Pulmonari Oxdative Stress

11
 Pulmonary and Systemic Oxidant/Antioxidant Imbalance in Chronic Obstructive Pulmonary Disease Willia m MacNee ELEGI, Colt Research Laboratories, MRC Centre for Inflammation Research, Medical School, University of Edinburgh, Edinburgh, Scotland, United Kingdom An imbalance between oxidants and antioxidants is considered to play a role in the pathogenesis of chronic obstructive pulmonary disease (COPD). There is considerable evidence that an increased oxidative burden occurs in the lungs of patients with this disorder, and this may be involved in many of the pathogenic processes, suchas dire ct injur y to lungcells, mucus hype rsecr etion , inac tivat ion of antip roteases, and enha ncing lung infla mmat ion throu gh activ a- tion of redox-sensitive transcription factors. COPD is now recog- nized to have multiple systemic consequences, such as weight loss and skeletal muscle dysfunction. Moreover, it is appreciated that oxidative stress extends beyond the lung and may, through similar oxidative stress mechanisms as those in the lung, contribute to several of the systemic manifestations in COPD. Keywords: chronic obstructive pulmonary disease; oxidant/antioxidant imbalance; oxidative stress; smoking; systemic inflammation The lungs are exposed continuou sly to oxidants generated either endogenously from phagocytes and other cell types or exoge- nously from air pollutants or cigarette smoke. In addition, intra- cellular oxidants, such as those derived from mitochondrial elec- tron transport, are involved in many cellular signaling pathways. Lung cells are protected against this oxidative challenge by well- developed enzymatic and nonenzymatic antioxidan t systems (1). When the balance between oxidants and antioxidants shifts in favor of the former, from either an excess of oxidants and/or depletion of antioxidants, oxidative stress occurs. Oxidative stress produces not only direct injurious effects in the lungs but also acti vate s mol ecu lar mechanisms that initiate lun g ina mmat ion (2). Smoking is the main etiologic factor in chronic obstructive pulmonary disease (COPD). Cigarette smoke contains around 10 17 oxidant molecules per puff, and this, together with a large body of evidence demonstrating increased oxidative stress in smokers with and without COPD, has led to the proposed role of oxidant/antioxid ant imbalance in the pathogenesis of this con- dition (3). Increasingly, COPD is recognized to affect not only the lungs but also to have signicant systemic consequences, such as muscle dysfunction and weight loss (4). Oxidative stress is alsobelieved to play an impo rtant role in the syste mic manif es- tations of COPD (5). HOW IS OXIDATIVE STRESS MEASURED? Oxi dat ivestresscan be mea sur ed in sev era l dif fer ent way s, eit her by dir ectmeasurements of theoxida tiv e burde n, as therespo nse s to oxidative stress, or by the effects of oxidative stress on target molecules (Table 1). Direct measurements of the oxidative bur- den in airspaces can be derived from measurement of hydrogen (Rec eiv ed in ori gin al for m Nov emb er 11,2004 ; acc ept ed in fi nalformDecember21,2004 ) Corr esp onde nce and requ estsfor reprints sho uld be addr esse d to W. MacN ee,M.B. Ch.B., M.D., ELEGI, Colt Research Laboratories, Wilkie Building, Medical School, Teviot Place, Edinburgh EH8 9AG, Scotland, UK. E-mail: [email protected] Proc Am Thora c Soc Vol 2. p p 50–60 , 2005 DOI: 10.1513/pats.200411-056SF Internet address: www.atsjournals.org peroxide (H 2 O 2 ) in bronchoalveolar lavage (BAL) uid or in exhaled breath condensate. Airspace leukocytes derived from BAL can also be assessed  ex vivo  for their ability to produce reactive oxygen species (ROS). Nitric oxide (NO) is produced in the lungs by the catalytic activity of NO synthase (NOS), which exists in both constitutive isoforms (cNOS) and an induc- ible isoform (iNOS) (6). The latter is induced by inammatory stimuli, and NO in exhaled breath is considered a marker of inammation and indirectly as a marker of oxidative stress. Measurements of the responses to oxidative stress can be obtained by assessing changes in antioxidants in BAL uid or in indu ced sputu m. The depl etion of antio xidants or upre gula tion of ant iox idantenzyme s canbe assessedin lun g tis sue(7). Per haps more important than the presence of oxidative stress are mea- surements of the effects of oxidative stress on target molecules. Oxidative stress renders proteins more susceptible to proteolytic degradation by modifying amino acid chains to form protein aggregates and cleaving peptide bonds (8). As part of this pro- cess, some amino acid residues are converted to carbonyl resi- dues. Exposure of human plasma to cigarette smoke  in vitro resul ts in depletio n of plas ma prote in sulfh ydry l grou ps and elevation of protein carbonyl levels (9). Plasma proteins can also be degr adedthrough nitra tion and oxid ationby reactive nitro gen species, the formation of which is stimulated by cigarette smok- ing (10). Oxidized proteins can be measured as protein carbonyls, ni- trotyrosine, or oxidative damage to DNA producing 8-hydroxy- deoxyguanine. The reaction of NO and superoxide anion pro- duces pe rox yni tri te (Fi gur e 1),whichcan the n cause thenitra tio n of tyrosine to produce nitrotyrosine, a marker of peroxynitrite that can be measured in blood, breath condensate, BAL, and lung tissue as an indicator of free-radical protein damage (11). NO may form stabl e nitro sothi ols with low- mole cula r-wei ght thiols, such as glutathione, to enhance its bioactivity (12). Nitrite is a further end-product of NO. Lipid peroxidation is a process of abstraction of a proton from a side chain of a fatty acid to produce a carbon-centered rad ica l, which its elfcan the n react wit h the fat ty aci d to pro duc ea lipid pero xidation prod uct and a furth er carbo n-cen tered radical, which can react again, resulting in a chain reaction (13). Lipid peroxidation products, such as F2-isoprostanes, 4-hydroxyno- nenal (4-HNE), and hydrocarbons, can be measured in breath, breath condensate, BAL, and lung tissue. EVIDENCE OF LOCAL OXIDATIVE STRESS IN THE LUNGS Numerous studies have shown that oxidative stress is increased in the lun gs of patien ts wit h COP D compar ed wit h hea lth y subjects, but also compared with smokers with similar smoking history but who have not developed airways obstruction (3). Smokers and patients with COPD have higher levels of H 2 O 2  in exhaled breath condensate, a direct measurement of airspace oxidative burden, than ex-smokers with COPD or nonsmokers (14, 15). H 2 O 2  levels are even higher during exacerbations of COPD (Figure 2) (14). The elevated level of H 2 O 2 in the exhaled Proc Am Thorac Soc 2005.2:50-60. Downloaded from www.atsjournals .org by 223.255.227.30 on 01/29/15. For personal use only.

description

j

Transcript of Pulmonari Oxdative Stress

  • Pulmonary and Systemic Oxidant/AntioxidantImbalance in Chronic Obstructive Pulmonary DiseaseWilliam MacNee

    ELEGI, Colt Research Laboratories, MRC Centre for Inflammation Research, Medical School, University of Edinburgh, Edinburgh, Scotland,United Kingdom

    An imbalance between oxidants and antioxidants is considered toplay a role in the pathogenesis of chronic obstructive pulmonarydisease (COPD). There is considerable evidence that an increasedoxidative burden occurs in the lungs of patients with this disorder,and this may be involved in many of the pathogenic processes,such asdirect injury to lung cells,mucushypersecretion, inactivationof antiproteases, and enhancing lung inflammation through activa-tion of redox-sensitive transcription factors. COPD is now recog-nized to have multiple systemic consequences, such as weight lossand skeletal muscle dysfunction. Moreover, it is appreciated thatoxidative stress extends beyond the lung and may, through similaroxidative stress mechanisms as those in the lung, contribute toseveral of the systemic manifestations in COPD.

    Keywords: chronic obstructive pulmonary disease; oxidant/antioxidantimbalance; oxidative stress; smoking; systemic inflammation

    The lungs are exposed continuously to oxidants generated eitherendogenously from phagocytes and other cell types or exoge-nously from air pollutants or cigarette smoke. In addition, intra-cellular oxidants, such as those derived from mitochondrial elec-tron transport, are involved in many cellular signaling pathways.Lung cells are protected against this oxidative challenge by well-developed enzymatic and nonenzymatic antioxidant systems (1).When the balance between oxidants and antioxidants shifts infavor of the former, from either an excess of oxidants and/ordepletion of antioxidants, oxidative stress occurs. Oxidative stressproduces not only direct injurious effects in the lungs but alsoactivates molecular mechanisms that initiate lung inflammation (2).

    Smoking is the main etiologic factor in chronic obstructivepulmonary disease (COPD). Cigarette smoke contains around1017 oxidant molecules per puff, and this, together with a largebody of evidence demonstrating increased oxidative stress insmokers with and without COPD, has led to the proposed roleof oxidant/antioxidant imbalance in the pathogenesis of this con-dition (3). Increasingly, COPD is recognized to affect not onlythe lungs but also to have significant systemic consequences,such as muscle dysfunction and weight loss (4). Oxidative stressis also believed to play an important role in the systemic manifes-tations of COPD (5).

    HOW IS OXIDATIVE STRESS MEASURED?

