upper limb muscle dysfunction executive summary

17
AMERICAN THORACIC SOCIETY DOCUMENTS An Ofcial American Thoracic Society/European Respiratory Society Statement: Update on Limb Muscle Dysfunction in Chronic Obstructive Pulmonary Disease Executive Summary François Maltais, Marc Decramer, Richard Casaburi, Esther Barreiro, Yan Burelle, Richard Debigar ´ e, P. N. Richard Dekhuijzen, Frits Franssen, Ghislaine Gayan-Ramirez, Joaquim Gea, Harry R. Gosker, Rik Gosselink, Maurice Hayot, Sabah N. A. Hussain, Wim Janssens, Michael I. Polkey, Josep Roca, Didier Saey, Annemie M. W. J. Schols, Martijn A. Spruit, Michael Steiner, Tanja Taivassalo, Thierry Troosters, Ioannis Vogiatzis, and Peter D. Wagner; on behalf of the ATS/ERS Ad Hoc Committee on Limb Muscle Dysfunction in COPD THIS OFFICIAL STATEMENT OF THE AMERICAN THORACIC SOCIETY (ATS) AND THE EUROPEAN RESPIRATORY SOCIETY (ERS) WAS APPROVED BY THE ATS BOARD OF DIRECTORS,NOVEMBER 2013, AND BY THE ERS EXECUTIVE COMMITTEE,SEPTEMBER 2013 Background: Limb muscle dysfunction is prevalent in chronic obstructive pulmonary disease (COPD) and has important clinical implications, such as reduced exercise tolerance, quality of life, and even survival. Since the last American Thoracic Society (ATS) and European Respiratory Society (ERS) statement on limb muscle dysfunction, important progress has been made in the characterization of this problem and on our understanding of its pathophysiology and clinical implications. Purpose: The purpose of this document is to update the 1999 ATS/ ERS statement on limb muscle dysfunction in COPD. Methods: An interdisciplinary committee of experts from the ATS and ERS determined that the scope of this document should be limited to limb muscles. Committee members conducted focused reviews of the literature on relevant topics. A librarian also performed a literature search. An ATS methodologist provided advice to the committee, ensuring that the methodological approach was consistent with ATS standards. Results: We identied important advances in our understanding of the extent and nature of the structural alterations in limb muscles in patients with COPD. Since the last update, studies have been published regarding mechanisms of development of limb muscle dysfunction in COPD and on the treatment of this condition. The clinical implications of limb muscle dysfunction have been delineated. Although exercise training is the most potent intervention to address this condition, other therapies, such as neuromuscular electrical stimulation, are emerging. Assessment of limb muscle function can identify patients who are at increased risk of poor clinical outcomes, such as exercise intolerance and premature mortality. Conclusions: Limb muscle dysfunction is a key systemic consequence of COPD. However, there are still important gaps in our knowledge about mechanisms of development of this problem. Strategies for early detection and specic treatments for this condition are needed. This Executive Summary is part of the full official Limb Muscle Dysfunction in COPD Update statement, which readers may access online at www.atsjournals.org/ doi/abs/10.1164/rccm.201402-0373ST. Only the Executive Summary is appearing in the print edition of the Journal. The article of record, and the one that should be cited, is: An Official American Thoracic Society/European Respiratory Society Statement: update on limb muscle dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2014;189:e15–e62. Available at www.atsjournals.org/doi/abs/10.1164/rccm.201402-0373ST. F.M. holds a CIHR/GSK Research Chair on COPD at Universit ´ e Laval. R.C. holds the Grancell/Burns Chair in the Rehabilitative Sciences. E.B.’s contribution to this statement was supported by CIBERES, FIS 11/02029, 2009-SGR-393, SEPAR 2010, FUCAP 2011, FUCAP 2012, and Marat ´ o TV3 (MTV3-07-1010) (Spain) and by the ERS COPD Research Award 2008. M.I.P.’s contribution to this statement was supported by the NIHR Respiratory Disease Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, who partially fund his salary. Am J Respir Crit Care Med Vol 189, Iss 9, pp 1121–1137, May 1, 2014 Copyright © 2014 by the American Thoracic Society DOI: 10.1164/rccm.201402-0373ST Internet address: www.atsjournals.org American Thoracic Society Documents 1121

Upload: rescateur

Post on 23-Dec-2015

8 views

Category:

Documents


1 download

DESCRIPTION

Ghid

TRANSCRIPT

Page 1: Upper Limb Muscle Dysfunction Executive Summary

AMERICAN THORACIC SOCIETYDOCUMENTS

An Official American Thoracic Society/European RespiratorySociety Statement: Update on Limb Muscle Dysfunctionin Chronic Obstructive Pulmonary DiseaseExecutive SummaryFrançois Maltais, Marc Decramer, Richard Casaburi, Esther Barreiro, Yan Burelle, Richard Debigare,P. N. Richard Dekhuijzen, Frits Franssen, Ghislaine Gayan-Ramirez, Joaquim Gea, Harry R. Gosker, Rik Gosselink,Maurice Hayot, Sabah N. A. Hussain, Wim Janssens, Michael I. Polkey, Josep Roca, Didier Saey,Annemie M. W. J. Schols, Martijn A. Spruit, Michael Steiner, Tanja Taivassalo, Thierry Troosters, Ioannis Vogiatzis,and Peter D. Wagner; on behalf of the ATS/ERS Ad Hoc Committee on Limb Muscle Dysfunction in COPD

THIS OFFICIAL STATEMENT OF THE AMERICAN THORACIC SOCIETY (ATS) AND THE EUROPEAN RESPIRATORY SOCIETY (ERS) WAS APPROVED BY THE ATS BOARD OF

DIRECTORS, NOVEMBER 2013, AND BY THE ERS EXECUTIVE COMMITTEE, SEPTEMBER 2013

Background: Limb muscle dysfunction is prevalent in chronicobstructive pulmonary disease (COPD) and has important clinicalimplications, such as reduced exercise tolerance, quality of life, andeven survival. Since the last American Thoracic Society (ATS)and European Respiratory Society (ERS) statement on limbmuscle dysfunction, important progress has been made in thecharacterization of this problem and on our understanding of itspathophysiology and clinical implications.

Purpose: The purpose of this document is to update the 1999 ATS/ERS statement on limb muscle dysfunction in COPD.

Methods: An interdisciplinary committee of experts from the ATSand ERS determined that the scope of this document should belimited to limb muscles. Committee members conducted focusedreviews of the literature on relevant topics. A librarian also performeda literature search. An ATS methodologist provided advice to thecommittee, ensuring that the methodological approach wasconsistent with ATS standards.

Results:We identified important advances in our understanding ofthe extent and nature of the structural alterations in limb musclesin patients with COPD. Since the last update, studies have beenpublished regarding mechanisms of development of limb muscledysfunction in COPD and on the treatment of this condition.The clinical implications of limb muscle dysfunction have beendelineated.Although exercise training is themost potent interventionto address this condition, other therapies, such as neuromuscularelectrical stimulation, are emerging. Assessment of limb musclefunction can identify patients who are at increased risk of poorclinical outcomes, such as exercise intolerance and prematuremortality.

Conclusions: Limb muscle dysfunction is a key systemicconsequence of COPD.However, there are still important gaps in ourknowledge about mechanisms of development of this problem.Strategies for early detection and specific treatments for thiscondition are needed.

This Executive Summary is part of the full official Limb Muscle Dysfunction in COPD Update statement, which readers may access online at www.atsjournals.org/doi/abs/10.1164/rccm.201402-0373ST. Only the Executive Summary is appearing in the print edition of the Journal. The article of record, and the one that shouldbe cited, is: An Official American Thoracic Society/European Respiratory Society Statement: update on limb muscle dysfunction in chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 2014;189:e15–e62. Available at www.atsjournals.org/doi/abs/10.1164/rccm.201402-0373ST.

F.M. holds a CIHR/GSK Research Chair on COPD at Universite Laval. R.C. holds the Grancell/Burns Chair in the Rehabilitative Sciences. E.B.’s contribution tothis statement was supported by CIBERES, FIS 11/02029, 2009-SGR-393, SEPAR 2010, FUCAP 2011, FUCAP 2012, and Marato TV3 (MTV3-07-1010)(Spain) and by the ERS COPD Research Award 2008. M.I.P.’s contribution to this statement was supported by the NIHR Respiratory Disease BiomedicalResearch Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, who partially fund his salary.

Am J Respir Crit Care Med Vol 189, Iss 9, pp 1121–1137, May 1, 2014

Copyright © 2014 by the American Thoracic Society

DOI: 10.1164/rccm.201402-0373ST

Internet address: www.atsjournals.org

American Thoracic Society Documents 1121

Page 2: Upper Limb Muscle Dysfunction Executive Summary

Contents

OverviewIntroductionMethodsScope and DefinitionLimb Muscles in Clinically StableCOPD

Muscle AtrophyStructural AlterationsMitochondrial Function andBioenergetics

Oxidative StressLimb Muscle Function

Limb Muscle Function during COPDExacerbation

Consequences of LimbMuscle Dysfunction in COPD

Etiology of Limb Muscle Dysfunctionin COPD

Disuse versus MyopathyMechanisms of Limb MuscleDysfunction in COPD

Assessment of LimbMuscle Functionin COPD

Effects of Interventions on LimbMuscle Function in COPD

Exercise TrainingOther Interventions

Suggestions for FutureResearch

Conclusions

Overview

Limb muscle dysfunction is an importantsystemic consequence of chronic obstructivepulmonary disease (COPD) because ofits impact on physical activity, exercisetolerance, quality of life, and even survival.Although some mechanisms underlyingdevelopment of limb muscle dysfunctionhave been identified (e.g., deconditioning),much needs to be learned about the impactof other potential contributors to this clinicalmanifestation of COPD. Limb muscledysfunction can be prevented and improved,in part, with exercise training, but it is clearthat novel therapies are needed to betteraddress this problem.

The purpose of this document is toupdate the 1999 American Thoracic Society/European Respiratory Society (ATS/ERS)statement on limb muscle dysfunction. Weintend to provide researchers and clinicianswith recent advances in this field, withemphasis on the following areas: (1)

structural and metabolic alterations foundin limb muscles, (2) consequences andclinical evaluation of limb muscledysfunction, (3) mechanisms ofdevelopment of this comorbidity, and (4)treatment approaches to limb muscledysfunction in COPD. Future researchdirections are also discussed.

Major conclusions of the statementinclude:

d Limb muscle dysfunction is prevalent inCOPD. Muscle atrophy and weaknesscarry important consequences, such asdifficulties in engaging in physical activity,exercise intolerance, poor quality of life,and premature mortality. Metabolicalterations in relation to structuralchanges within the lower limb muscle arealso involved in exercise limitation.

d Lower limb muscle function is furthercompromised during COPDexacerbations. Patients experiencingexacerbations may be targeted forrehabilitative interventions aiming atpreserving limb muscle function.

d Assessment of limb muscle functionshould be encouraged.

d Knowledge of the biochemical regulationof muscle mass will likely lead todevelopment of specific therapy formuscle atrophy in COPD.

d Although physical inactivity is involvedin the development of limb muscledysfunction development in COPD,other mechanisms, such as inflammation,oxidative stress, nutritional imbalance,and hypoxemia, likely play a role.

d The most potent currently availabletreatment option for limb muscledysfunction is exercise training, a keycomponent of integrated COPDmanagement.

d Neuromuscular electrical stimulation isemerging as a useful training modality inseverely impaired COPD and duringexacerbations.