    Oxidative stress can bemeasured in several different ways, eitherby direct measurements of the oxidative burden, as the responsesto oxidative stress, or by the effects of oxidative stress on targetmolecules (Table 1). Direct measurements of the oxidative bur-den in airspaces can be derived from measurement of hydrogen

    (Received in original formNovember 11, 2004; accepted in final formDecember 21,2004)

    Correspondence and requests for reprints should be addressed toW.MacNee, M.B.Ch.B., M.D., ELEGI, Colt Research Laboratories, Wilkie Building, Medical School,Teviot Place, Edinburgh EH8 9AG, Scotland, UK. E-mail: [email protected]

    Proc Am Thorac Soc Vol 2. pp 5060, 2005DOI: 10.1513/pats.200411-056SFInternet address: www.atsjournals.org

    peroxide (H2O2) in bronchoalveolar lavage (BAL) fluid or inexhaled breath condensate. Airspace leukocytes derived fromBAL can also be assessed ex vivo for their ability to producereactive oxygen species (ROS). Nitric oxide (NO) is producedin the lungs by the catalytic activity of NO synthase (NOS),which exists in both constitutive isoforms (cNOS) and an induc-ible isoform (iNOS) (6). The latter is induced by inflammatorystimuli, and NO in exhaled breath is considered a marker ofinflammation and indirectly as a marker of oxidative stress.

    Measurements of the responses to oxidative stress can beobtained by assessing changes in antioxidants in BAL fluid orin induced sputum.The depletion of antioxidants or upregulationof antioxidant enzymes can be assessed in lung tissue (7). Perhapsmore important than the presence of oxidative stress are mea-surements of the effects of oxidative stress on target molecules.Oxidative stress renders proteins more susceptible to proteolyticdegradation by modifying amino acid chains to form proteinaggregates and cleaving peptide bonds (8). As part of this pro-cess, some amino acid residues are converted to carbonyl resi-dues. Exposure of human plasma to cigarette smoke in vitroresults in depletion of plasma protein sulfhydryl groups andelevation of protein carbonyl levels (9). Plasma proteins can alsobe degraded through nitration and oxidation by reactive nitrogenspecies, the formation of which is stimulated by cigarette smok-ing (10).

    Oxidized proteins can be measured as protein carbonyls, ni-trotyrosine, or oxidative damage to DNA producing 8-hydroxy-deoxyguanine. The reaction of NO and superoxide anion pro-duces peroxynitrite (Figure 1), which can then cause the nitrationof tyrosine to produce nitrotyrosine, a marker of peroxynitritethat can be measured in blood, breath condensate, BAL, andlung tissue as an indicator of free-radical protein damage (11).NO may form stable nitrosothiols with low-molecular-weightthiols, such as glutathione, to enhance its bioactivity (12). Nitriteis a further end-product of NO.

    Lipid peroxidation is a process of abstraction of a protonfrom a side chain of a fatty acid to produce a carbon-centeredradical, which itself can then react with the fatty acid to produce alipid peroxidation product and a further carbon-centered radical,which can react again, resulting in a chain reaction (13). Lipidperoxidation products, such as F2-isoprostanes, 4-hydroxyno-nenal (4-HNE), and hydrocarbons, can be measured in breath,breath condensate, BAL, and lung tissue.

    EVIDENCE OF LOCAL OXIDATIVE STRESSIN THE LUNGS

    Numerous studies have shown that oxidative stress is increasedin the lungs of patients with COPD compared with healthysubjects, but also compared with smokers with similar smokinghistory but who have not developed airways obstruction (3).Smokers and patients with COPD have higher levels of H2O2 inexhaled breath condensate, a direct measurement of airspaceoxidative burden, than ex-smokers with COPD or nonsmokers(14, 15). H2O2 levels are even higher during exacerbations ofCOPD (Figure 2) (14). The elevated level of H2O2 in the exhaled

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • MacNee: Systemic Oxidative Stress in COPD 51

    TABLE 1. MEASUREMENTS OF OXIDATIVE STRESS

    Direct measurements of oxidative burden Hydrogen peroxide in breath condensate or BAL fluidBAL/peripheral blood leukocyte reactive oxygen species productionNitric oxide in exhaled breath

    Responses to oxidative stressCarbon monoxide in breath (reflecting hemoxygenase activity)Antioxidants, antioxidant enzymes in blood, sputum, BAL, and lung tissue

    Effects of oxidative stress on target moleculesOxidized proteins (e.g., carbonyl residues, oxidase, and nitrated proteins)Lipid peroxidation products (e.g., F2-isoprostains, 4-hydroxy-2-nonenal,hydrocarbons) in breath condensate, sputum,BAL, blood, urine, lung tissue

    Definition of abbreviation: BAL bronchoalveolar lavage.

    breathof smokers is believed toderive partly from increased releaseof superoxide anion (O2) by alveolar macrophages (16, 17).

    Because the iron content of smokers alveolar macrophagesis also increased compared with that of nonsmokers (18), theresultant increased free iron in the airspaces of smokers wouldstimulate the generation of ROS through the Fenton reaction(10). A further source of both O2 and H2O2 is the xanthine/xanthine oxidase reaction. Xanthine oxidase activity is increasedin cell-free BAL fluid and in the plasma of smokers and inpatients with COPD compared with healthy subjects and non-smoking subjects, respectively (19, 20).

    NO has been used as a marker of airway inflammation andindirectly as a measure of oxidative stress. Increased NO levelsin exhaled breath occurs in some studies of COPD but are not ashigh as the NO levels reported in asthma (2123). Other studieshave found either normal or even lower exhaled NO in patientswith stable COPD, compared with normal subjects (24, 25).

    The rapid reaction of NO with O2, as described previously,or with thiols may alter breath NO levels. Nitrosothiol levelshave been shown to be higher in breath condensate in smokersand patients with COPD compared with subjects who do notsmoke (Figure 3) (6). Smoking increases directly exhaled NOlevels, which limits the usefulness of this marker in COPD.Peroxynitrite, formed by the reaction of NO with superoxideanion, can cause nitrosylation of tyrosine to produce nitrotyro-sine (11). Nitrotyrosine levels are elevated in sputum leukocytesof patients with COPD (Figure 3) and are correlated negativelywith the FEV1 (26).

    Exhaled carbon monoxide (CO) as a measure of the responseof heme oxygenase to oxidative stress (Figure 4) has been shownto be elevated in exhaled breath in patients with COPD com-

    Figure 1. Nitric oxide (NO) and NO-related products. FAD flavinadenine dinucleotide; FMN flavin mononucleotide; NADP nicotin-amide adenine dinucleotide phosphate; NADPH reduced form ofNADP; S-GSNO S-nitrosoglutathione. Adapted from Reference 6.

    Figure 2. Individual data for hydrogen peroxide (H2O2) concentrationsin breath condensate in control subjects and in patients with stable andunstable chronic obstructive pulmonary disease (COPD). Modified fromReference 14 by permission.

    pared with normal subjects (Figure 5) (27). Again, CO is presentin cigarette smoke, which limits the usefulness of this markerof oxidative stress in patients who smoke.

    Lipid peroxidation products, such as thiobarbituric acid-reacting substances, are elevated in sputum in patients withCOPD,and correlate negatively with the FEV1 (28, 29). Isoprostanes,produced by ROS-mediated peroxidation of arachidonic acid,circulate in plasma and can be excreted in the urine (30). Thelevels of 8-isoprostane in breath condensate have been shownto be elevated in patients with COPD compared with normalsubjects and smokers who have not developed the disease, andcorrelate with the degree of obstruction (31). The levels of8-isoprostane in breath condensate in patients with COPD alsoshow a positive correlation with the percentage of neutrophilsin induced sputum, suggesting a role for oxidative stress in theairway inflammation (Figure 6) (32). Isoprostanes may also re-flect systemic effects caused by ROS. In plasma, the levels offree F2-isoprostanes are increased in smokers and decreasedafter smoking cessation (33). In urine, the levels of F2-isopros-tanes are elevated in patients with COPD in comparison tohealthy control subjects, with the highest levels observed duringexacerbations (34).

    Lipid peroxides can interact with enzymatic or nonenzymaticantioxidants or decompose by reacting with metal ions or iron-

    Figure 3. Left: nitrosothiols, breath condensate in smokers and patientswith COPD. Right: increased inducible NO (iNOS) and nitrotyrosineimmunoreactivity in sputum leukocytes in COPD. *p 0.05. Modifiedby permission from Reference 26.

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • 52 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005

    Figure 4. Hemeoxygenase system-synthesis of carbon monoxide (CO).

    containing proteins, thereby forming hydrocarbon gases and un-saturated aldehydes (13). Hydrocarbons are by-products of fattyacid peroxidation (27). Patients with COPD show an increasedlevel of exhaled ethane in breath compared with control subjects,the levels being correlated negatively with lung function (28, 35).

    There is increasing evidence that these markers of oxidativestress are also reflected in lung tissue in patients with COPD.Increased nitrotyrosine immunoreactivity has been shown insputum leukocytes and lung tissue in patients with COPD com-pared with healthy subjects (26). The lipid peroxidation product4-HNE reacts quickly with cellular proteins to form adducts.These adducts have been shown to be present in greater quanti-ties in airway epithelial and endothelial cells in the lungs ofpatients with COPD, compared with smokers with a similarsmoking history who have not developed the disease (Figure 7)(36). Lipid peroxidation products, such as 8-isoprostane, canact as signaling molecules and cause release of inflammatorymediators, such as interleukin 8 (IL-8) from lung cells (37).4-HNE can cause the upregulation of transforming growth factor (TGF-) (38) and oxidant enzyme gene expression (39).

    Thus, many studies show increased markers of airway oxida-tive stress in exhaled breath or breath condensate and in lungtissue in patients with COPD compared with normal subjectsand smokers who have not developed the disease. In addition,many of thesemarkers correlatewith the degree of airflow limita-tion in COPD, suggesting a role for oxidative stress in the declinein lung function in COPD. Furthermore, products of oxidativestress, such as lipid peroxides, can act as signaling molecules toenhance inflammation in COPD.