Introduction

Limb muscle atrophy and weakness areprevalent in COPD, affecting up to one-third of patients with COPD seen inspecialized centers (1–3). Furthermore, thisproblem is not only confined to advanceddisease; it may also occur in mild COPD(3). More than 10 years have elapsed since

the first ATS/ERS statement on limbmuscles in COPD (4). Since then, abundantevidence shows that limb muscle atrophyand weakness are strongly related toexercise intolerance, quality of life, andeven premature mortality in COPD(1, 5–7). Another emerging message is thatCOPD exacerbations are associated withfurther decreases in quadriceps strengthand mass (8–10). This finding is relevantbecause the preservation of muscle strengthcould become an important treatment goalduring COPD exacerbation.

The involvement of limb muscles inCOPD goes beyond simple issues of atrophyand weakness. Important morphological andmetabolic alterations have been reported inthis condition at rest and during exercise. Thesechanges lead to premature use of glycolyticmetabolism in the contracting muscles andpromote muscle fatigue and exerciseintolerance (11–13). Together, muscle atrophyand weakness as well as morphological andmetabolic alterations are components of limbmuscle dysfunction in COPD.

Limb muscle weakness and atrophy inCOPD are likely of multifactorial origin, butlittle is known about the interplay and therelative role of the mechanisms underlyingthe phenomena. Molecular and biochemicalmechanisms involved in muscle massregulation in COPD are emerging (14–18).This line of research will, we hope, leadto development of specific therapies formuscle atrophy in COPD. As it stands now,exercise training remains the most potenttherapeutic intervention to improve limbmuscle function in COPD.

The primary objective of this documentis to update current scientific and clinicalknowledge on this topic and to provideguidance for future research directions. Thisdocument should be useful to scientists andalso to clinicians, for whom we wish toraise awareness regarding the clinical relevanceof limb muscle dysfunction.

Methods

The present document is intended to bea Clinical Statement. Methods involved in theproduction of this document are summarizedin Table 1 and in the full document (online).

Scope and Definition

This document focuses on limb muscles, ascommittee members agreed that respiratory

AMERICAN THORACIC SOCIETY DOCUMENTS

1122 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 3: Upper Limb Muscle Dysfunction Executive Summary

muscles are a separate topic. Limb muscledysfunction is used in this document toreflect morphological and functionalchanges seen in limb muscles in patientswith COPD with no implications onunderlying mechanisms.

Limb Muscles in ClinicallyStable COPD

Several structural changes of the limbmuscles have been reported in patients withCOPD. They are described in this sectionand summarized in Figure 1. In comparisonto the quadriceps, upper limb muscles arerelatively spared.

Muscle AtrophyUsing World Health Organizationclassification for body mass index (BMI),underweight in COPD is found in up to 30%of patients with Global Initiative forChronic Obstructive Lung Disease (GOLD)class 4 COPD (2). Studies assessing bodycomposition revealed that reduced musclemass occurs in 4 to 35% of patients withCOPD (2, 19, 20), this proportion beingdependent on the criteria used to definelow muscle mass (1, 2, 21). Low fat-freemass index (FFMI) is reported in 26% ofpatients with COPD with a normal BMI,underscoring the importance of assessingbody composition to quantify preciselymuscle atrophy (2). Even in the presenceof a normal BMI, low FFMI is a strong

predictor of mortality (1). Lower limbmuscles are particularly vulnerable toatrophy in COPD (22, 23).

Structural Alterations

Muscle fiber atrophy and fiber type shift. Allfiber types of the quadriceps are affected bythe atrophying process (24), although someauthors argue that the type IIx fibers aremore specifically affected (25–27). A shiftin quadriceps fiber type distribution, fromtype I to type IIx fibers, is a typical featureof advanced COPD (23, 24, 28, 29). Thisfinding is inconsistent with normal aging,which is not associated with a shift towardtype II fibers (30, 31). The proportion oftype I fibers correlates inversely withdisease severity (32, 33). The fiber typedistribution shift in quadriceps and tibialisanterior (34) muscles is not observed inupper extremity muscles (35), indicatingthat muscle structural abnormalities are nothomogeneously distributed among musclegroups.

Changes in capillarization. Totalnumbers of capillaries and capillariesper muscle fibers are reduced in limbmusclesof COPD (24, 36). This is not a universalfinding (37), perhaps because, in somestudies, patients were involved in exercisetraining, which could improve musclecapillarization (38). However, reduction inthe capillary to muscle fiber cross-sectionalarea has not been reported (39).

Mitochondrial Functionand BioenergeticsMitochondrial functionality is altered inCOPD limb muscles (38). Locomotormuscle oxidative capacity is reduced inCOPD (40, 41), and this is consistentwith type I to type IIx fiber type shift foundin COPD (24, 28). Limb muscle metabolicprofile of patients with COPD shows,at rest, low levels of ATP, inosinemonophosphate, and phosphocreatinelevels compared with age-matched healthycontrol subjects (41, 42). Half-timephosphocreatine recovery after cyclingexercise is slower and phosphocreatineto inorganic phosphate ratio is lower inCOPD than in healthy control subjects(43). In addition, intermediate markers ofglycolysis as well as phosphofructokinaseand lactate dehydrogenase activitiesare elevated in resting COPD muscles(41, 44, 45).

Oxidative StressEvidence of oxidative stress with higherlevels of lipid peroxidation, oxidizedglutathione, and protein oxidation andnitration is consistently found in the bloodand limb muscles of patients with COPD(17, 27, 32, 46–64), particularly those withmuscle atrophy (65). The development ofoxidative stress in limb muscles of patientswith COPD may result from enhancedinflammatory cell infiltration and cytokineproduction. However, there is no strongrelationship between muscle oxidativestress and muscle inflammation in patientswith COPD (17, 27, 52, 62, 64). Muscleoxidative stress may also be related tomitochondrial dysfunction. Interestingly,chronic cigarette smoke exposure alsoinduces a significant increase in severaloxidative stress markers in limb muscles ofhealthy smokers (17) and in limb musclesof animals chronically exposed to cigarettesmoke (17, 66).

Limb Muscle FunctionStrength and endurance are the two maindefining features of muscle function; theycan vary independently in such a way thatone cannot be used to predict the other (67).Strength refers to the ability of the muscleto generate force; endurance is defined asthe ability of the muscle to sustain a giventask. Muscle weakness and reducedendurance are frequent in COPD.Quadriceps strength is reduced by anaverage of 20 to 30% in these individuals

Table 1: Methods

Methods Checklist Yes No

Panel assemblyIncluded experts from relevant clinical and nonclinical disciplines XIncluded individual who represents views of patients and societyat large

X

Included methodologist with documented expertise XLiterature reviewPerformed in collaboration with librarian XSearched multiple electronic databases XReviewed reference lists of retrieved articles X

Evidence synthesisApplied prespecified inclusion and exclusion criteria XEvaluated included studies for sources of bias XExplicitly summarized benefits and harms XUsed PRISMA (252) to report systematic review XUsed GRADE (253) to describe quality of evidence X

Generation of recommendationsUsed GRADE to rate the strength of recommendations X

Definition of abbreviations: GRADE = Grading of Recommendations Assessment, Development andEvaluation; PRISMA = Preferred Reporting Items for Systematic reviews and Meta-Analyses.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1123

Page 4: Upper Limb Muscle Dysfunction Executive Summary

(3, 8, 10, 54, 67–80), and weakness isfound in patients with mild disease (3).Conflicting data exist about the rate ofdecline in quadriceps strength in patientswith COPD, which was reported to beincreased (8) or similar to the healthyaging population (81). Muscle weaknessis not similar among muscle groups:strength of the upper limb muscle isbetter preserved than that of lower limbmuscles (35, 70, 82). Muscle weakness isa direct consequence of reduced musclemass (54, 68, 71, 83, 84), althoughduring chronic or repeated exposure tosystemic corticosteroids, strength lossout of proportion to muscle massreduction can occur (68, 85).

Muscle endurance and fatigue. Volitional(57, 59, 67, 73, 86–89) and nonvolitional(90) quadriceps endurance is decreased inCOPD. Magnitude of decrease is highly

variable (range, 32–77%), probablybecause of differences in test procedures.Impaired quadriceps endurance is alsopresent in patients with mild to moderateCOPD, even in those with relativelynormal physical activity (67). Impairedquadriceps endurance is the consequenceof reduced oxidative capacity andoxidative stress (73), rather than reducedmuscle mass (83). Endurance of theupper extremity muscles (69, 74, 91) ispreserved in patients with COPD,providing additional indication aboutheterogeneity of muscle abnormalities inthis disease.

Fatigue is defined as a failure of forcegeneration after loaded muscle contractionsthat is reversible by rest (87, 92). Quadricepsfatigue can be objectively demonstrated in48 to 58% of patients with COPD aftercycling exercise (6, 72, 79, 93). Taking into

account the amount of work performed, thesepatients exhibit greater susceptibility tofatigue than healthy individuals (72).The gastrocnemius and tibialis anteriorof patients with COPD may also be moresusceptible to fatigue during walking(80, 94). Susceptibility to muscle fatiguein COPD can be related to intrinsicmuscle morphometric and metabolicderangements seen during exercise in thisdisease (11,13, 95–97). Insufficientoxygen delivery to contractile muscle mayalso contribute to premature lower limbmuscle fatigue (98, 99).

Limb Muscle Function duringCOPD Exacerbation

Quadriceps strength often decreases duringhospitalization for COPD exacerbation(10, 100, 101). Reduction in quadriceps

Figure 1. Morphological and structural alterations reported in limb muscles in patients with chronic obstructive pulmonary disease (COPD). CS = citratesynthase; HADH = 3-hydroxyacyl CoA dehydrogenase.

AMERICAN THORACIC SOCIETY DOCUMENTS

1124 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 5: Upper Limb Muscle Dysfunction Executive Summary

force during hospitalization is associatedwith less improvement in walking time1 month after discharge (100). Importantly,quadriceps force only partially recovered3 months after hospital discharge (10).Muscle weakness has been associated withincreased risk of hospital readmissionsdue to acute exacerbation (102). Musclemass and strength maintenance may becompromised during an exacerbation,with activation of multiple atrophyingbiochemical pathways during this process(101, 103). Causes of muscle dysfunctionduring exacerbations may involveinflammation (10), impaired energybalance (104, 105), inactivity (100, 106),and systemic corticosteroid use (85).

Consequences of LimbMuscle Dysfunction in COPD

The most troublesome consequence ofmuscle dysfunction is its association with

reduced life expectancy (1, 5, 7) (Figure 2).Muscle atrophy (1, 5) and quadricepsweakness (7) are predictors of mortality insubjects with COPD: (1) midthigh musclecross-sectional area less than 70 cm2 asassessed by computed tomographyscanning is associated with fourfoldincrease in mortality after adjusting forage, sex, and FEV1 (5); (2) FFMI less than16 kg/m2 in men and less than 15 kg/m2 inwomen is associated with 1.9-fold increasein mortality after adjusting for age, sex, andFEV1 (1); and (3) a quadriceps strength(kg) to BMI (kg/m2) ratio less than 120%is associated with increased mortality; each10% increment of this ratio is associatedwith a 9% reduction in mortality (7). Theseindicators stress the importance forclinicians to carefully monitor bodycomposition and muscle strength inpatients with COPD. Muscle atrophy andweakness are also associated with reducedquality of life (107) and greater healthcare

resource use (108). Quadriceps strengthis a strong predictor of exercise capacity inpatients with pulmonary diseases (109):a twofold increase in muscle strength isassociated with 1.4- to 1.6-fold increase inwork capacity (110). One explanation forthis is the influence of muscle strengthon leg effort perception during exercise(110), the main limiting symptom in anappreciable portion of patients withCOPD (111) (Figure 2). Premature legfatigue reduces the ability ofbronchodilators to produce improvementin exercise tolerance (6, 112).Perturbation in muscle energymetabolism during exercise seen inCOPD is also relevant to whole-bodyexercise capacity, as it is associated withpremature fatigue (11, 13, 95–97) andincreased ventilatory requirements duringexercise (46, 113, 114), imposing anadditional burden on already overloadedrespiratory muscles.