    ANTIOXIDANTS AND COPD

    Several studies have investigated a relationship between anti-oxidants, pulmonary function, and the development of COPD(40, 41). The National Health and Nutrition Examination Survey(NHANES) and the Dutch Monitoring Project for the RiskFactors for Chronic Diseases (MORGEN) suggested relation-ships between dietary intake and airflow limitation. In NHANESI, lower dietary vitamin C related directly to lower FEV1 levelsin a population survey study and, in addition, the protectiveeffect of vitamin C was even greater in subjects with bronchitis(40). Data from NHANES II showed an inverse relationshipbetween both dietary and serum vitamin C and chronic respira-tory symptoms (41). Moreover, in NHANES III, the levels ofdietary vitamin C, vitamin E, selenium, and beta-carotene werepositively associated with lung function (42). Analysis of thedata obtained in the MORGEN study also shows higher intakeof vitamin C and beta-carotene is associated with a higher levelFEV1, comparedwith a low intake of these antioxidants (43). Theassociation between dietary vitaminE intake and lung function isless consistent (4345). Several studies, including theMORGEN

    Figure 5. Exhaled CO in patients with COPD with and without cortico-steroid therapy. ns not significant. Modified by permission fromReference 27.

    study, have shown associations between fruit intake and higherFEV1 and lower symptoms in patients with COPD (4649). Datafrom the MORGEN study have also shown the beneficial effectsof flavonoids on FEV1 (50).

    IS OXIDATIVE STRESS IMPORTANT IN THEPATHOGENESIS OF COPD?

    Biomarkers of oxidative stress have been related to the develop-ment of airflow limitation, and many studies have shown higheroxidant levels in patients with COPD, compared with healthysmokers. Furthermore, several studies show relationships be-tween oxidative stress markers and the degree of airflow limita-tion in COPD (3, 5, 51). However, the presence of oxidativestress and its relationship to airflow limitation may be epipheno-mena, because it occurs in inflammation, a characteristic featureof COPD (10). There are as yet no longitudinal studies showingthat the presence of enhanced oxidative stress relates to thedecline in FEV1 or to the progression of the disease.

    OXIDATIVE STRESS ANDPROTEASE/ANTIPROTEASE IMBALANCE

    An increased protease burden in the lungs occurs as a result ofthe influx and activation of inflammatory leukocytes. It has beenproposed that a relative deficiency of antiproteases, such as1-antitrypsin, because of their inactivation by oxidants, createsa protease/antiprotease imbalance in the lungs. This forms the

    Figure 6. Isoprostane levels in exhaled breath condensate (EBC) insmokers with COPD (left). Significant correlation between 8-isoprostanelevels in EBC and percentage of neutrophils in induced sputum (right).Adapted from Reference 32.

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • MacNee: Systemic Oxidative Stress in COPD 53

    Figure 7. Immunostaining for lipidperoxidation product 4-hydroxy-2-nonenal (4-HNE) adduct in thelungs of smokers with and withoutCOPD. (A ) Immunohistochemistryshowing increased staining in bothbronchial and alveolar epitheliumin patients with COPD who havethe same smoking history as agroup of smokers without COPD.(B ) Staining score showing in-creased staining in COPD. (C ) Cor-relation between 4-HNE stainingscore in the lungs and percent pre-dicted FEV1. Modified by permis-sion from Reference 36.

    basis of the protease/antiprotease theory of the pathogenesis ofemphysema (52, 53). Inactivation of 1-antitrypsin occurs byoxidation of a critical methionine residue at its active site byoxidants from cigarette smoke or released from inflammatoryleukocytes, resulting in a dramatic reduction in its inhibitorycapacity in vitro (54, 55). The acute effects of cigarette smokeon functional activity of 1-antitrypsin have been studied in vivo,and show a transient, but nonsignificant, fall in the antiproteaseactivity of BAL fluid 1 hour after smoking (56). However, aprotease/antiprotease imbalance involving 1-antitrypsin andneutrophil elastase is likely an oversimplification, because otherproteases and other antiproteases are likely to have a role inthe pathogenesis of COPD.

    OXIDANTS AND MUCUS HYPERSECRETION

    Oxidant-generating systems, such as xanthine/xanthine oxidase,can cause airway epithelial mucus secretion (57). Oxidants arealso involved in the signaling pathways for epidermal growthfactor, which has an important role in mucus production (58).H2O2 or HOCl, in relatively low concentrations (100 M), causessignificant impairment of ciliary beating and even complete stasis(59). Oxidant-mediated hypersecretion and impaired mucocili-ary clearance may result in airway mucus accumulation contrib-uting to airflow limitation (60).

    OXIDATIVE STRESS AND AIRSPACE EPITHELIAL INJURY

    Because of their direct contact with the environment, the lungepithelial surfaces are especially vulnerable to the effects ofoxidative stress. Injury to the airway epithelium is an importantearly event after exposure to cigarette smoke as shown by anincrease in airspace epithelial permeability. This increased per-meability can be shown to result from cigarette smoke exposure

    both in vitro and in vivo (6163) and is partially reversible byantioxidants. Extra- and intracellular glutathione appears to becritical to the maintenance of epithelial integrity after exposureto smoke. This has been shown in studies of the permeabilityof epithelial cell monolayers in vitro and in animal models in vivoafter exposure to cigarette smoke condensate, which is associatedwith profound changes in the homeostasis of the antioxidant gluta-thione (6366). Depletion of lung glutathione alone can induceincreased airspace epithelial permeability (63, 66). Interindividualvariability in antioxidant defenses may be one factor in determin-ing whether COPD develops after cigarette smoking.

    OXIDATIVE STRESS AND NEUTROPHIL INFLUXIN THE LUNGS

    The lungs contain a large pool of noncirculating (marginated)neutrophils, which either are retained ormove only slowly withinthe pulmonary microcirculation (67). This is because of thesmaller average diameter of the neutrophil ( 7 m) comparedwith the pulmonary capillary diameter ( 5 m), and results ina proportion of the circulating neutrophils having to deform andthus move slowly to negotiate the small capillaries. In normalsubjects, there is a correlation between neutrophil deformability,measured in vitro, and their subsequent sequestration in thepulmonary microcirculation after reinjectionthe less deform-able the cells, the greater their sequestration (68). This mecha-nism of neutrophil sequestration in the pulmonary bed allowstime to interact and adhere to capillary endothelium before theirsubsequent migration across the alveolarcapillary membraneto the interstitial airspaces. During smoking, there is a transientincrease in neutrophil sequestration in the lungs (69) becauseof a decrease in circulating neutrophil deformability (67, 70).Studies in vitro show that the decreased neutrophil deformabilityinduced by cigarette smoke is abolished by antioxidants (71).

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • 54 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005

    Figure 8. Oxidative stress in exacerbations of COPD. Super-oxide anion release from peripheral blood neutrophils (PMN;spontaneous andphorbolmyristate acetate [PMA] activated)in normal subjects and in patients with acute and stableCOPD (left). Plasma antioxidant capacity (Trolox equivalentantioxidant capacity [TEAC]) in normal subjects, and in thosewith stable and exacerbated COPD (right). ROS reactiveoxygen species. *p 0.05; **p 0.01. Adapted from Refer-ence 108.

    Oxidative stress occurs systemically during smoking as shownby a profound decrease in the antioxidant capacity of plasma(Figure 8), which may reduce neutrophil deformability becauseof actin polymerization (71). These may be the initial eventsthat cause the influx of neutrophils into the lungs of cigarettesmokers, and variability in this effect (69, 70) may be one reasonwhy not all smokers develop enhanced lung inflammation andtherefore COPD. It is possible that activation of neutrophilssequestered in the pulmonary microvasculature could also in-duce the release of ROS and proteases within a microenviron-ment, which limits access for free-radical scavengers and anti-proteases. Thus, destruction of the alveolar wall, as occurs inemphysema, could result from a proteolytic/oxidant insult de-rived from the intravascular space without the need for theneutrophils to migrate into the airspaces.

    OXIDATIVE STRESS AND LUNG INFLAMMATION

    As mentioned previously, oxidative stress is present whereverthere is inflammation. Regardless, oxidative stress may also bea mechanism for enhancing the airspace inflammation that is acharacteristic feature of COPD (10). Oxidative stress can causethe release of chemotactic factors, such as IL-8, from airwayepithelial cells (72), and epithelial cells from patients with COPDrelease more IL-8 than those of smokers or healthy individuals(73). Numerousmarkers of inflammation, such as IL-8 and tumornecrosis factor-, have been shown to be elevated in the sputumof patients with COPD (74), and there is overwhelming evidencethat COPD is associated with enhanced airway inflammation asconfirmed by biopsy studies (75). Recent studies have also showna relationship between markers of oxidative stress in breath andthe number of neutrophils in induced sputum (32). Epidemio-logic studies have shown a relationship between circulating neu-trophil numbers and FEV1 (76) and between the changes inperipheral blood neutrophil numbers and airflow limitation overtime (77). There is also a relationship between peripheral bloodneutrophil oxidant release and measures of airflow limitation inyoung smokers (78). In addition, lipid peroxidation products inplasma have also been shown to correlate inversely with thepercent predicted FEV1 in a population study (79).

    Oxidative stress may have a fundamental role enhancing in-flammation through the upregulation of redox-sensitive tran-scription factors, such as nuclear factor B (NF-B) and activat-ing protein 1 (AP-1), and also the extracellular signal-regulatedkinase (ERK), c-Jun N-terminal kinase (JNK), and p38mitogen-activated protein kinase pathways. Cigarette smoke has beenshown to activate all of these signaling mechanisms (2, 8082).Genes for many inflammatory mediators are regulated by NF-B,

    which is present in the cytosol in an inactive form linked to itsinhibitory protein IB. Many stimuli, including cytokines andoxidants, result in activation of IBkinase, resulting in phosphor-ylation of IB, cleaving of IB from NF-B, and the destructionof IB in the proteasome. This critical event in the inflammatoryresponse is redox-sensitive (83, 84). Studies in macrophage celllines and alveolar and bronchial epithelial cells show that oxi-dants cause the release of inflammatory mediators, such as IL-8,IL-1, andNO, and that these events are associatedwith increasedexpression of the genes for these inflammatory mediators andincreased nuclear binding or activation of NF-B (85, 86). Thelinking of NF-B to its consensus site in the nucleus leads toenhanced transcription of proinflammatory genes and thereforeinflammation, which itself will produce more oxidative stress,creating a vicious circle of enhanced inflammation resulting fromthe increased oxidative stress (Figure 9). Animal models ofsmoke exposure show that neutrophil influx in the lungs is associ-ated with increased IL-8 gene expression and protein releaseandwithNF-Bactivation (87).All of these events are associatedwith oxidative stress because they can be abrogated by antioxi-dant therapy (Figure 10). NF-B can be activated and translo-cated to the nucleus in lung tissue in smokers and in patients withCOPD compared with healthy subjects (88). NF-B activation inlung tissue has been shown to correlate with the FEV1 (89).