Figure 2. Relationships between muscle mass and strength and clinical outcomes in patients with chronic obstructive pulmonary disease (COPD). Amidthigh muscle cross-sectional area (MTCSA), 70 cm2 (A) (5), a low fat-free mass index (FFMI) defined as an FFMI , 16 kg/m2 in men and , 15 kg/m2

in women (B) (1), and a reduced quadriceps strength defined as a quadriceps strength (kg) to body mass index (BMI, kg/m2) ratio , 120% (C) (7)are predictors of mortality in COPD after adjusting for traditional mortality risk factor such as age and FEV1. The strength of the quadriceps is a significantcontributor to exercise capacity in COPD (D) (110). All panels adapted by permission from the indicated references.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1125

Page 6: Upper Limb Muscle Dysfunction Executive Summary

Etiology of Limb MuscleDysfunction in COPD

Several factors have been hypothesized toinitiate and/or promote changes in limbmusclesof patients with COPD (Table 2). Together,they have the ability to activate variousbiochemical pathways initiating or enhancingalterations in fiber type expression, contractileproteolysis, metabolic alterations, andregenerative defects in limb muscles.

Disuse versus MyopathyOne important question is whether limbmuscle dysfunction simply reflects years of

physical inactivity or whether some COPD-specific myopathy (115–119) can beinvoked. Against the hypothesis of COPDmyopathy are observations that manyfunctional and cellular findings in COPDare identical to those of disuse and thatlimb muscle function and oxidative profilecan be improved with training (115, 118,120–124). The fact that full recovery ofmuscle function is unusual does notnecessarily indicate a myopathy, becauseintensity and/or duration of training maybe insufficient to correct alterations thathave developed over several decades (119,125, 126). On the other hand, several

observations favor the existence ofa myopathy in patients with COPD. Musclestructure and function are poorly related tothe degree of physical activity (127–129).Disparities in muscle function and/orstructure remain when patients with COPDare compared with control subjectshaving similar degree of physical activity(116, 130). Muscle typology does notimprove to the same extent after exercise inCOPD (24, 131, 132) compared withhealthy subjects (133–135).

Mechanisms of Limb MuscleDysfunction in COPD

Inflammation. Through transcriptionalactivities of nuclear factor-kappa B (NF-kB)and forkhead box O (FOXO) (136),production of proinflammatory cytokinesduring the inflammatory response canenhance ubiquitin proteasome systemactivity, apoptosis (137), andmacroautophagy occurrence (138), whichhave all been linked to muscle atrophydevelopment (139, 140). Increasedexpression of E3-ligases Atrogin-1, musclering finger protein 1 (MuRF-1), and neuralprecursor cell expressed developmentallydown-regulated protein 4 (Nedd4) isobserved in quadriceps of patients withCOPD (14, 15). Despite these convincingarguments supporting a role forinflammation, data supporting its presencein quadriceps of patients with COPD areequivocal (45, 52, 141–144), even inestablished muscle atrophy and duringperiods of acute exacerbation wheninflammatory bursts should be expected(103).

Oxidative stress. In COPD, systemic(56) and local (27, 48, 49) oxidative stresshas been reported at rest, during acuteexercise bouts (54, 56), and during acuteexacerbation (145, 146). Oxidative stresslevels are directly related to quadriceps forceand endurance in patients with severeCOPD (17, 52, 53, 57, 59). Oxidative stresscould alter the integrity of proteins,enhancing their degradation (147). Directoxidative stress exposure (148) or indirectproduction of reactive oxygen speciesthrough inflammatory response (149, 150)induces proteolysis and increases expressionof ubiquitin-proteasome components.Induction of NF-kB (149) and FOXO (151)transcriptional activities or p38 kinaseactivity (18, 152) by oxidative stress arebelieved to regulate proteolytic signaling.

Table 2: Etiologies of Limb Muscle Atrophy, Weakness, and Susceptibility to Fatigue

Mechanisms Involved

Factors leading to muscle atrophy andweakness

Disuse Associated with weakness, atrophy,changes in fiber type distribution andmetabolic alterations (118, 120–123)

Inflammation Triggering of the muscle proteolysiscascade (14, 15, 139, 140)

Oxidative stress Triggering of the muscle proteolysiscascade (147, 149, 150)

Associated with reduced muscleendurance (51, 57, 59)

Protein carbonylation possibly involved inexercise intolerance and weakness (55)

Hypoxemia Decreased muscle protein synthesisActivation of muscle degradation throughhypoxia-inducible factor/vonHippel–Lindau signaling cascade(158, 254–256)

Hypercapnia Intracellular acidosis/alterations incontractile protein synthesis/degradation(163, 164)

Low levels of anabolic hormones andgrowth factors

Associated with reduced muscle proteinsynthesis (257, 258)

Impaired energy balance Associated with reduced muscle proteinsynthesis (175, 259)

Corticosteroids Reduced muscle protein synthesis andenhanced proteolysis through increasedmyostatin levels and reduced insulin-likegrowth factor-1 levels (178)

Vitamin D deficiency Associated with muscle weakness, type IIatrophy impaired calcium metabolism(180, 184, 260)

Factors leading to muscle susceptibilityto fatigue

Central fatigue—afferent feedback fromlimb muscles

Reduced motor output to the contractingmuscles (261)

Reduced O2 delivery (impaired cardiacoutput, blood flow competitionbetween the respiratory and limbmuscle, reduced capillarity)

Changes in muscle metabolism in favor ofglycolysis; accumulation of musclemetabolites associated with musclefatigue

Muscle metabolic alteration (reducedoxidative enzyme activity, reducedmitochondrial function)

Preferential use of glycolysis andaccumulation of muscle metabolitesassociated with muscle fatigue (45, 96,262–264)

AMERICAN THORACIC SOCIETY DOCUMENTS

1126 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 7: Upper Limb Muscle Dysfunction Executive Summary

Hypoxia. Hypoxic conditions decreasemuscle mass (153, 154). Patients withCOPD having reduced arterial PO2 (155)or O2 delivery (156) tend to have lowerbody mass than those with preservedoxygenation. Biochemical mechanismsinvolved in the negative muscle massregulation by hypoxemia are complex andinvolve hypoxia inducible factor-1 signalingcascade (157, 158). Both inflammatoryresponse (159) and increased productionof reactive oxygen species (59, 160) asa reaction to hypoxia have been hypothesizedto link hypoxia to cellular signaling yieldingmuscle atrophy. Hypoxemia also may alsocompromise muscle oxidative capacity andcapillarization (161).

Hypercapnia. Increase in cellularCO2 content in chronic hypercapniamay worsen in exacerbated patients.Consequently, tissue pH decreases (162)and acidosis can alter contractile proteinsynthesis and degradation (163). In muscletissue, acidosis increases expression ofgenes encoding proteins of the ubiquitin-proteasome pathway (164) and impairsinsulin/IRS/AKT signaling (165).

Low levels of anabolic hormones andgrowth factors. Low testosterone levelshave been reported in COPD (166–170).However, association of limb musclefunction indices with serum testosteronelevels in COPD is inconsistent acrossstudies (166, 169). Myostatin, atransforming growth factor-b (TGF-b)family member that acts as a negativeregulator of limb muscle growth (171, 172),may be elevated in blood and muscles inCOPD (16, 173, 174).

Impaired energy balance. Muscleatrophy may result from disturbed balancebetween energy expenditure and dietaryintake, which may affect muscle proteinsynthesis rate, thus creating an imbalancebetween protein synthesis and breakdown.Altered appetite regulation, oxidative stress,and systemic inflammationmay all contributeto loss in muscle tissue in COPD (175).

Corticosteroids. Although short-termexposure to systemic corticosteroids doesnot appear to compromise limb musclestrength and function (176), chronic orrepeated exposure can potentiate muscleatrophy and weakness (85, 177).Corticosteroids decrease protein synthesisand simultaneously promote proteindegradation (178). These actions on proteinsynthesis are believed to occur throughinhibition of key hypertrophic biochemical

pathways. Muscle protein degradation inthe context of corticosteroids exposureis related to activation of ubiquitin-proteasome and lysosomal systems, twomajor proteolytic pathways (178).

Vitamin D deficiency. Vitamin Ddeficiency is highly prevalent in COPDcompared with age-matched smokingcontrol subjects (179), and it is suggestedthat vitamin D deficiency could contributeto limb muscle dysfunction (180, 181).However, published evidence isunconvincing in this regard (182, 183). Inone study, genetic polymorphisms in thevitamin D receptor associated withquadriceps strength (184), indicating thatthe vitamin D pathway may affect muscleindependent of serum levels.

Smoking. Cigarette smoking is unlikelyto be the main mechanism involved in limbmuscle dysfunction in COPD because,in several studies, smoking history wasmatched in patients with COPD and healthycontrol subjects. However, smoking by itselfmay be associated with muscle atrophyand weakness in otherwise healthy subjects (3,64, 81, 185–188). Smoking is also associatedwith decreased type I fiber cross-sectionalarea, reduced type I fiber proportion, reducedcytochrome oxidase activity, increased lactatedehydrogenase activity, and higher level ofprotein oxidation in the quadriceps(189–191).

Assessment of Limb MuscleFunction in COPD

The existing relationships between limbmuscle mass and strength and importantclinical outcomes in patients with COPD(see previous section on the consequencesof limb muscle dysfunction) suggest thatassessing body composition and limbmuscle strength in the clinical evaluation ofCOPD can identify patients who are atincreased risk for exercise intoleranceand premature mortality. Bioelectricalimpedance and dual-energy X-rayabsorptiometry provide reasonably accurateassessment of body composition (192).Learning about limb muscle status mayhelp clinicians understand the mechanismsof exercise limitation in a given patient.Limb muscle strength assessment is alsouseful to prescribe adequate loads forresistance training. One strategy could be toevaluate body composition and quadricepsstrength at the time of referral to theexercise laboratory for assessment of

dyspnea and/or exercise tolerance. Someexercise laboratories implemented thispractice several years ago (110).

Various methodologies exist to measuremuscle strength (193), including handhelddynamometry (70, 194), the one-repetitionmeasurement (the maximum value thata patient can move over the full range ofmotion) (195, 196), and hydraulic resistances(68, 197). They are discussed morecompletely in the full document (online).

We favor using strain gauges tomeasure isometric maximal quadricepsstrength; this methodology could beimplemented in clinical practice to providereliable and reproducible measurements(198, 199) (Figure 3). The maximalvoluntary contraction force hashistorically been reported in kilograms,because weights are used to calibrate theapparatus. Force could also be reportedin Newtons, the product of force inkilograms and gravitational pull (9.81 m/s2).Maximal voluntary contraction force istypically reported as the best of threereproducible maneuvers. Isometric muscleforce can also be assessed on specificallybuilt computerized dynamometers (e.g.,Cybex or Biodex), but these devices areexpensive and not widely available. Onecurrent limitation of assessment ofquadriceps strength is that there are nowidely accepted reference values. Thiscould be circumvented by expressingquadriceps strength in kilograms asa percentage of BMI (7). Another limitationof volitional assessment of strength is itsdependence on patient cooperation.Nonvolitional assessment of limb musclestrength involves electrical or magneticstimulation of a peripheral nerve (6, 78).This technique is currently practiced onlyin research laboratories.

Effects of Interventions onLimb Muscle Function inCOPD

The effects of interventions designed toimprove limb muscle mass and function aresummarized in Table 3.

Exercise TrainingExercise training is the only interventionthat can be indisputably recommended fortreatment of limb muscle dysfunction inCOPD. Rehabilitative exercise trainingimproves limb muscle function and

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1127

Page 8: Upper Limb Muscle Dysfunction Executive Summary

morphology in patients with COPD (93,125, 126, 200–203). Endurance exercisetraining (either of interval or constant-loadmodality) improves the cross-sectional area(CSA) of all fiber types within the vastuslateralis muscle (24, 33, 204, 205). Inaddition, endurance training has beenreported to modify quadriceps muscle fibertype distribution in favor of type I fibers

(204–206). These morphological andtypological adaptations in limb musclefibers are not different across GOLD classes2 to 4 (33). Exercise training also shiftsmuscle energy metabolism from glycolyticto oxidative metabolism (41, 43).Phenotypical changes within limb musclesare accompanied by positive adaptations inmuscle mass regulating pathways (132, 196,

205, 206). Resistance exercise, alone or incombination with aerobic exercise, alsohelps to improve muscle mass and strengthin COPD (197, 201, 203, 207).

An important question is whether theresponse to exercise training is normal inCOPD. Muscle angiogenic and molecularresponse to training may be blunted inCOPD (208, 209); whether this is entirelyexplained by an insufficient trainingintensity is uncertain. Gene expressionresponse after endurance training inpatients with COPD differs from that seenin healthy sedentary subjects (210).Qualitatively, genes associated withoxidative stress, ubiquitin proteasome, andcyclooxygenase pathways are distinctlyinduced in COPD, potentially reflectingspecific molecular response of muscleto exercise and suggesting additionalmechanisms for exercise limitation (210).The occurrence of oxidative (61) andnitrosative (53) stress at the muscle levelhas been reported after exercise training;cachectic patients seem to be more prone tothis effect (211). However, the functionalsignificance of these observations is unclear(209), and exercise training is a safe andeffective intervention to treat limb muscledysfunction in COPD.

The magnitude of response topulmonary rehabilitation in COPD is highlyvariable among patients (212), and thereseems also to be a genetic component tothis phenomenon (213, 214). Patientsdeveloping quadriceps contractile fatigueduring training sessions show greatertraining effects in terms of functionalexercise capacity and health-related qualityof life than those who do not (215). Thisargues in favor of training intensity beingan important determinant of exercisetraining outcome (62, 113, 216).

Exercise training early after anexacerbation. Several interventions toprevent or counteract muscle impairmentduring episodes of exacerbation have beenconsidered (217). Resistance traininginitiated during the second day ofhospitalization may counteract limbmuscle dysfunction: quadriceps forceincreased by 10%, and 6-minute walkdistance improved at discharge (9). Thiswas associated with a more favorableanabolic/catabolic balance in muscle (9).Moreover, 1 month after discharge,functional status and muscle forceremained better in the group that trainedduring the exacerbation (9).

Figure 3. Standard operating procedure for isometric quadriceps strength assessment. During themaneuver, vigorous encouragement of the patient is needed. Patient is positioned in a standardizedfashion (typically sitting with knees and hips in 908 flexion or, less often, supine [3]). Maximal voluntarycontraction force (reported in kilograms or Newtons) can be reliably assessed as the best of threereproducible maneuvers. Maximal voluntary contraction is recorded as the maximal force that can bemaintained for 1 full second.

AMERICAN THORACIC SOCIETY DOCUMENTS

1128 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 9: Upper Limb Muscle Dysfunction Executive Summary

Neuromuscular stimulation. Transcutaneousneuromuscular electrical stimulation canbe particularly useful for severely disabledpatients with COPD (218, 219), as theload on the cardiopulmonary system is low(220). Indeed, it may even be consideredfor home use (221) and in patients whoare unstable (222). Increase in midthighmuscle and type II fiber CSA, decrease infiber type I CSA, change in fiber typedistribution in favor of type I fibers,decreased muscle oxidative stress, alongwith a more favorable anabolic to catabolicbalance have been reported (222–224).Larger trials are needed to clarify theoptimal stimulation parameters(frequency, intensity, and duration ofstimulation) and in which population thistraining option should be offered.

Other InterventionsSeveral interventions have beeninvestigated for their ability to improvelimb muscle mass and function (Table 3).These interventions have not reachedwidespread clinical application becauseof uncertain efficacy and potential ofadverse effects.

Nutritional supplementation has beeninvestigated in COPD (225, 226). As bodyweight gain is associated with improvedprognosis in COPD (227), efforts should bemade to establish appropriate treatmentsfor the underweight patients. A caveat isthat weight gain may predominantly consistof fat mass with little effect on limb musclemass and function (226, 228). Combination ofnutritional intervention with an anabolicstimulus, such as exercise training, is

warranted to ensure that nutritionalintervention impacts muscle mass andfunction (229). Patients with COPD arevulnerable to nutritional depletionduring an acute disease exacerbation.Adequate nutritional support, especiallyin patients with already impaired energybalance, is also important (105, 230).Although some improvement in FFM andin muscle strength can be achieved withnutritional intervention, particularlywhen coupled with an anabolic stimulus(229), nutritional interventions are notwidely used because of equivocal benefits(231). Well-designed randomizedcontrolled trials are needed to addressthis question.

A variety of anabolic drugs andbioactive nutrients have been tested withthe hope of improving muscle mass andstrength in COPD. None of them canbe recommended as routine treatment.Testosterone supplementation and itsanalogs are able to improve muscle strengthand mass (196, 232–236), but this does notnecessarily translate into improvedfunctional capacity. Also, the potential ofadverse events, including carcinogenesisand virilization, has limited the applicationof this intervention in clinical practice.Growth hormone and its analogs(237–241), megestrol acetate (242), creatine(243–246), L-carnitine (247), antioxidants(57, 248–250), and vitamin Dsupplementation (183, 251), alone or incombination with exercise training, havebeen tested in COPD. The role of thesetherapies is uncertain, and additionalstudies are warranted.

Suggestions for FutureResearch

Despite progressmade since the first ATS/ERSStatement on limb muscle in COPD, muchremains to be learned about this importantsystemic consequence. In the context ofa worldwide epidemic of obesity, the diagnosisof muscle atrophy may become more difficultas clinicians are likely to be misled bynormal or increased BMI, believing that thisimplies normal muscle mass. Althoughmuscle atrophy is less prevalent when BMI isnormal or increased, there is still anappreciable portion of patients who expressesmuscle atrophy and weakness (1).

There are some recommendations forfuture research:

1. If the question about presence of disuseversus specific COPD-related myopathy isto be answered, patients with COPDshould be matched with control subjectshaving a similar degree of physical activity,as assessed with activity monitors.

2. Widely accepted normative values forquadriceps muscle strength should bedetermined.

3. There is a need to know more about theonset of limb muscle dysfunction inCOPD. A focus on patientswith mild disease would be ofinterest. Longitudinal studies willbe important to understand whenthe muscle pathological processesstart and how they evolve over time.

4. Investigation of molecular mechanismsinvolved in initiation and developmentof limb muscle dysfunction is a key issue

Table 3: Effects of Treatments for Limb Muscle Dysfunction in Chronic Obstructive Pulmonary Disease

Treatment Mass Strength Exercise Tolerance Survival

Exercise O (201) O (200, 201) O (265) ?Oxygen ? ? O (266–268) ONutrition alone No (269) No (269) No (269) ?Nutrition with exercise training O (228, 229, 270) O (229, 270) O (228, 229, 270) ?Nutrition with exercise training and anabolichormone supplementation

O (271) O (271) O (271) ?

Testosterone O (196) O (196) No (196, 233) ?Growth hormones O (240) No (240) No (240) ?Ghrelin ? ? ? ?Megestrol No (242) ? No (242) ?Creatine ? ? No (243, 244, 246) ?Antioxidants ? ? ? ?Vitamin D alone ? ? ? ?Vitamin D with exercise training ? ? ? ?

O: Studies support that the treatment has a favorable effect on the outcome; No: studies support that the treatment has no favorable effect on theoutcome; ?: there are no supporting data for a treatment effect on the outcome.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1129

Page 10: Upper Limb Muscle Dysfunction Executive Summary

for development of specific and safetherapy for this problem.

5. Clinical trials are warranted to evaluateto which extent treatment of limbmuscle function affects clinicaloutcomes (exercise capacity, quality oflife, and survival) in COPD.

6. Large and multicenter studies inthoroughly phenotyped patients will beinstrumental in understanding specificrisk factors for developing limb muscledysfunction and evaluating treatmentfor this condition.

7. Whether muscle abnormalities can becompletely normalized with exercisetraining is a question that should beaddressed specifically in an adequatelypowered clinical trial.

8. In the context of increasingprevalence of (abdominal) obesity inCOPD, emphasis should be placedon possible biochemical cross-talkbetween fat and muscle. For example,it may be that limb muscledysfunction influences prevalence ofmetabolic syndrome.

Conclusions

Limb muscle dysfunction is a clinicallyrelevant systemic manifestation ofCOPD, because it influences importantclinical outcomes. This comorbidcondition can be treated with exercisetraining. Future research shouldallow a better understanding ofmechanisms involved in developmentof skeletal muscle dysfunction, with thehope for development of specifictherapies. n

This statement was prepared by an ad hoc subcommittee of the ATS Assembly on Pulmonary Rehabilitation and the ERS Scientific Group 01.02“Rehabilitation and Chronic Care.”

Members of this committee were as follows:

FRANCOIS MALTAIS, M.D. (Chair)MARC DECRAMER, PH.D., M.D. (Co-Chair)ESTHER BARREIRO, M.D., PH.D.YAN BURELLE, PH.D.RICHARD CASABURI, PH.D., M.D.RICHARD DEBIGARE, PH.D.P. N. RICHARD DEKHUIJZEN, M.D., PH.D.FRITS FRANSSEN, M.D., PH.D.GHISLAINE GAYAN-RAMIREZ, PH.D.JOAQUIM GEA, M.D.HARRY R. GOSKER, PH.D.RIK GOSSELINK, PH.D.MAURICE HAYOT, PH.D., M.D.SABAH N. A. HUSSAIN, PH.D., M.D.WIM JANSSENS, PH.D., M.D.MICHAEL I. POLKEY, PH.D.JOSEP ROCA, M.D.DIDIER SAEY, P.T., PH.D.ANNEMIE M. W. J. SCHOLS, PH.D.

MARTIJN A. SPRUIT, PH.D.MICHAEL STEINER, M.D.TANJA TAIVASSALO, PH.D.THIERRY TROOSTERS, PH.D.IOANNIS VOGIATZIS, PH.D.PETER D. WAGNER, M.D.

Author Disclosures: F.M. was on an advisorycommittee and received speaker fees fromGlaxoSmithKline ($5,000–24,999 combined); hereceived research support from GlaxoSmithKline($50,000–99,999). R.C. received researchsupport from Novartis ($100,000–249,999). J.G.holds a patent for a training valve, with proceedspaid to his institution. W.J. received speaker feesfrom AstraZeneca ($1–4,999), was on anadvisory committee and received speaker feesfrom Boehringer Ingelheim ($1–4,999combined), and received research supportfrom Boehringer Ingelheim ($5,000–24,999);

he was on an advisory committee of Chiesi($1–4,999), and on an advisory committeeand received speaker fees fromGlaxoSmithKline ($1–4,999 combined); hewas on an advisory committee and receivedspeaker fees from Novartis ($1–4,999combined). M.D., E.B., Y.B., R.D., P.N.R.D.,F.F., G.G-R., H.R.G., R.G., M.H., S.N.A.H.,M.I.P., J.R., D.S., A.M.W.J.S., M.A.S.,M.S., T. Taivassalo, T. Troosters., I.V., andP.D.W. reported no relevant commercialinterests.