    A further event induced by oxidative stress, which may en-hance lung inflammation, is chromatin remodeling. Under nor-mal circumstances, DNA is wound tightly around a core of his-tones, and in this configuration prevents excessive transcriptionby factors such as NF-B, as well as reduced access of RNApolymerase to DNA, thereby resulting in transcriptional repres-sion and gene silencing (Figure 11). Under the influence of his-tone acetyltransferases, histone residues are acetylated, causinga change in their charge and the unwinding of DNA. This thenallows access for transcription factors such as NF-B and/or RNApolymerase to the transcriptional machinery, therefore enhanc-ing gene expression. This process is reversed by histone deacety-lases (HDAC), enzymes that deacetylate histone residues, re-sulting in the rewinding of DNA and gene silencing. Theseprocesses are known to be redox-sensitive. Histone acetylationcan be shown to occur after cigarette smoke exposure of epithe-lial cells and is prevented by the antioxidant N-acetylcysteine,indicating that the process is redox-sensitive (90). Furthermore,animal models of cigarette smoke exposure have been shownto result in increased acetylated histone in lung and decreasedHDAC activity, and both of these events would enhance geneexpression (91). HDAC activity in alveolar macrophages, specifi-cally HDAC2 activity, has also been shown to be downregulated

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • MacNee: Systemic Oxidative Stress in COPD 55

    Figure 9. Activation of signal pathways by oxidative stress. AP-1 acti-vating protein 1; ERK extracellular signal-related kinase; ikK inhibitorkB kinase; JNK c-Jun N-terminal kinase; NF-B nuclear factor B;P phosphate.

    in alveolar macrophages obtained from cigarette smokers, a pro-cess that would enhance gene expression (Figure 12) (92). Recentstudies suggest that acetylatedhistone residues, specifically histone4 (H4), are present to a greater extent in smokers and in patientswith COPD who smoke, with an associated decrease in HDAC2,specifically in patients with COPD who smoke and in patientswith severe COPD (88). Acetylated histone 3 appears to beelevated in lung tissue in ex-smoking patients with COPD andmay be a mechanism for persistent inflammation in COPD aftersmoking cessation (88). A correlation has also been shown be-tween decreased HDAC activity in lung tissue and FEV1 inpatients with COPD (88, 93).

    Many of the markers of oxidative stress do not respond totherapy with corticosteroids, and it is believed that oxidativestress may be involved in the relative resistance to these drugsin COPD. HDAC recruitment is believed to be required forthe antiinflammatory action of corticosteroids in smokers and inpatients with COPD. Amechanism involving nitration of HDACs,and therefore downregulation of HDACs in lung tissue, mayprevent the action of corticosteroids (94).

    Recent evidence suggests that latent adenoviral infectionmaybe associated with the pathogenesis of COPD by enhancing lunginflammation (95). E1A protein derived from latent adenoviral

    Figure 10. Effect of recombinant superoxide dismutase (rhSOD) on cig-arette smoke (CS-)induced neutrophil influx, interleukin (IL-)-8 geneexpression, and NF-B nuclear binding in guinea pig lungs. *p 0.05;**p 0.01. Modified by permission from Reference 86.

    Figure 11. Model of histone acetylation/deacetylation. HAT histoneacetyltransferase.

    infection interacts with transcription factor cofactors and en-hances nuclear binding of transcription factors (95). E1A hasbeen noted to be more prevalent in the lungs of smokers whodevelop COPD than in smokers who do not develop the disease(96). Cells transfected with the E1A protein have also beenshown to have increased IL-8 release in response to oxidants,such as H2O2, and enhanced NF-B activation (97). Latent ade-noviral infection may, therefore, render lung cells more suscepti-ble to oxidative stress, thus enhancing the release of proinflam-matory mediators. The presence of E1A also enhances smoke-induced inflammation and emphysema in animal models (98).

    The pathogenesis of emphysema has been proposed recentlyto result from loss of alveolar endothelial cells via apoptosis,and this may be as an initial event in the development of emphy-sema (99). Apoptosis occurs to a greater extent in emphysema-tous lungs than in lungs of nonsmokers (100). The process ofendothelial apoptosis is believed to be under the influence ofvascular endothelial growth factor (VEGF)R2 receptors. Down-regulation of VEGF-R2 has been shown in animals to produceemphysema (100), and reduced expression of VEGF-R2 is evi-dent in emphysematous human lungs (100). Studies have alsoshown that the apoptosis/emphysema produced by VEGF in-hibition in animal models is associated with increased markersof oxidative stress and prevented by antioxidants (101).

    Ablation in mice of nuclear factor, erythroid-derived 2, like2 (Nrf2), a redox-sensitive transcription factor that is involvedin the regulation of many antioxidant genes results in moreextensive cigarette smokeinduced emphysema than occurs inwild-type mice (102), and this was associated with increasedmarkers of oxidative stress and increased apoptosis in the lungs,suggesting a role for theNrf2 pathway in determining the suscep-tibility to tobacco smokeinduced emphysema through a mecha-nism involving upregulation of antioxidant defenses.

    Other events involving oxidative stress and epithelial cellsmay be relevant to the development of COPD. For example,TGF-1 expression is increased in small airway epithelial cellsof smokers compared with nonsmokers and increased evenmorein patients with COPD (101, 102). TGF- mRNA levels corre-lated positively with smoking history and the degree of airwayobstruction (103, 104).

    Furthermore, TGF-1 itself may increase oxidative stress be-cause this substance produces profound changes in endothelialand epithelial cell glutathione (105, 106) by a mechanism involv-ing the downregulation of -glutamylcysteine synthetase (GCS)RNA in alveolar epithelial cells (39, 106). In addition, oxidativestress has been shown to activate TGF-.

    Studies in vitro show that the lipid peroxidation product

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • 56 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005

    Figure 12. Histone deacetylase(HDAC) activity (left) and im-munoreactivity (right) in alveolarmacrophages from smokers andnonsmokers. Modified by permis-sion from Reference 90.

    4-HNE is present in increased amounts in lung tissue in patientswith COPD (36). 4-HNE increases TGF- expression by amech-anism dependent on the activation of macrophage AP-1 (38).Furthermore, 4-HNE induces GCS in alveolar epithelial cells(39), and increased GCS expression has been demonstrated inthe lungs of patients with COPD (107). Thus, it has been sug-gested that 4-HNE can act as a second messenger in the regula-tion of protective antioxidant genes, such as GCS, and a varietyof other genes, such as TGF- (107).

    Thus, as a response to oxidative stress induced by cigarettesmoke, there appears to be upregulation of protective antioxi-dant genes. Glutathione is concentrated in epithelial lining fluidcompared with plasma (108). Human studies have shown thatglutathione is elevated in the epithelial lining fluid in chroniccigarette smokers compared with nonsmokers, an increase thatdoes not occur during acute cigarette smoking (61). The effectsof chronic cigarette smoking can be mimicked by exposure ofairspace epithelial cells to cigarette smoke condensate in vitro.This produces an initial decrease in intracellular glutathione(GSH) with a rebound increase after 24 hours (64, 109). Thiseffect is mimicked by a similar change in glutathione in rat lungsin vivo after exposure to cigarette smoke (109), associated withan increase in the oxidized form of glutathione (GSSG). Theincrease in glutathione after cigarette smoke exposure is causedby transcriptional upregulation of GCSmRNA, the rate-limitingenzyme in GSH synthesis (109). Themechanism of the upregula-tion of GCS mRNA is by the activation by cigarette smoke ofthe redox-sensitive transcription factor AP-1 (108, 110). Theseevents likely account for the increased glutathione levels seenin epithelial lining fluid in chronic cigarette smokers, a protectivemechanism. The injurious effects of cigarette smoke may occurrepeatedly during and immediately after cigarette smoking whenthe lung is depleted of antioxidants, including glutathione.TNF-, which is believed to have a role in the lung inflammationof COPD, also decreases intracellular glutathione levels initiallyin epithelial cells by a mechanism involving intracellular oxida-tive stress, which is followed 24 hours thereafter by a reboundincrease in intracellular glutathione as a result of AP-1 activationand increased GCS expression (108). Animals exposed to wholecigarette smoke for up to 14 days also show increased expressionof a number of antioxidant genes, including manganese super-oxidedismutase,metallothionine, andglutathioneperoxidase(111).

    Sensitivity to oxidative stress may also be a cofactor in thedevelopment of a protease/antiprotease imbalance and conse-quent emphysema. Strains of mice that decreased their lung anti-oxidant defenses developed emphysema when exposed chroni-cally to cigarette smoke, whereas emphysema did not developin strains of mice that showed upregulation of antioxidants inresponse to smoke (112).

    SYSTEMIC OXIDATIVE STRESS

    An increased systemic oxidative burden has been shown to occurin smokers. In COPD, peripheral blood neutrophils have beenshown to release more ROS than in normal subjects and this isenhanced still further in exacerbation, being associated withmarked depletion of the plasma antioxidant capacity, indicatingincreased systemic oxidative stress (Figure 8) (113). Products oflipid peroxidation are also increased in plasma in smokers withCOPD, particularly during exacerbations (113). Increased levelsof nitrotyrosine have also been shown to occur in the plasma ofpatients with COPD (25).