Acknowledgment: The authors thankAnnette De Bruyne and Eline Lahousse fortheir secretarial assistance and Helene Trudel,for drawing the figures. They also thankKevin Wilson and Guy Brusselle for theirguidance during the preparation of thisdocument.

References

1. Schols AM, Broekhuizen R, Weling-Scheepers CA, Wouters EF. Bodycomposition and mortality in chronic obstructive pulmonary disease.Am J Clin Nutr 2005;82:53–59.

2. Vestbo J, Prescott E, Almdal T, Dahl M, Nordestgaard BG, Andersen T,Sørensen TI, Lange P. Body mass, fat-free body mass, and prognosisin patients with chronic obstructive pulmonary disease from a randompopulation sample: findings from the Copenhagen City Heart Study.Am J Respir Crit Care Med 2006;173:79–83.

3. Seymour JM, Spruit MA, Hopkinson NS, Natanek SA, Man WD, JacksonA, Gosker HR, Schols AM, Moxham J, Polkey MI, et al. The prevalenceof quadriceps weakness in COPD and the relationship with diseaseseverity. Eur Respir J 2010;36:81–88.

4. Skeletal muscle dysfunction in chronic obstructive pulmonarydisease: a statement of the American Thoracic Society andEuropean Respiratory Society. Am J Respir Crit Care Med 1999;159:S1–S40.

5. Marquis K, Debigare R, Lacasse Y, LeBlanc P, Jobin J, Carrier G,Maltais F. Midthigh muscle cross-sectional area is a better predictor ofmortality than body mass index in patients with chronic obstructivepulmonary disease. Am J Respir Crit Care Med 2002;166:809–813.

6. Saey D, Debigare R, LeBlanc P, Mador MJ, Cote CH, Jobin J, Maltais F.Contractile leg fatigue after cycle exercise: a factor limiting exercise inpatients with chronic obstructive pulmonary disease. Am J Respir CritCare Med 2003;168:425–430.

7. Swallow EB, Reyes D, Hopkinson NS, Man WD, Porcher R, Cetti EJ,Moore AJ, Moxham J, Polkey MI. Quadriceps strength predictsmortality in patients with moderate to severe chronic obstructivepulmonary disease. Thorax 2007;62:115–120.

8. Hopkinson NS, Tennant RC, Dayer MJ, Swallow EB, Hansel TT, MoxhamJ, Polkey MI. A prospective study of decline in fat free mass andskeletal muscle strength in chronic obstructive pulmonary disease.Respir Res 2007;8:25.

9. Troosters T, Probst VS, Crul T, Pitta F, Gayan-Ramirez G, Decramer M,Gosselink R. Resistance training prevents deterioration in quadricepsmuscle function during acute exacerbations of chronic obstructivepulmonary disease. Am J Respir Crit Care Med 2010;181:1072–1077.

10. Spruit MA, Gosselink R, Troosters T, Kasran A, Gayan-Ramirez G,Bogaerts P, Bouillon R, Decramer M. Muscle force during an acuteexacerbation in hospitalised patients with COPD and its relationshipwith CXCL8 and IGF-I. Thorax 2003;58:752–756.

11. Steiner MC, Evans R, Deacon SJ, Singh SJ, Patel P, Fox J, GreenhaffPL, Morgan MD. Adenine nucleotide loss in the skeletal musclesduring exercise in chronic obstructive pulmonary disease. Thorax2005;60:932–936.

12. Calvert LD, Shelley R, Singh SJ, Greenhaff PL, Bankart J, Morgan MD,Steiner MC. Dichloroacetate enhances performance and reduces bloodlactate during maximal cycle exercise in chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 2008;177:1090–1094.

13. Saey D, Lemire BB, Gagnon P, Bombardier E, Tupling AR, Debigare R,Cote CH, Maltais F. Quadriceps metabolism during constantworkrate cycling exercise in chronic obstructive pulmonary disease.J Appl Physiol (1985) 2011;110:116–124.

AMERICAN THORACIC SOCIETY DOCUMENTS

1130 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 11: Upper Limb Muscle Dysfunction Executive Summary

14. Doucet M, Russell AP, Leger B, Debigare R, Joanisse DR, Caron MA,LeBlanc P, Maltais F. Muscle atrophy and hypertrophy signaling inpatients with chronic obstructive pulmonary disease. Am J RespirCrit Care Med 2007;176:261–269.

15. Plant PJ, Brooks D, Faughnan M, Bayley T, Bain J, Singer L, Correa J,Pearce D, Binnie M, Batt J. Cellular markers of muscle atrophy inchronic obstructive pulmonary disease. Am J Respir Cell Mol Biol2010;42:461–471.

16. Man WD, Natanek SA, Riddoch-Contreras J, Lewis A, Marsh GS, KempPR, Polkey MI. Quadriceps myostatin expression in COPD. EurRespir J 2010;36:686–688.

17. Barreiro E, Peinado VI, Galdiz JB, Ferrer E, Marin-Corral J, Sanchez F,Gea J, Barbera JA; ENIGMA in COPD Project. Cigarette smoke-induced oxidative stress: a role in chronic obstructive pulmonarydisease skeletal muscle dysfunction. Am J Respir Crit Care Med2010;182:477–488.

18. Lemire BB, Debigare R, Dube A, Theriault ME, Cote CH, Maltais F.MAPK signaling in the quadriceps of patients with chronicobstructive pulmonary disease. J Appl Physiol (1985) 2012;113:159–166.

19. Schols AM, Soeters PB, Dingemans AM, Mostert R, Frantzen PJ,Wouters EF. Prevalence and characteristics of nutritional depletion inpatients with stable COPD eligible for pulmonary rehabilitation. AmRev Respir Dis 1993;147:1151–1156.

20. Coronell C, Orozco-Levi M, Gea J. COPD and body weight ina Mediterranean population. Clin Nutr 2002;21:437–438, author reply437–438.

21. Schutz Y, Kyle UU, Pichard C. Fat-free mass index and fat mass indexpercentiles in Caucasians aged 18-98 y. Int J Obes Relat MetabDisord 2002;26:953–960.

22. Engelen MP, Schols AM, Does JD, Wouters EF. Skeletal muscleweakness is associated with wasting of extremity fat-freemass but not with airflow obstruction in patients with chronicobstructive pulmonary disease. Am J Clin Nutr 2000;71:733–738.

23. Caron MA, Debigare R, Dekhuijzen PN, Maltais F. Comparativeassessment of the quadriceps and the diaphragm in patients withCOPD. J Appl Physiol (1985) 2009;107:952–961.

24. Whittom F, Jobin J, Simard PM, Leblanc P, Simard C, Bernard S,Belleau R, Maltais F. Histochemical and morphologicalcharacteristics of the vastus lateralis muscle in patients with chronicobstructive pulmonary disease. Med Sci Sports Exerc 1998;30:1467–1474.

25. Gosker HR, Engelen MP, van Mameren H, van Dijk PJ, van der VusseGJ, Wouters EF, Schols AM. Muscle fiber type IIX atrophy is involvedin the loss of fat-free mass in chronic obstructive pulmonary disease.Am J Clin Nutr 2002;76:113–119.

26. Natanek SA, Riddoch-Contreras J, Marsh GS, Hopkinson NS, Man WD,Moxham J, Polkey MI, Kemp PR. Yin Yang 1 expression andlocalisation in quadriceps muscle in COPD. Arch Bronconeumol2011;47:296–302.

27. Fermoselle C, Rabinovich R, Ausın P, Puig-Vilanova E, Coronell C,Sanchez F, Roca J, Gea J, Barreiro E. Does oxidative stressmodulate limb muscle atrophy in severe COPD patients? Eur RespirJ 2012;40:851–862.

28. Gosker HR, Zeegers MP, Wouters EF, Schols AM. Muscle fibre typeshifting in the vastus lateralis of patients with COPD is associatedwith disease severity: a systematic review and meta-analysis. Thorax2007;62:944–949.

29. Kim HC, Mofarrahi M, Hussain SN. Skeletal muscle dysfunction inpatients with chronic obstructive pulmonary disease. Int J ChronObstruct Pulmon Dis 2008;3:637–658.

30. Lexell J, Taylor CC, Sjostrom M. What is the cause of the ageingatrophy? Total number, size and proportion of different fiber typesstudied in whole vastus lateralis muscle from 15- to 83-year-old men.J Neurol Sci 1988;84:275–294.

31. Larsson L. Histochemical characteristics of human skeletal muscleduring aging. Acta Physiol Scand 1983;117:469–471.

32. Doucet M, Debigare R, Joanisse DR, Cote C, Leblanc P, Gregoire J,Deslauriers J, Vaillancourt R, Maltais F. Adaptation of the diaphragmand the vastus lateralis in mild-to-moderate COPD. Eur Respir J2004;24:971–979.

33. Vogiatzis I, Terzis G, Stratakos G, Cherouveim E, Athanasopoulos D,Spetsioti S, Nasis I, Manta P, Roussos C, Zakynthinos S. Effect ofpulmonary rehabilitation on peripheral muscle fiber remodeling inpatients with COPD in GOLD stages II to IV. Chest 2011;140:744–752.

34. Eliason G, Abdel-Halim S, Arvidsson B, Kadi F, Piehl-Aulin K. Physicalperformance and muscular characteristics in different stages ofCOPD. Scand J Med Sci Sports 2009;19:865–870.

35. Gea JG, Pasto M, Carmona MA, Orozco-Levi M, Palomeque J,Broquetas J. Metabolic characteristics of the deltoid muscle inpatients with chronic obstructive pulmonary disease. Eur Respir J2001;17:939–945.

36. Jobin J, Maltais F, Doyon JF, LeBlanc P, Simard PM, Simard AA,Simard C. Chronic obstructive pulmonary disease: capillarity andfiber-type characteristics of skeletal muscle. J Cardiopulm Rehabil1998;18:432–437.

37. Richardson RS, Leek BT, Gavin TP, Haseler LJ, Mudaliar SR, Henry R,Mathieu-Costello O, Wagner PD. Reduced mechanical efficiency inchronic obstructive pulmonary disease but normal peak VO2 withsmall muscle mass exercise. Am J Respir Crit Care Med 2004;169:89–96.

38. Rabinovich RA, Vilaro J. Structural and functional changes of peripheralmuscles in chronic obstructive pulmonary disease patients. CurrOpin Pulm Med 2010;16:123–133.

39. Eliason G, Abdel-Halim SM, Piehl-Aulin K, Kadi F. Alterations in themuscle-to-capillary interface in patients with different degrees ofchronic obstructive pulmonary disease. Respir Res 2010;11:97.

40. Gosker HR, Hesselink MK, Duimel H, Ward KA, Schols AM. Reducedmitochondrial density in the vastus lateralis muscle of patients withCOPD. Eur Respir J 2007;30:73–79.

41. Green HJ, Burnett ME, D’Arsigny CL, O’Donnell DE, Ouyang J, WebbKA. Altered metabolic and transporter characteristics of vastuslateralis in chronic obstructive pulmonary disease. J Appl Physiol(1985) 2008;105:879–886.