    Exacerbations of COPD result from increased levels of airpollutants, specifically particulate air pollution (see article in thisissue by van Eeden and coworkers, pp. 6167) (114). Particulateair pollution causes oxidative stress in the airways (115), andenhanced inflammation by mechanisms similar to those de-scribed previously, through oxidative stressinducedNF-B acti-vation, decreased histone deacetylation, and increased histoneacetylation (72, 86). Air pollution has also been associated withincreased cardiovascular deaths. The mechanism of this effectmay involve oxidative stressinduced changes in fibrinolytic bal-ance, resulting in enhanced plasminogen activator inhibitor type1 and decreased tissue-type plasminogen activator, both of whichwould reduce fibrinolysis of clots forming on ruptured athero-sclerotic plaques and therefore lead to acute cardiac events, suchas myocardial infarction and death (discussed in detail in thearticles in this issue by MacCallum, pp. 3443, and Tapson, pp.7177) (116). Furthermore, COPD is an independent risk factorfor the development of ischemic heart disease and death frommyocardial infarction, as reviewed by Sin and Man in this issue,pp. 811 (117).

    It is now recognized that COPD is not only a disease thataffects the lungs but has important systemic consequences, such

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • MacNee: Systemic Oxidative Stress in COPD 57

    Figure 13. Altered glutamatemetabolism associatedwith reducedmus-cle glutathione levels in patients with emphysema. **p 0.01; ***p 0.001. Adapted from Reference 125.

    as cachexia of skeletalmuscle function (118). Increasing evidencesuggests that similar mechanisms involving oxidative stress andinflammation in the lungs may also be responsible for many ofthe systemic effects of COPD (5, 118).

    Patients with COPD often display weight loss, which is seenas an independent predictor of outcome (119, 120). Loss of fat-free mass also results in peripheral muscle dysfunction, de-creased exercise capacity, and reduced health status (121123).There are several factors that influence the loss of weight andfat-free mass in patients with COPD, including malnutrition,imbalance in overall protein turnover and hormones involvedin this process, tissue hypoxia, and pulmonary inflammation (118,122, 124, 125). The roles of body weight and fat-free mass inthe pathogenesis andmorbidity of COPD are reviewed byWout-ers in this issue (pp. 2633).

    The cachexia and loss of fat-free mass that occur in COPDmay involve oxidative stress. Skeletal muscle is exposed continu-ously to changes in the redox environment as occurs duringexercise. There is increased ROS production by the mitochon-drial respiratory chain after exercise in patients with COPD. Ithas been shown that lipid peroxidation products increase inserum during exercise accompanied by an increase in GSSG/GSH ratio (125128). The increased oxidative stress was muchgreater than in healthy individuals. Thus, although redox homeo-stasis seems to be conserved in COPD, it requires only a littlestress to disturb this balance. Skeletal muscle cells adapt tooxidative stress by upregulating antioxidant enzymes, such asmanganese, copper, and zinc superoxide dismutase (SOD), cata-lase, and glutathione peroxidase (129). In patients with COPDsubmitted to a training protocol, GSSG/GSH ratios increased,whereas no such increase was seen in healthy individuals (130).This finding suggests that the adaptor response of skeletal muscleto oxidative stress may be impaired in COPD. There is alsoevidence of disturbed redox homeostasis in the phenotype ofCOPD associated with emphysema, in which glutathione levelsin the vastus lateralis muscle were decreased. This was associatedwith reduced concentrations of glutamate, an important sub-strate in the synthesis of glutamine and glutathione (Figure 13)(131). This suggests that glutathione metabolism may be im-paired in COPD, which is supported further by studies thatdemonstrate a decrease in glutathione peroxidase activity, ele-vated glutathione reductase activity, and increased lipid peroxi-dation indicative of oxidative damage in skeletalmuscle of exper-imental emphysema in hamsters (132).

    The mitochondrial electron transport chain may be responsi-ble for the increased ROS production in skeletal muscle duringexercise and the mitochondrial electron transport chain may bestimulated by TNF (133), which is elevated in the circulation in

    patients with COPD who lose weight (134). Leukocytes thatinfiltrate muscles in patients with COPD may be another sourceof ROS (135). Moreover, exercise also increases xanthine/xan-thine oxidase activity, another source of ROS (136). Reactivenitrogen species may also contribute to oxidative stress. Induc-ible NO expression in skeletal muscle in response to inflamma-tory cytokines has been reported and depends on NF-B activa-tion (135). Muscle function may be directly compromised byoxidative stress, which has been shown in vitro to decrease con-tractility and ex vivo to increase fragility of muscle to oxidants(136, 137). Proteins in the contractile apparatus of musclemay beoxidized by ROS, critically sulfhydryl residues in the contractileproteins, which may impair force development (138). In additionto impairing muscle contractility and muscle fatigue, oxidativestress may also compromise muscle function directly and inducemuscle atrophy. Muscle mass can be reduced as a result of animbalance in muscle protein metabolism, which has been de-scribed in studies of oxidative stressinduced inhibition of mus-cle-specific protein expression (139, 140). In addition, oxidativestress may result in apoptosis, which has also been described forskeletal monocytes and may contribute to oxidative stressdependent muscle atrophy (141, 142).

    In conclusion, there is now considerable evidence of bothlocal and systemic oxidative stress in patients with COPD. Thereis also increasing evidence that oxidative stress is involved inthe pathogenesis of local lung inflammation as well as in systemicphenomena, such as skeletal muscle dysfunction, and perhapseven the increased cardiovascular risk of mortality that resultsfrom COPD.

    Conflict of Interest Statement : W.M. has been reimbursed for travel by Glaxo-SmithKline, Zambon, AstraZeneca, Boehringer Ingelheim, Pfizer, and Micromet forattending conferences and has received honoraria from GlaxoSmtihKline, Astra-Zeneca, Zambon, and Pfizer for participating as a speaker in scientific meetings,and serves on advisory boards for GlaxoSmithKline, Pfizer, Almirall, Amgen, Bayer,and Micromet, and serves as a consultant for Pfizer and SMB Pharmaceuticals, andresearch grants to support work carried out in his laboratory come from SMB, Pfizer,Ceremedix, GlaxoSmithKline, Chugi, and Novartis.

    References

    1. Halliwell B. Antioxidants in human health and disease. Annu Rev Nutr1996;16:3350.

    2. Rahman I, MacNee W. Role of transcription factors in inflammatorylung diseases. Thorax 1998;53:601612.

    3. MacNee W. Oxidants/antioxidants and COPD. Chest 2000;117:303S317S.

    4. Agusti AG. Systemic effects of chronic obstructive pulmonary disease.Novartis Found Symp 2001;234:242249.

    5. Langen RC, Korn SH, Wouters EF. ROS in the local and systemicpathogenesis of COPD. Free Radic Biol Med 2003;35:226235.

    6. Corradi M, Montuschi P, Donnelly LE, Pesci A, Kharitonov SA, BarnesPJ. Increased nitrosothiols in exhaled breath condensate in inflam-matory airway diseases.Am JRespir Crit CareMed 2001;163:854858.

    7. MacNee W. Antioxidants. Hansel TT, Barnes PJ, editors. New drugsfor asthma, allergy and COPD. Basel, Switzerland: Karger; 2001. pp.151155.

    8. Stadtman ER. Metal ion-catalyzed oxidation of proteins: biochemicalmechanism and biological consequences. Free Radic Biol Med 1990;9:315325.

    9. Nagler R, Lischinsky S, Diamond E, Drigues N, Klein I, Reznick AZ.Effect of cigarette smoke on salivary proteins and enzyme activities.Arch Biochem Biophys 2000;379:229236.

    10. MacNee W. Oxidative stress and lung inflammation in airways disease.Eur J Pharmacol 2001;429:195207.

    11. Petruzzelli S, Puntoni R, Mimotti P, Pulera N, Baliva F, Fornai E,Giuntini C. Plasma 3-nitro tyrosine in cigarette smokers. Am J RespirCrit Care Med 1997;156:19021907.

    12. Gaston B, Reilly J, Drazen JM, Fackler J, Ramdev P, Arnelle D,MullinsME, Sugarbaker DJ, Chee C, Singel DJ, et al. Endogenous nitrogenoxides and bronchodilator S-nitrosothiols in human airways. ProcNatl Acad Sci USA 1993;90:1095710961.

    13. Repine JE, Bast A, Lankhorst I. Oxidative stress in chronic obstructive

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • 58 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005

    pulmonary disease. Oxidative Stress Study Group. Am J Respir CritCare Med 1997;156:341357.

    14. Dekhuijzen PN, Aben KK, Dekker I, Aarts LP, Wielders PL, vanHerwaarden CL, Bast A. Increased exhalation of hydrogen peroxidein patients with stable and unstable chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 1996;154:813816.

    15. Nowak D, Kasielski M, Pietras T, Bialasiewicz P, Antczak A. Cigarettesmoking does not increase hydrogen peroxide levels in expired breathcondensate of patients with stable COPD. Monaldi Arch Chest Dis1998;53:268273.

    16. Schaberg T, Haller H, Rau M, Kaiser D, Fassbender M, Lode H. Super-oxide anion release induced by platelet-activating factor is increasedin human alveolar macrophages from smokers. Eur Respir J 1992;5:387393.

    17. Hoidal JR, Fox RB, LeMarbe PA, Perri R, Repine JE. Altered oxidativemetabolic responses in vitro of alveolar macrophages from asympto-matic cigarette smokers. Am Rev Respir Dis 1981;123:8589.

    18. Thompson AB, Bohling T, Heires A, Linder J, Rennard SI. Lowerrespiratory tract iron burden is increased in association with cigarettesmoking. J Lab Clin Med 1991;117:493499.