42. Pouw EM, Schols AM, van der Vusse GJ, Wouters EF. Elevated inosinemonophosphate levels in resting muscle of patients with stablechronic obstructive pulmonary disease. Am J Respir Crit Care Med1998;157:453–457.

43. Sala E, Roca J, Marrades RM, Alonso J, Gonzalez De Suso JM, MorenoA, Barbera JA, Nadal J, de Jover L, Rodriguez-Roisin R, et al. Effectsof endurance training on skeletal muscle bioenergetics in chronicobstructive pulmonary disease. Am J Respir Crit Care Med 1999;159:1726–1734.

44. Green HJ, Bombardier E, Duhamel TA, Stewart RD, Tupling AR,Ouyang J. Metabolic, enzymatic, and transporter responses inhuman muscle during three consecutive days of exercise andrecovery. Am J Physiol Regul Integr Comp Physiol 2008;295:R1238–R1250.

45. Jakobsson P, Jorfeldt L, Henriksson J. Metabolic enzyme activity in thequadriceps femoris muscle in patients with severe chronicobstructive pulmonary disease. Am J Respir Crit Care Med 1995;151:374–377.

46. Maltais F, LeBlanc P, Whittom F, Simard C, Marquis K, Belanger M,Breton MJ, Jobin J. Oxidative enzyme activities of the vastus lateralismuscle and the functional status in patients with COPD. Thorax2000;55:848–853.

47. Ribera F, N’Guessan B, Zoll J, Fortin D, Serrurier B, Mettauer B, BigardX, Ventura-Clapier R, Lampert E. Mitochondrial electron transportchain function is enhanced in inspiratory muscles of patients withchronic obstructive pulmonary disease. Am J Respir Crit Care Med2003;167:873–879.

48. Allaire J, Maltais F, LeBlanc P, Simard PM, Whittom F, Doyon JF,Simard C, Jobin J. Lipofuscin accumulation in the vastus lateralismuscle in patients with chronic obstructive pulmonary disease.Muscle Nerve 2002;25:383–389.

49. Barreiro E, Gea J, Corominas JM, Hussain SN. Nitric oxide synthasesand protein oxidation in the quadriceps femoris of patients withchronic obstructive pulmonary disease. Am J Respir Cell Mol Biol2003;29:771–778.

50. Barreiro E, de la Puente B, Minguella J, Corominas JM, Serrano S,Hussain SN, Gea J. Oxidative stress and respiratory muscledysfunction in severe chronic obstructive pulmonary disease. AmJ Respir Crit Care Med 2005;171:1116–1124.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1131

Page 12: Upper Limb Muscle Dysfunction Executive Summary

51. Barreiro E, Gea J, Matar G, Hussain SN. Expression and carbonylationof creatine kinase in the quadriceps femoris muscles of patients withchronic obstructive pulmonary disease. Am J Respir Cell Mol Biol2005;33:636–642.

52. Barreiro E, Schols AM, Polkey MI, Galdiz JB, Gosker HR, Swallow EB,Coronell C, Gea J; ENIGMA in COPD project. Cytokine profile inquadriceps muscles of patients with severe COPD. Thorax 2008;63:100–107.

53. Barreiro E, Rabinovich R, Marin-Corral J, Barbera JA, Gea J, Roca J.Chronic endurance exercise induces quadriceps nitrosative stress inpatients with severe COPD. Thorax 2009;64:13–19.

54. Couillard A, Maltais F, Saey D, Debigare R, Michaud A, Koechlin C,LeBlanc P, Prefaut C. Exercise-induced quadriceps oxidative stressand peripheral muscle dysfunction in patients with chronicobstructive pulmonary disease. Am J Respir Crit Care Med 2003;167:1664–1669.

55. Engelen MP, Schols AM, Does JD, Deutz NE, Wouters EF. Alteredglutamate metabolism is associated with reduced muscleglutathione levels in patients with emphysema. Am J Respir Crit CareMed 2000;161:98–103.

56. Heunks LM, Viña J, van Herwaarden CL, Folgering HT, Gimeno A,Dekhuijzen PN. Xanthine oxidase is involved in exercise-inducedoxidative stress in chronic obstructive pulmonary disease. AmJ Physiol 1999;277:R1697–R1704.

57. Koechlin C, Couillard A, Simar D, Cristol JP, Bellet H, Hayot M, PrefautC. Does oxidative stress alter quadriceps endurance in chronicobstructive pulmonary disease? Am J Respir Crit Care Med 2004;169:1022–1027.

58. Koechlin C, Couillard A, Cristol JP, Chanez P, Hayot M, Le Gallais D,Prefaut C. Does systemic inflammation trigger local exercise-induced oxidative stress in COPD? Eur Respir J 2004;23:538–544.

59. Koechlin C, Maltais F, Saey D, Michaud A, LeBlanc P, Hayot M, PrefautC. Hypoxaemia enhances peripheral muscle oxidative stress in chronicobstructive pulmonary disease. Thorax 2005;60:834–841.

60. Marin-Corral J, Minguella J, Ramırez-Sarmiento AL, Hussain SN, Gea J,Barreiro E. Oxidised proteins and superoxide anion production in thediaphragm of severe COPD patients. Eur Respir J 2009;33:1309–1319.

61. Rabinovich RA, Ardite E, Troosters T, Carbo N, Alonso J, Gonzalez deSuso JM, Vilaro J, Barbera JA, Polo MF, Argiles JM, et al. Reducedmuscle redox capacity after endurance training in patients withchronic obstructive pulmonary disease. Am J Respir Crit Care Med2001;164:1114–1118.

62. Rodriguez DA, Kalko S, Puig-Vilanova E, Perez-Olabarrıa M, Falciani F,Gea J, Cascante M, Barreiro E, Roca J. Muscle and blood redoxstatus after exercise training in severe COPD patients. Free RadicBiol Med 2012;52:88–94.

63. Van Helvoort HA, Heijdra YF, Thijs HM, Viña J, Wanten GJ, DekhuijzenPN. Exercise-induced systemic effects in muscle-wasted patientswith COPD. Med Sci Sports Exerc 2006;38:1543–1552.

64. Barreiro E, Ferrer D, Sanchez F, Minguella J, Marin-Corral J, Martinez-Llorens J, Lloreta J, Gea J. Inflammatory cells and apoptosis inrespiratory and limb muscles of patients with COPD. J Appl Physiol(1985) 2011;111:808–817.

65. Rabinovich RA, Bastos R, Ardite E, Llinas L, Orozco-Levi M, Gea J,Vilaro J, Barbera JA, Rodrıguez-Roisin R, Fernandez-Checa JC, et al.Mitochondrial dysfunction in COPD patients with low body massindex. Eur Respir J 2007;29:643–650.

66. Barreiro E, del Puerto-Nevado L, Puig-Vilanova E, Perez-Rial S,Sanchez F, Martınez-Galan L, Rivera S, Gea J, Gonzalez-MangadoN, Peces-Barba G. Cigarette smoke-induced oxidative stress inskeletal muscles of mice. Respir Physiol Neurobiol 2012;182:9–17.

67. Coronell C, Orozco-Levi M, Mendez R, Ramırez-Sarmiento A, GaldizJB, Gea J. Relevance of assessing quadriceps endurance in patientswith COPD. Eur Respir J 2004;24:129–136.

68. Bernard S, LeBlanc P, Whittom F, Carrier G, Jobin J, Belleau R, MaltaisF. Peripheral muscle weakness in patients with chronic obstructivepulmonary disease. Am J Respir Crit Care Med 1998;158:629–634.

69. Clark CJ, Cochrane LM, Mackay E, Paton B. Skeletal muscle strengthand endurance in patients with mild COPD and the effects of weighttraining. Eur Respir J 2000;15:92–97.

70. Gosselink R, Troosters T, Decramer M. Distribution of muscleweakness in patients with stable chronic obstructive pulmonarydisease. J Cardiopulm Rehabil 2000;20:353–360.

71. Debigare R, Cote CH, Hould FS, LeBlanc P, Maltais F. In vitro andin vivo contractile properties of the vastus lateralis muscle in maleswith COPD. Eur Respir J 2003;21:273–278.

72. Mador MJ, Bozkanat E, Kufel TJ. Quadriceps fatigue after cycleexercise in patients with COPD compared with healthy controlsubjects. Chest 2003;123:1104–1111.

73. Allaire J, Maltais F, Doyon JF, Noel M, LeBlanc P, Carrier G, SimardC, Jobin J. Peripheral muscle endurance and the oxidative profileof the quadriceps in patients with COPD. Thorax 2004;59:673–678.

74. Franssen FM, Broekhuizen R, Janssen PP, Wouters EF, Schols AM.Limb muscle dysfunction in COPD: effects of muscle wasting andexercise training. Med Sci Sports Exerc 2005;37:2–9.

75. Man WD, Hopkinson NS, Harraf F, Nikoletou D, Polkey MI, Moxham J.Abdominal muscle and quadriceps strength in chronic obstructivepulmonary disease. Thorax 2005;60:718–722.

76. Vivodtzev I, Flore P, Levy P, Wuyam B. Voluntary activation during kneeextensions in severely deconditioned patients with chronicobstructive pulmonary disease: benefit of endurance training.MuscleNerve 2008;37:27–35.

77. Seymour JM, Ward K, Sidhu PS, Puthucheary Z, Steier J, Jolley CJ,Rafferty G, Polkey MI, Moxham J. Ultrasound measurement of rectusfemoris cross-sectional area and the relationship with quadricepsstrength in COPD. Thorax 2009;64:418–423.

78. Man WD, Soliman MG, Nikoletou D, Harris ML, Rafferty GF, Mustfa N,Polkey MI, Moxham J. Non-volitional assessment of skeletal musclestrength in patients with chronic obstructive pulmonary disease.Thorax 2003;58:665–669.

79. Man WD, Soliman MG, Gearing J, Radford SG, Rafferty GF, Gray BJ,Polkey MI, Moxham J. Symptoms and quadriceps fatigability afterwalking and cycling in chronic obstructive pulmonary disease. Am JRespir Crit Care Med 2003;168:562–567.

80. Gagnon P, Maltais F, Bouyer L, Ribeiro F, Coats V, Brouillard C, NoelM, Rousseau-Gagnon M, Saey D. Distal leg muscle function inpatients with COPD. COPD 2013;10:235–242.

81. van den Borst B, Koster A, Yu B, Gosker HR, Meibohm B, BauerDC, Kritchevsky SB, Liu Y, Newman AB, Harris TB, et al. Is age-related decline in lean mass and physical function acceleratedby obstructive lung disease or smoking? Thorax 2011;66:961–969.

82. Heijdra YF, Pinto-Plata V, Frants R, Rassulo J, Kenney L, Celli BR.Muscle strength and exercise kinetics in COPD patients witha normal fat-free mass index are comparable to control subjects.Chest 2003;124:75–82.

83. Vilaro J, Rabinovich R, Gonzalez-deSuso JM, Troosters T, Rodrıguez D,Barbera JA, Roca J. Clinical assessment of peripheral musclefunction in patients with chronic obstructive pulmonary disease. AmJ Phys Med Rehabil 2009;88:39–46.

84. Malaguti C, Napolis LM, Villaça D, Neder JA, Nery LE, Dal Corso S.Relationship between peripheral muscle structure and functionin patients with chronic obstructive pulmonary disease withdifferent nutritional status. J Strength Cond Res 2011;25:1795–1803.

85. Decramer M, Lacquet LM, Fagard R, Rogiers P. Corticosteroidscontribute to muscle weakness in chronic airflow obstruction. Am JRespir Crit Care Med 1994;150:11–16.

86. Couillard A, Koechlin C, Cristol JP, Varray A, Prefaut C. Evidence oflocal exercise-induced systemic oxidative stress in chronicobstructive pulmonary disease patients. Eur Respir J 2002;20:1123–1129.

87. Janaudis-Ferreira T, Wadell K, Sundelin G, Lindstrom B. Thigh musclestrength and endurance in patients with COPD compared withhealthy controls. Respir Med 2006;100:1451–1457.

88. Serres I, Gautier V, Varray A, Prefaut C. Impaired skeletal muscleendurance related to physical inactivity and altered lung function inCOPD patients. Chest 1998;113:900–905.

89. Van’t Hul A, Harlaar J, Gosselink R, Hollander P, Postmus P, KwakkelG. Quadriceps muscle endurance in patients with chronic obstructivepulmonary disease. Muscle Nerve 2004;29:267–274.

AMERICAN THORACIC SOCIETY DOCUMENTS

1132 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 13: Upper Limb Muscle Dysfunction Executive Summary

90. Swallow EB, Gosker HR, Ward KA, Moore AJ, Dayer MJ, HopkinsonNS, Schols AM, Moxham J, Polkey MI. A novel technique fornonvolitional assessment of quadriceps muscle endurance inhumans. J Appl Physiol (1985) 2007;103:739–746.

91. Newell SZ, McKenzie DK, Gandevia SC. Inspiratory and skeletal musclestrength and endurance and diaphragmatic activation in patientswith chronic airflow limitation. Thorax 1989;44:903–912.

92. Luo YM, Hart N, Mustfa N, Lyall RA, Polkey MI, Moxham J. Effect ofdiaphragm fatigue on neural respiratory drive. J Appl Physiol (1985)2001;90:1691–1699.

93. Mador MJ, Kufel TJ, Pineda LA, Steinwald A, Aggarwal A, Upadhyay AM,Khan MA. Effect of pulmonary rehabilitation on quadriceps fatiguabilityduring exercise. Am J Respir Crit Care Med 2001;163:930–935.

94. Marquis N, Debigare R, Bouyer L, Saey D, Laviolette L, Brouillard C, MaltaisF. Physiology of walking in patients with moderate to severe chronicobstructive pulmonary disease.Med Sci Sports Exerc 2009;41:1540–1548.

95. Saey D, Michaud A, Couillard A, Cote CH, Mador MJ, LeBlanc P, JobinJ, Maltais F. Contractile fatigue, muscle morphometry, and bloodlactate in chronic obstructive pulmonary disease. Am J Respir CritCare Med 2005;171:1109–1115.

96. Maltais F, Simard AA, Simard C, Jobin J, Desgagnes P, LeBlanc P.Oxidative capacity of the skeletal muscle and lactic acid kineticsduring exercise in normal subjects and in patients with COPD. Am JRespir Crit Care Med 1996;153:288–293.

97. Calvert LD, Singh SJ, Greenhaff PL, Morgan MD, Steiner MC. Theplasma ammonia response to cycle exercise in COPD. Eur Respir J2008;31:751–758.

98. Dempsey JA, Sheel AW, St Croix CM, Morgan BJ. Respiratoryinfluences on sympathetic vasomotor outflow in humans. RespirPhysiol Neurobiol 2002;130:3–20.

99. Amann M, Regan MS, Kobitary M, Eldridge MW, Boutellier U, PegelowDF, Dempsey JA. Impact of pulmonary system limitations onlocomotor muscle fatigue in patients with COPD. Am J Physiol RegulIntegr Comp Physiol 2010;299:R314–R324.

100. Pitta F, Troosters T, Probst VS, Spruit MA, Decramer M, Gosselink R.Physical activity and hospitalization for exacerbation of COPD.Chest 2006;129:536–544.

101. Crul T, Spruit MA, Gayan-Ramirez G, Quarck R, Gosselink R,Troosters T, Pitta F, Decramer M. Markers of inflammation anddisuse in vastus lateralis of chronic obstructive pulmonary diseasepatients. Eur J Clin Invest 2007;37:897–904.

102. Vilaro J, Ramirez-Sarmiento A, Martınez-Llorens JM, Mendoza T, AlvarezM, Sanchez-Cayado N, Vega A, Gimeno E, Coronell C, Gea J, et al.Global muscle dysfunction as a risk factor of readmission to hospitaldue to COPD exacerbations. Respir Med 2010;104:1896–1902.

103. Crul T, Testelmans D, Spruit MA, Troosters T, Gosselink R, GeeraertsI, Decramer M, Gayan-Ramirez G. Gene expression profiling invastus lateralis muscle during an acute exacerbation of COPD. CellPhysiol Biochem 2010;25:491–500.

104. Vermeeren MA, Schols AM, Wouters EF. Effects of an acuteexacerbation on nutritional and metabolic profile of patients withCOPD. Eur Respir J 1997;10:2264–2269.

105. Vermeeren MA, Wouters EF, Geraerts-Keeris AJ, Schols AM. Nutritionalsupport in patients with chronic obstructive pulmonary diseaseduring hospitalization for an acute exacerbation; a randomizedcontrolled feasibility trial. Clin Nutr 2004;23:1184–1192.

106. Donaldson GC, Wilkinson TM, Hurst JR, Perera WR, Wedzicha JA.Exacerbations and time spent outdoors in chronic obstructivepulmonary disease. Am J Respir Crit Care Med 2005;171:446–452.

107. Mostert R, Goris A, Weling-Scheepers C, Wouters EF, Schols AM.Tissue depletion and health related quality of life in patients withchronic obstructive pulmonary disease. Respir Med 2000;94:859–867.

108. Decramer M, Gosselink R, Troosters T, Verschueren M, Evers G.Muscle weakness is related to utilization of health care resources inCOPD patients. Eur Respir J 1997;10:417–423.

109. Gosselink R, Troosters T, Decramer M. Peripheral muscle weaknesscontributes to exercise limitation in COPD. Am J Respir Crit CareMed 1996;153:976–980.

110. Hamilton AL, Killian KJ, Summers E, Jones NL. Muscle strength,symptom intensity, and exercise capacity in patients withcardiorespiratory disorders. Am J Respir Crit Care Med 1995;152:2021–2031.

111. Killian KJ, Leblanc P, Martin DH, Summers E, Jones NL, Campbell EJ.Exercise capacity and ventilatory, circulatory, and symptomlimitation in patients with chronic airflow limitation. Am Rev RespirDis 1992;146:935–940.

112. Deschenes D, Pepin V, Saey D, LeBlanc P, Maltais F. Locus ofsymptom limitation and exercise response to bronchodilation inchronic obstructive pulmonary disease. J Cardiopulm Rehabil Prev2008;28:208–214.

113. Casaburi R, Patessio A, Ioli F, Zanaboni S, Donner CF, Wasserman K.Reductions in exercise lactic acidosis and ventilation as a result ofexercise training in patients with obstructive lung disease. Am RevRespir Dis 1991;143:9–18.

114. Maltais F, Bernard S, Jobin J, Belleau R, LeBlanc P. Lactate kineticsduring exercise in chronic obstructive pulmonary disease. CanRespir J 1997;4:251–257.

115. Polkey MI, Moxham J. Attacking the disease spiral in chronicobstructive pulmonary disease. Clin Med 2006;6:190–196.

116. Couillard A, Prefaut C. From muscle disuse to myopathy in COPD:potential contribution of oxidative stress. Eur Respir J 2005;26:703–719.

117. Polkey MI, Rabe KF. Chicken or egg: physical activity in COPDrevisited. Eur Respir J 2009;33:227–229.

118. Polkey MI, Moxham J. Attacking the disease spiral in chronicobstructive pulmonary disease: an update. Clin Med 2011;11:461–464.

119. Wagner PD. Skeletal muscles in chronic obstructive pulmonary disease:deconditioning, or myopathy? Respirology 2006;11:681–686.

120. Booth FW, Gollnick PD. Effects of disuse on the structure and functionof skeletal muscle. Med Sci Sports Exerc 1983;15:415–420.

121. Larsson L, Ansved T. Effects of long-term physical training anddetraining on enzyme histochemical and functional skeletal musclecharacteristic in man. Muscle Nerve 1985;8:714–722.

122. Coyle EF, Martin WH III, Bloomfield SA, Lowry OH, Holloszy JO.Effects of detraining on responses to submaximal exercise. J ApplPhysiol (1985) 1985;59:853–859.

123. Casaburi R. Deconditioning. In: Fishman AP, editor. Pulmonaryrehabilitation. New York: Marcel Dekker; 1996. pp. 213–230.

124. Brønstad E, Rognmo O, Tjonna AE, Dedichen HH, Kirkeby-Garstad I,Haberg AK, Bjørk Ingul C, Wisløff U, Steinshamn S. High-intensityknee extensor training restores skeletal muscle function in COPDpatients. Eur Respir J 2012;40:1130–1136.

125. Man WD, Kemp P, Moxham J, Polkey MI. Exercise and muscledysfunction in COPD: implications for pulmonary rehabilitation. ClinSci (Lond) 2009;117:281–291.

126. Troosters T, Gosselink R, Decramer M. Short- and long-term effects ofoutpatient rehabilitation in patients with chronic obstructivepulmonary disease: a randomized trial. Am JMed 2000;109:207–212.

127. Bossenbroek L, de Greef MH, Wempe JB, Krijnen WP, Ten HackenNH. Daily physical activity in patients with chronic obstructivepulmonary disease: a systematic review. COPD 2011;8:306–319.

128. van den Borst B, Slot IG, Hellwig VA, Vosse BA, Kelders MC, BarreiroE, Schols AM, Gosker HR. Loss of quadriceps muscle oxidativephenotype and decreased endurance in patients with mild-to-moderate COPD. J Appl Physiol 2013;114:1319–1328.

129. Natanek SA, Gosker HR, Slot IG, Marsh GS, Hopkinson NS, Man WD,Tal-Singer R, Moxham J, Kemp PR, Schols AM, et al. Heterogeneityof quadriceps muscle phenotype in chronic obstructive pulmonarydisease (COPD); implications for stratified medicine? Muscle Nerve2013;48:488–497.

130. Gouzi F, Prefaut C, Abdellaoui A, Vuillemin A, Molinari N, Ninot G,Caris G, Hayot M. Evidence of an early physical activity reduction inchronic obstructive pulmonary disease patients. Arch Phys MedRehabil 2011;92:1611–1617, e2.

131. Gosker HR, Schrauwen P, Broekhuizen R, Hesselink MK, Moonen-Kornips E, Ward KA, Franssen FM, Wouters EF, Schols AM.Exercise training restores uncoupling protein-3 content in limbmuscles of patients with chronic obstructive pulmonary disease.Am J Physiol Endocrinol Metab 2006;290:E976–E981.

132. Lewis MI, Fournier M, Storer TW, Bhasin S, Porszasz J, Ren SG, Da X,Casaburi R. Skeletal muscle adaptations to testosterone andresistance training in men with COPD. J Appl Physiol (1985) 2007;103:1299–1310.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1133

Page 14: Upper Limb Muscle Dysfunction Executive Summary

133. Howald H, Hoppeler H, Claassen H, Mathieu O, Straub R. Influences ofendurance training on the ultrastructural composition of the differentmuscle fiber types in humans. Pflugers Arch 1985;403:369–376.

134. Russell AP, Feilchenfeldt J, Schreiber S, Praz M, Crettenand A,Gobelet C, Meier CA, Bell DR, Kralli A, Giacobino JP, et al.Endurance training in humans leads to fiber type-specific increasesin levels of peroxisome proliferator-activated receptor-gammacoactivator-1 and peroxisome proliferator-activated receptor-alphain skeletal muscle. Diabetes 2003;52:2874–2881.