    19. Heunks LM, Vina J, van Herwaarden CL, Folgering HT, Gimeno A,Dekhuijzen PN. Xanthine oxidase is involved in exercise-inducedoxidative stress in chronic obstructive pulmonary disease.Am J Phys-iol 1999;277:R1697R1704.

    20. Pinamonti S, Muzzoli M, Chicca MC, Papi A, Ravenna F, Fabbri LM,Ciaccia A. Xanthine oxidase activity in bronchoalveolar lavage fluidfrom patients with chronic obstructive pulmonary disease. Free RadicBiol Med 1996;21:147155.

    21. Delen FM, Sippel JM, Osborne ML, Law S, Thukkani N, Holden WE.Increased exhaled NO in chronic bronchitis: comparison with asthmaand COPD. Chest 2000;117:695701.

    22. Maziak W, Loukides S, Culpitt S, Sullivan P, Kharitonov SA, BarnesPJ. Exhaled NO in chronic obstructive pulmonary disease. Am JRespir Crit Care Med 1998;157:9981002.

    23. Corradi M, Majori M, Cacciani GC, Consigli GF, deMunari E, PesciA. Increased exhaled NO in patients with stable chronic obstructivepulmonary disease. Thorax 1999;54:572575.

    24. Rutgers SR, van der Mark TW, Coers W, Moshage H, Timens W,Kauffman HF, Koeter GH, Postma DS. Markers of NO metabolismin sputum and exhaled air are not increased in chronic obstructivepulmonary disease. Thorax 1999;54:576580.

    25. Clini E, Bianchi L, PaganiM,Ambrosino N. EndogenousNO in patientswith stable COPD: correlates with severity of disease. Thorax 1998;53:881883.

    26. Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato K. Increase inreactive nitrogen species production in chronic obstructive pulmonarydisease airways. Am J Respir Crit Care Med 2000;162:701706.

    27. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, Barnes PJ.Exhaled ethane, a marker of lipid peroxidation, is elevated in chronicobstructive pulmonary disease. Am J Respir Crit Care Med 2000;162:369373.

    28. Nowak D, Kasielski M, Antczak A, Pietras T, Bialasiewicz P. Increasedcontent of thiobarbituric acid-reactive substances and hydrogen per-oxide in the expired breath condensate of patients with stable chronicobstructive pulmonary disease: no significant effect of cigarette smok-ing. Respir Med 1999;93:389396.

    29. Tsukagoshi H, Shimizu Y, Iwamae S, Hisada T, Ishizuka T, Iizuka K,Dobashi K, Mori M. Evidence of oxidative stress in asthma andCOPD: potential inhibitory effect of theophylline. Respir Med 2000;94:584588.

    30. Morrow JD, Roberts LJ. The isoprostanes: unique bioactive productsof lipid peroxidation. Prog Lipid Res 1997;36:121.

    31. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, Corradi M, KhariSA, Barnes PJ. Exhaled 8-isoprostane as an in vivo biomarker oflung oxidantive stress in patients with COPD and healthy smokers.Am J Respir Crit Care Med 2000;162:11751177.

    32. Frangulyan R, Anderson D, Drost E, Hill A, MacNee W. Exhaledmarkers of inflammation in breath condensate in patients with bron-chiectasis and COPD. Thorax 2003;58:S116.

    33. Morrow JD, Frei B, Longmire AW, Gaziano JM, Lynch SM, Shyr Y,Strauss WE, Oates JA, Roberts LJ II. Increase in circulating productsof lipid peroxidation (F2-isoprostanes) in smokers: smoking as a causeof oxidative damage. N Engl J Med 1995;332:11981203.

    34. PraticoD,Basili S, VieriM,Cordova C,Violi F, FitzGeraldGA.Chronicobstructive pulmonary disease is associated with an increase in urinarylevels of isoprostane F2alpha-III, an index of oxidant stress. Am JRespir Crit Care Med 1998;158:17091714.

    35. Habib MP, Clements NC, Garewal HS. Cigarette smoking and ethaneexhalation in humans.AmJRespir Crit CareMed 1995;151:13681372.

    36. Rahman I, van Schadewijk AA, Crowther AJ, Hiemstra PS, Stolk J,MacNeeW,De BoerWI. 4-Hydroxy-2-nonenal, a specific lipid perox-idation product, is elevated in lungs of patients with chronic obstruc-tive pulmonary disease.Am J Respir Crit Care Med 2002;166:490495.

    37. Scholz H, Yndestad A, Damas JK, Waehre T, Tonstad S, Aukrust P,Halvorsen B. 8-isoprostane increases expression of interleukin-8 inhuman macrophages through activation of mitogen-activated proteinkinases. Cardiovasc Res 2003;59:945954.

    38. Leonarduzzi G, Scavazza A, Biasi F, Chiarpotto E, Camandola S, VogelS, Dargel R, Poli G. The lipid peroxidation end product 4-hydroxy-2,3-nonenal up-regulates transforming growth factor beta1 expressionin the macrophage lineage: a link between oxidative injury and fibro-sclerosis. FASEB J 1997;11:851857.

    39. Arsalane K, Dubois CM, Muanza T, Begin R, Boudreau F, Asselin C,Cantin AM. Transforming growth factor-beta1 is a potent inhibitorof glutathione synthesis in the lung epithelial cell line A549: transcrip-tional effect on the GSH rate-limiting enzyme gamma-glutamylcys-teine synthetase. Am J Respir Cell Mol Biol 1997;17:599607.

    40. Schwartz J, Weiss ST. Relationship between dietary vitamin C intakeand pulmonary function in the First National Health and NutritionExamination Survey (NHANES I). Am J Clin Nutr 1994;59:110114.

    41. Schwartz J, Weiss ST. Dietary factors and their relation to respiratorysymptoms: the Second National Health and Nutrition ExaminationSurvey. Am J Epidemiol 1990;132:6776.

    42. Hu G, Cassano PA. Antioxidant nutrients and pulmonary function: theThird National Health and Nutrition Examination Survey (NHANESIII). Am J Epidemiol 2000;151:975981.

    43. Grievink L, Smit HA, Ocke MC. vant Veer P, Kromhout D. Dietaryintake of antioxidant (pro)-vitamins, respiratory symptoms and pul-monary function: the MORGEN study. Thorax 1998;53:166171.

    44. Dow L, Tracey M, Villar A, Coggon D, Margetts BM, Campbell MJ,Holgate ST. Does dietary intake of vitamins C and E influence lungfunction in older people? Am J Respir Crit Care Med 1996;154:14011404.

    45. Britton JR, Pavord ID, Richards KA, Knox AJ, Wisniewski AF, LewisSA, Tattersfield AE, Weiss ST. Dietary antioxidant vitamin intakeand lung function in the general population. Am J Respir Crit CareMed 1995;151:13831387.

    46. Tabak C, Smit HA, Heederik D, Ocke MC, Kromhout D. Diet andchronic obstructive pulmonary disease: independent beneficial effectsof fruits, whole grains, and alcohol (the MORGEN study). Clin ExpAllergy 2001;31:747755.

    47. Miedema I, Feskens EJ, Heederik D, Kromhout D. Dietary determi-nants of long-term incidence of chronic nonspecific lung diseases: theZutphen Study. Am J Epidemiol 1993;138:3745.

    48. Strachan DP, Cox BD, Erzinclioglu SW, Walters DE, Whichelow MJ.Ventilatory function and winter fresh fruit consumption in a randomsample of British adults. Thorax 1991;46:624629.

    49. Tabak C, Feskens EJ, Heederik D, Kromhout D, Menotti A, BlackburnHW. Fruit and fish consumption: a possible explanation for popula-tion differences in COPDmortality (the Seven Countries Study). EurJ Clin Nutr 1998;52:819825.

    50. TabakC,Arts IC, Smit HA, HeederikD, KromhoutD. Chronic obstruc-tive pulmonary disease and intake of catechins, flavonols, and fla-vones: the MORGEN study. Am J Respir Crit Care Med 2001;164:6164.

    51. BootsAW,HaenenGR,BastA.Oxidantmetabolism in chronic obstruc-tive pulmonary disease. Eur Respir J Suppl 2003;46:14s27s.

    52. Janoff A, Carp H, Laurent P, Raju L. The role of oxidative processesin emphysema. Am Rev Respir Dis 1983;127:S31S38.

    53. Stockley RA. Proteases and antiproteases. Novartis Found Symp 2001;234:189199.

    54. Bieth JG. The antielastase screen of the lower respiratory tract. Eur JRespir Dis Suppl 1985;139:5761.

    55. Evans MD, Pryor WA. Damage to human alpha-1-proteinase inhibitorby aqueous cigarette tar extracts and the formation of methioninesulfoxide. Chem Res Toxicol 1992;5:654660.

    56. Abboud RT, Fera T, Richter A, Tabona MZ, Johal S. Acute effect ofsmoking on the functional activity of alpha1-protease inhibitor inbronchoalveolar lavage fluid. Am Rev Respir Dis 1985;131:7985.

    57. Adler KB, Holden-Stauffer WJ, Repine JE. Oxygen metabolites stimu-late release of high-molecular-weight glycoconjugates by cell andorgan cultures of rodent respiratory epithelium via an arachidonicacid-dependent mechanism. J Clin Invest 1990;85:7585.

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • MacNee: Systemic Oxidative Stress in COPD 59

    58. Nadel JA. Role of epidermal growth factor receptor activation in regu-lating mucin synthesis. Respir Res 2001;2:8589.

    59. Feldman C, Anderson R, Kanthakumar K, Vargas A, Cole PJ, WilsonR. Oxidant-mediated ciliary dysfunction in human respiratory epithe-lium. Free Radic Biol Med 1994;17:110.

    60. Rogers DF. Mucus hypersecretion in chronic obstructive pulmonarydisease. In: Chadwick D, Goode JA, editors. Chronic obstructivepulmonary disease: pathogenesis to treatment. Chichester, UK: JohnWiley & Sons; 2001. pp. 6583.

    61. Morrison D, Rahman I, Lannan S, MacNee W. Epithelial permeability,inflammation, and oxidant stress in the air spaces of smokers. Am JRespir Crit Care Med 1999;159:473479.

    62. Rusznak C, Mills PR, Devalia JL, Sapsford RJ, Davies RJ, LozewiczS. Effect of cigarette smoke on the permeability and IL-1beta andsICAM-1 release from cultured human bronchial epithelial cells ofnever-smokers, smokers, and patients with chronic obstructive pulmo-nary disease. Am J Respir Cell Mol Biol 2000;23:530536.

    63. Li XY, Donaldson K, Rahman I, MacNee W. An investigation of therole of glutathione in increased epithelial permeability induced bycigarette smoke in vivo and in vitro. Am J Respir Crit Care Med 1994;149:15181525.

    64. Rahman I, Li XY, Donaldson K, Harrison DJ, MacNee W. Glutathionehomeostasis in alveolar epithelial cells in vitro and lung in vivo underoxidative stress. Am J Physiol 1995;269:L285L292.

    65. Rahman I, Bel A, Mulier B, Lawson MF, Harrison DJ, MacNee W,Smith CA. Transcriptional regulation of gamma-glutamylcysteinesynthetase-heavy subunit by oxidants in human alveolar epithelialcells. Biochem Biophys Res Commun 1996;229:832837.

    66. Li XY, Rahman I, Donaldson K, MacNee W. Mechanisms of cigarettesmoke induced increased airspace permeability. Thorax 1996;51:465471.

    67. MacNee W, Selby C. New perspectives on basic mechanisms in lungdisease. 2. Neutrophil traffic in the lungs: role of haemodynamics,cell adhesion, and deformability. Thorax 1993;48:7988.

    68. Selby C, Drost E, Wraith PK, MacNee W. In vivo neutrophil sequestra-tion within lungs of humans is determined by in vitro filterability.J Appl Physiol 1991;71:19962003.

    69. MacNee W, Wiggs B, Belzberg AS, Hogg JC. The effect of cigarettesmoking on neutrophil kinetics in human lungs. N Engl J Med 1989;321:924928.

    70. Drost EM, Selby C, Bridgeman MM, MacNee W. Decreased leukocytedeformability after acute cigarette smoking in humans.AmRevRespirDis 1993;148:12771283.

    71. Drost EM, Selby C, Lannan S, Lowe GD, MacNee W. Changes inneutrophil deformability following in vitro smoke exposure: mecha-nism and protection. Am J Respir Cell Mol Biol 1992;6:287295.

    72. Gilmour PS, Rahman I, Donaldson K, MacNee W. Histone acetylationregulates epithelial IL-8 release mediated by oxidative stress fromenvironmental particles. Am J Physiol Lung Cell Mol Physiol 2003;284:L533L540.

    73. ProfitaM, Chiappara G,Mirabella F, DiGiorgi R, Chimenti L, CostanzoG, Riccobono L, Bellia V, Bousquet J, Vignola AM. Effect of cilomi-last (Ariflo) on TNF-alpha, IL-8, and GM-CSF release by airwaycells of patients with COPD. Thorax 2003;58:573579.

    74. Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences in interleu-kin-8 and tumor necrosis factor-alpha in induced sputum frompatientswith chronic obstructive pulmonary disease or asthma. Am J RespirCrit Care Med 1996;153:530534.

    75. Jeffery PK. Structural and inflammatory changes in COPD: a compari-son with asthma. Thorax 1998;53:129136.

    76. Chan-Yeung M, Dybuncio A. Leucocyte count, smoking and lung func-tion. Am J Med 1984;76:3137.

    77. Chan-Yeung M, Abboud R, Buncio AD, Vedal S. Peripheral leucocytecount and longitudinal decline in lung function. Thorax 1988;43:462466.

    78. RichardsGA,TheronAJ,Van derMerweCA,AndersonR. Spirometricabnormalities in young smokers correlate with increased chemilumi-nescence responses of activated blood phagocytes. Am Rev RespirDis 1989;139:181187.

    79. Schunemann HJ, Muti P, Freudenheim JL, Armstrong D, Browne R,Klocke RA, Trevisan M. Oxidative stress and lung function. Am JEpidemiol 1997;146:939948.

    80. Di Stefano A, Caramori G, Oates T, Capelli A, Lusuardi M, GnemmiI, Ioli F, Chung KF, Donner CF, Barnes PJ, et al. Increased expressionof nuclear factor-kappaB in bronchial biopsies from smokers andpatients with COPD. Eur Respir J 2002;20:556563.

    81. Rahman I. Oxidative stress, transcription factors and chromatin remod-elling in lung inflammation. Biochem Pharmacol 2002;64:935942.

    82. MacNee W, Rahman I. Is oxidative stress central to the pathogenesisof chronic obstructive pulmonary disease? Trends Mol Med 2001;7:5562.

    83. Janssen-Heininger YM, Macara I, Mossman BT. Cooperativity betweenoxidants and tumor necrosis factor in the activation of nuclear factor(NF)-B: requirement of Ras/mitogen-activated protein kinases inthe activation of NF-B by oxidants. Am J Respir Cell Mol Biol 1999;20:942952.

    84. Flohe L, Brigelius-Flohe R, Saliou C, Traber MG, Packer L. Redoxregulation of NF-kappa B activation. Free Radic Biol Med 1997;22:11151126.

    85. Parmentier M, Hirani N, Rahman I, Donaldson K, MacNee W, Antoni-celli F. Regulation of lipopolysaccharide-mediated interleukin-1betarelease by N-acetylcysteine in THP-1 cells. Eur Respir J 2000;16:933939.

    86. Jimenez LA, Thompson J, Brown DA, Rahman I, Antonicelli F, DuffinR, Drost EM, Hay RT, Donaldson K, MacNee W. Activation of NF-kappaB by PM(10) occurs via an iron-mediated mechanism in theabsence of IkappaB degradation. Toxicol Appl Pharmacol 2000;166:101110.

    87. NishikawaM, Kakemizu N, Ito T, KudoM, Kaneko T, SuzukiM, UdakaN, Ikeda H, Okubo T. Superoxide mediates cigarette smoke-inducedinfiltration of neutrophils into the airways through nuclear factor-kappaB activation and IL-8 mRNA expression in guinea pigs in vivo.Am J Respir Cell Mol Biol 1999;20:189198.

    88. Szulakowski P, Drost E, Jimenez LA, Wickenden JA, Crowther AJ,Donaldson K. MacNee. Role of histone acetylation in transcriptionalregulation and promotion of inflammatory process in patients withchronic obstructive pulmonary disease [abstract]. Am J Respir CritCare Med 2004;169:A275.

    89. Crowther AJ, Rahman I, Antonicelli F, Jimenez LA, Salter D, MacNeeW. Oxidative stress and transcription factors AP-1 and NF-B inhuman lung tissue [abstract]. Am J Respir Crit Care Med 1999;159:A816.

    90. Anderson C, Kilty I, Marwick JA, MacNee W, Rahman I. Cigarettesmoke and H2O2-mediated decrease in histone deacetylase activityis attenuated by N-acetyl-L-cysteine but not be I-B kinase inhibitionin A549 cells [abstract]. Am J Respir Crit Care Med 2004;169:A424.

    91. Marwick JA, Kirkham P, Gilmour PS, Donaldson K, MacNee W, Rah-man I. Cigarette smoke-induced oxidative stress and TGF-1 increasep21waf1/cip1 expression in alveolar epithelial cells. Ann N Y AcadSci 2002;973:278283.

    92. Ito K, Lim S, Caramori G, Chung KF, Barnes PJ, Adcock IM. Cigarettesmoking reduces histone deacetylase 2 expression, enhances cytokineexpression, and inhibits glucocorticoid actions in alveolar macro-phages. FASEB J 2001;15:11101112.

    93. To Y, Elliott WM, Ito M, Hayashi S, Adcock IM, Hogg JC, Barnes PJ,Ito K. Total histone deacetylase activity decreases with an increasingclinical stage of COPD [abstract].Am J Respir Crit Care Med 2004;169:A276.

    94. Barnes PJ, Ito K, Adcock IM. Corticosteroid resistance in chronic ob-structive pulmonary disease: inactivation of histone deacetylase.Lan-cet 2004;28,363:731733.

    95. Vitalis TZ, Kern I, Croome A, Behzad H, Hayashi S, Hogg JC. Theeffect of latent adenovirus 5 infection on cigarette smoke-inducedlung inflammation. Eur Respir J 1998;11:664669.

    96. Matsuse T, Hayashi S, Kuwano K, Keunecke H, Jefferies WA, HoggJC. Latent adenoviral infection in the pathogenesis of chronic airwaysobstruction. Am Rev Respir Dis 1992;146:177184.

    97. Gilmour PS, Rahman I, Hayashi S, Hogg JC, Donaldson K, MacNeeW. Adenoviral E1A primes alveolar epithelial cells to PM10-inducedtranscription of interleukin-8. Am J Physiol Lung Cell Mol Physiol2001;281:L598L606.

    98. Meshi B, Vitalis TZ, Ionescu D, Elliott WM, Liu C, Wang XD, HayashiS, Hogg JC. Emphysematous lung destruction by cigarette smoke:the effects of latent adenoviral infection on the lung inflammatoryresponse. Am J Respir Cell Mol Biol 2002;26:5257.

    99. Voelkel NF, Tuder R. COPD: exacerbation. Chest 2000;117(5 Suppl 2):376S379S.

    100. Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF.Endothelial cell death and decreased expression of vascular endothe-lial growth factor and vascular endothelial growth factor receptor 2in emphysema. Am J Respir Crit Care Med 2001;163:737744.

    101. Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y,Salvemini D, Voelkel NF, Flores SC. Oxidative stress and apoptosis

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.

  • 60 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005

    interact and cause emphysema due to vascular endothelial growthfactor receptor blockade. Am J Respir Cell Mol Biol 2003;29:8897.

    102. Rangasamy T, ChoCY, Thimmulappa RK, Zhen L, Srisuma SS, KenslerTW, YamamotoM, Petrache I, Tuder RM, Biswal S. Genetic ablationof Nrf2 enhances susceptibility to cigarette smoke-induced emphy-sema in mice. J Clin Invest 2004;114:12481259.

    103. Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, OkadaY, Yamasawa F, Nakahara K, Umeda A. Increased expression oftransforming growth factor-beta1 in small airway epithelium fromtobacco smokers and patients with chronic obstructive pulmonarydisease (COPD). Am J Respir Crit Care Med 2001;163:14761483.

    104. de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J, HiemstraPS, van Krieken JH. Transforming growth factor beta1 and recruit-ment of macrophages and mast cells in airways in chronic obstructivepulmonary disease. Am J Respir Crit Care Med 1998;158:19511957.

    105. White AC, Das SK, Fanburg BL. Reduction of glutathione is associatedwith growth restriction and enlargement of bovine pulmonary arteryendothelial cells produced by transforming growth factor-beta 1. AmJ Respir Cell Mol Biol 1992;6:364368.

    106. Jardine H, MacNee W, Donaldson K, Rahman I. Molecular mechanismof transforming growth factor (TNF)-beta1-induced glutathionedepletion in alveolar epithelial cells: involvement of AP-1/ARE andFra-1. J Biol Chem 2002;277:2115821166.

    107. Rahman I, MacNee W. Regulation of redox glutathione levels andgene transcription in lung inflammation: therapeutic approaches. FreeRadic Biol Med 2000;28:14051420.

    108. Rahman I, Antonicelli F, MacNee W. Molecular mechanism of theregulation of glutathione synthesis by tumor necrosis factor-alphaand dexamethasone in human alveolar epithelial cells. J Biol Chem1999;274:50885096.

    109. Rahman I, Smith CA, Lawson MF, Harrison DJ, MacNee W. Inductionof gamma-glutamylcysteine synthetase by cigarette smoke is associ-ated with AP-1 in human alveolar epithelial cells. FEBS Lett 1996;396:2125.

    110. Rahman I, Smith CA, Antonicelli F, MacNee W. Characterisation ofgamma-glutamylcysteine synthetase-heavy subunit promoter: a criti-cal role for AP-1. FEBS Lett 1998;427:129133.

    111. Gilks CB, Price K, Wright JL, Churg A. Antioxidant gene expressionin rat lung after exposure to cigarette smoke. Am J Pathol 1998;152:269278.

    112. Cavarra E, Bartalesi B, Lucattelli M, Fineschi S, Lunghi B, GambelliF, Ortiz LA, Martonana PA, Lungarella G. Effects of cigarette smokein mice with different levels of alpha(1)-proteinase inhibitor andsensitivity to oxidants. Am J Respir Crit Care Med 2001;164:886890.

    113. Rahman I, Morrison D, Donaldson K, MacNee W. Systemic oxidativestress in asthma, COPD, and smokers. Am J Respir Crit Care Med1996;154:10551060.

    114. MacNeeW, Donaldson K. Environmental factors in COPD. In: VoelkelNF, MacNee W, editors. Chronic obstructive lung disease. London:B.C. Decker; 2002. pp. 145160.

    115. Donaldson K, Brown DM,Mitchell C, Dineva M, Beswick PH, GilmourP, MacNee W. Free radical activity of PM10: iron-mediated genera-tion of hydroxyl radicals. Environ Health Perspect 1997;105:12851289.

    116. Donaldson K, Stone V, Seaton A, MacNee W. Ambient particle inhala-tion and the cardiovascular system: potential mechanisms. EnvironHealth Perspect 2001;109:523527.

    117. Sin DD, Man SF. Why are patients with chronic obstructive pulmonarydisease at increased risk of cardiovascular diseases? The potential roleof systemic inflammation in chronic obstructive pulmonary disease.Circulation 2003;107:15141519.

    118. Wouters EF, Creutzberg EC, Schols AM. Systemic effects in COPD.Chest 2002;121(5 Suppl):127S130S.

    119. Gray-Donald K, Gibbons L, Shapiro SH, Macklem PT, Martin JG.Nutritional status and mortality in chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 1996;153:961966.

    120. Landbo C, Prescott E, Lange P, Vestbo J, Almdal TP. Prognostic valueof nutritional status in chronic obstructive pulmonary disease. Am JRespir Crit Care Med 1999;160:18561861.

    121. Baarends EM, Schols AM, Mostert R, Wouters EF. Peak exercise re-

    sponse in relation to tissue depletion in patients with chronic obstruc-tive pulmonary disease. Eur Respir J 1997;10:28072813.

    122. Engelen MP, Schols AM, Does JD, Wouters EF. Skeletal muscle weak-ness is associated with wasting of extremity fat-free mass but not withairflow obstruction in patients with chronic obstructive pulmonarydisease. Am J Clin Nutr 2000;71:733738.

    123. Palange P, Forte S, Felli A, Galassetti P, Serra P, Carlone S. Nutritionalstate and exercise tolerance in patients with COPD. Chest 1995;107:12061212.

    124. Eid AA, Ionescu AA, Nixon LS, Lewis-Jenkins V, Matthews SB, Grif-fiths TL, Shale DJ. Inflammatory response and body composition inchronic obstructive pulmonary disease. Am J Respir Crit Care Med2001;164:14141418.

    125. Jenkins RC, Ross RJ. Growth hormone therapy for protein catabolism.QJM 1996;89:813819.

    126. Heunks LM, Dekhuijzen PN. Respiratory muscle function and freeradicals: from cell to COPD. Thorax 2000;55:704716.

    127. Sastre J, Asensi M, Gasco E, Pallardo FV, Ferrero JA, Furukawa T,Vina J. Exhaustive physical exercise causes oxidation of glutathionestatus in blood: prevention by antioxidant administration.Am J Phys-iol 1992;263:R992R995.

    128. Vina J, Servera E, Asensi M, Sastre J, Pallardo FV, Ferrero JA, Garcia-De-La-Asuncion J, Anton V, Marin J. Exercise causes blood glutathi-one oxidation in chronic obstructive pulmonary disease: preventionby O2 therapy. J Appl Physiol 1996;81:21982202.

    129. Franco AA, Odom RS, Rando TA. Regulation of antioxidant enzymegene expression in response to oxidative stress and during differentia-tion of mouse skeletal muscle. Free Radic Biol Med 1999;27:11221132.

    130. Rabinovich RA, Ardite E, Troosters T, Carbo N, Alonso J, Gonzalezde Suso JM,Vilaro J, Barbera JA, PoloMF,Argiles JM, et al.Reducedmuscle redox capacity after endurance training in patients withchronic obstructive pulmonary disease. Am J Respir Crit Care Med2001;164:11141118.

    131. Engelen MP, Schols AM, Does JD, Deutz NE, Wouters EF. Alteredglutamate metabolism is associated with reduced muscle glutathionelevels in patients with emphysema. Am J Respir Crit Care Med 2000;161:98103.

    132. Mattson JP, Sun J, Murray DM, Poole DC. Lipid peroxidation in theskeletalmuscle of hamsters with emphysema.Pathophysiology 2002;8:215221.

    133. Li YP, Atkins CM, Sweatt JD, Reid MB. Mitochondria mediate tumornecrosis factor-alpha/NF-kappaB signaling in skeletal muscle myo-tubes. Antioxid Redox Signal 1999;1:97104.

    134. Di Francia M, Barbier D, Mege JL, Orehek J. Tumor necrosis factor-alpha levels and weight loss in chronic obstructive pulmonary disease.Am J Respir Crit Care Med 1994;150:14531455.

    135. Adams V, Nehrhoff B, Spate U, Linke A, Schulze PC, Baur A, GielenS, Hambrecht R, Schuler G. Induction of iNOS expression in skeletalmuscle by IL-1beta and NFkappaB activation: an in vitro and in vivostudy. Cardiovasc Res 2002;54:95104.

    136. Andrade FH, Reid MB, Allen DG, Westerblad H. Effect of hydrogenperoxide and dithiothreitol on contractile function of single skeletalmuscle fibres from the mouse. J Physiol 1998;509:565575.

    137. Barclay JK, Hansel M. Free radicals may contribute to oxidative skeletalmuscle fatigue. Can J Physiol Pharmacol 1991;69:279284.

    138. MacFarlane NG, Miller DJ. Depression of peak force without alteringcalcium sensitivity by the superoxide anion in chemically skinnedcardiac muscle of rat. Circ Res 1992;70:12171224.

    139. Buck M, Chojkier M. Muscle wasting and dedifferentiation induced byoxidative stress in a murine model of cachexia is prevented by inhibi-tors of NO synthesis and antioxidants. EMBO J 1996;15:17531765.

    140. Langen RC, Van Der Velden JL, Schols AM, Kelders MC, Wouters EF,Janssen-Heininger YM. Tumor necrosis factor-alpha inhibits myogenicdifferentiation through MyoD protein destabilization. FASEB J 2004;18:227237.

    141. Stangel M, Zettl UK, Mix E, Zielasek J, Toyka KV, Hartung HP, GoldR. H2O2 and NO-mediated oxidative stress induce apoptosis in ratskeletal muscle myoblasts. J Neuropathol Exp Neurol 1996;55:3643.

    142. Agusti A, Morla M, Sauleda J, Saus C, Busquets X. NF-B activationand iNOS upregulation in skeletal muscle of patients with COPDand low body weight. Thorax 2004;59:483487.

    Proc Am Thorac Soc 2005.2:50-60.

    Downloaded from www.atsjournals.org by 223.255.227.30 on 01/29/15. For personal use only.