135. Simoneau JA, Lortie G, Boulay MR, Marcotte M, Thibault MC,Bouchard C. Human skeletal muscle fiber type alteration with high-intensity intermittent training. Eur J Appl Physiol Occup Physiol1985;54:250–253.

136. Cai D, Frantz JD, Tawa NE Jr, Melendez PA, Oh BC, Lidov HG,Hasselgren PO, Frontera WR, Lee J, Glass DJ, et al. IKKbeta/NF-kappaB activation causes severe muscle wasting in mice. Cell2004;119:285–298.

137. Siu PM. Muscle apoptotic response to denervation, disuse, and aging.Med Sci Sports Exerc 2009;41:1876–1886.

138. Harris J. Autophagy and cytokines. Cytokine 2011;56:140–144.139. Spate U, Schulze PC. Proinflammatory cytokines and skeletal muscle.

Curr Opin Clin Nutr Metab Care 2004;7:265–269.140. Glass DJ. Skeletal muscle hypertrophy and atrophy signaling

pathways. Int J Biochem Cell Biol 2005;37:1974–1984.141. Rabinovich RA, Figueras M, Ardite E, Carbo N, Troosters T, Filella X,

Barbera JA, Fernandez-Checa JC, Argiles JM, Roca J. Increasedtumour necrosis factor-alpha plasma levels during moderate-intensity exercise in COPD patients. Eur Respir J 2003;21:789–794.

142. Agustı A, Morla M, Sauleda J, Saus C, Busquets X. NF-kappaBactivation and iNOS upregulation in skeletal muscle of patients withCOPD and low body weight. Thorax 2004;59:483–487.

143. Montes de Oca M, Torres SH, De Sanctis J, Mata A, Hernandez N,Talamo C. Skeletal muscle inflammation and nitric oxide in patientswith COPD. Eur Respir J 2005;26:390–397.

144. Debigare R, Maltais F, Cote CH, Michaud A, Caron MA, Mofarrahi M,Leblanc P, Hussain SN. Profiling of mRNA expression in quadricepsof patients with COPD and muscle wasting. COPD 2008;5:75–84.

145. Karadag F, Karul AB, Cildag O, Yilmaz M, Ozcan H. Biomarkers ofsystemic inflammation in stable and exacerbation phases of COPD.Lung 2008;186:403–409.

146. Stanojkovic I, Kotur-Stevuljevic J, Milenkovic B, Spasic S, Vujic T,Stefanovic A, Llic A, Ivanisevic J. Pulmonary function, oxidativestress and inflammatory markers in severe COPD exacerbation.Respir Med 2011;105:S31–S37.

147. Moylan JS, Reid MB. Oxidative stress, chronic disease, and musclewasting. Muscle Nerve 2007;35:411–429.

148. Li YP, Chen Y, Li AS, Reid MB. Hydrogen peroxide stimulatesubiquitin-conjugating activity and expression of genes for specificE2 and E3 proteins in skeletal muscle myotubes. Am J Physiol CellPhysiol 2003;285:C806–C812.

149. Li YP, Schwartz RJ, Waddell ID, Holloway BR, Reid MB. Skeletalmuscle myocytes undergo protein loss and reactive oxygen-mediated NF-kappaB activation in response to tumor necrosisfactor alpha. FASEB J 1998;12:871–880.

150. Buck M, Chojkier M. Muscle wasting and dedifferentiation induced byoxidative stress in a murine model of cachexia is prevented byinhibitors of nitric oxide synthesis and antioxidants. EMBO J 1996;15:1753–1765.

151. Furukawa-Hibi Y, Kobayashi Y, Chen C, Motoyama N. FOXOtranscription factors in cell-cycle regulation and the response tooxidative stress. Antioxid Redox Signal 2005;7:752–760.

152. Li YP, Chen Y, John J, Moylan J, Jin B, Mann DL, Reid MB. TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitinligase atrogin1/MAFbx in skeletal muscle. FASEB J 2005;19:362–370.

153. Magalhães J, Ascensão A, Soares JM, Ferreira R, Neuparth MJ,Oliveira J, Amado F, Marques F, Duarte JA. Acute and chronicexposition of mice to severe hypoxia: the role of acclimatization againstskeletal muscle oxidative stress. Int J Sports Med 2005;26:102–109.

154. Hoppeler H, Kleinert E, Schlegel C, Claassen H, Howald H, Kayar SR,Cerretelli P. Morphological adaptations of human skeletal muscle tochronic hypoxia. Int J Sports Med 1990;11:S3–S9.

155. Takabatake N, Nakamura H, Abe S, Inoue S, Hino T, Saito H, Yuki H,Kato S, Tomoike H. The relationship between chronic hypoxemiaand activation of the tumor necrosis factor-alpha system in patientswith chronic obstructive pulmonary disease. Am J Respir Crit CareMed 2000;161:1179–1184.

156. Pitsiou G, Kyriazis G, Hatzizisi O, Argyropoulou P, Mavrofridis E,Patakas D. Tumor necrosis factor-alpha serum levels, weight lossand tissue oxygenation in chronic obstructive pulmonary disease.Respir Med 2002;96:594–598.

157. Turan N, Kalko S, Stincone A, Clarke K, Sabah A, Howlett K, CurnowSJ, Rodriguez DA, Cascante M, O’Neill L, et al. A systems biologyapproach identifies molecular networks defining skeletal muscleabnormalities in chronic obstructive pulmonary disease. PLOSComput Biol 2011;7:e1002129.

158. Caron MA, Theriault ME, Pare ME, Maltais F, Debigare R. Hypoxiaalters contractile protein homeostasis in L6 myotubes. FEBS Lett2009;583:1528–1534.

159. Gonzalez NC, Wood JG. Alveolar hypoxia-induced systemicinflammation: what low PO(2) does and does not do. Adv Exp MedBiol 2010;662:27–32.

160. Chandel NS, McClintock DS, Feliciano CE, Wood TM, Melendez JA,Rodriguez AM, Schumacker PT. Reactive oxygen speciesgenerated at mitochondrial complex III stabilize hypoxia-induciblefactor-1alpha during hypoxia: a mechanism of O2 sensing. J BiolChem 2000;275:25130–25138.

161. de Theije C, Costes F, Langen RC, Pison C, Gosker HR. Hypoxiaand muscle maintenance regulation: implications for chronicrespiratory disease. Curr Opin Clin Nutr Metab Care 2011;14:548–553.

162. Fiaccadori E, Del Canale S, Vitali P, Coffrini E, Ronda N, Guariglia A.Skeletal muscle energetics, acid-base equilibrium and lactatemetabolism in patients with severe hypercapnia and hypoxemia.Chest 1987;92:883–887.

163. England BK, Chastain JL, Mitch WE. Abnormalities in proteinsynthesis and degradation induced by extracellular pH in BC3H1myocytes. Am J Physiol 1991;260:C277–C282.

164. Bailey JL, Wang X, England BK, Price SR, Ding X, Mitch WE. Theacidosis of chronic renal failure activates muscle proteolysis in ratsby augmenting transcription of genes encoding proteins of theATP-dependent ubiquitin-proteasome pathway. J Clin Invest 1996;97:1447–1453.

165. Franch HA, Raissi S, Wang X, Zheng B, Bailey JL, Price SR. Acidosisimpairs insulin receptor substrate-1-associated phosphoinositide3-kinase signaling in muscle cells: consequences on proteolysis.Am J Physiol Renal Physiol 2004;287:F700–F706.

166. Van Vliet M, Spruit MA, Verleden G, Kasran A, Van Herck E, PittaF, Bouillon R, Decramer M. Hypogonadism, quadricepsweakness, and exercise intolerance in chronic obstructivepulmonary disease. Am J Respir Crit Care Med 2005;172:1105–1111.

167. Kamischke A, Kemper DE, Castel MA, Luthke M, Rolf C, Behre HM,Magnussen H, Nieschlag E. Testosterone levels in men with chronicobstructive pulmonary disease with or without glucocorticoidtherapy. Eur Respir J 1998;11:41–45.

168. Karadag F, Ozcan H, Karul AB, Yilmaz M, Cildag O. Sex hormonealterations and systemic inflammation in chronic obstructivepulmonary disease. Int J Clin Pract 2009;63:275–281.

169. Laghi F, Langbein WE, Antonescu-Turcu A, Jubran A, Bammert C,Tobin MJ. Respiratory and skeletal muscles in hypogonadal menwith chronic obstructive pulmonary disease. Am J Respir Crit CareMed 2005;171:598–605.

170. Debigare R, Marquis K, Cote CH, Tremblay RR, Michaud A, LeBlancP, Maltais F. Catabolic/anabolic balance and muscle wasting inpatients with COPD. Chest 2003;124:83–89.

171. Lee SJ. Regulation of muscle mass by myostatin. Annu Rev Cell DevBiol 2004;20:61–86.

172. Elliott B, Renshaw D, Getting S, Mackenzie R. The central role ofmyostatin in skeletal muscle and whole body homeostasis. ActaPhysiol (Oxf) 2012;205:324–340.

173. Ju CR, Chen RC. Serum myostatin levels and skeletal muscle wastingin chronic obstructive pulmonary disease. Respir Med 2012;106:102–108.

AMERICAN THORACIC SOCIETY DOCUMENTS

1134 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 15: Upper Limb Muscle Dysfunction Executive Summary

174. Hayot M, Rodriguez J, Vernus B, Carnac G, Jean E, Allen D, Goret L,Obert P, Candau R, Bonnieu A. Myostatin up-regulation isassociated with the skeletal muscle response to hypoxic stimuli.Mol Cell Endocrinol 2011;332:38–47.

175. Remels AH, Gosker HR, Langen RC, Schols AM. The mechanisms ofcachexia underlying muscle dysfunction in COPD. J Appl Physiol(1985) 2013;114:1253–1262.

176. Hopkinson NS, Man WD, Dayer MJ, Ross ET, Nickol AH, Hart N,Moxham J, Polkey MI. Acute effect of oral steroids on musclefunction in chronic obstructive pulmonary disease. Eur Respir J2004;24:137–142.

177. Decramer M, de Bock V, Dom R. Functional and histologic picture ofsteroid-induced myopathy in chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 1996;153:1958–1964.

178. Schakman O, Gilson H, Kalista S, Thissen JP. Mechanisms of muscleatrophy induced by glucocorticoids. Horm Res 2009;72:36–41.

179. Janssens W, Bouillon R, Claes B, Carremans C, Lehouck A,Buysschaert I, Coolen J, Mathieu C, Decramer M, Lambrechts D.Vitamin D deficiency is highly prevalent in COPD and correlates withvariants in the vitamin D-binding gene. Thorax 2010;65:215–220.

180. Janssens W, Lehouck A, Carremans C, Bouillon R, Mathieu C,Decramer M. Vitamin D beyond bones in chronic obstructivepulmonary disease: time to act. Am J Respir Crit Care Med 2009;179:630–636.

181. Vogelmeier CF, Wouters EF. Treating the systemic effects of chronicobstructive pulmonary disease. Proc Am Thorac Soc 2011;8:376–379.

182. Jackson AS, Shrikrishna D, Kelly JL, Hart N, Moxham J, Polkey MI,Kemp P, Hopkinson NS. Vitamin D and skeletal muscle strengthand endurance in COPD. Eur Respir J 2013;41:309–316.

183. Hornikx M, Van Remoortel H, Lehouck A, Mathieu C, Maes K, Gayan-Ramirez G, Decramer M, Troosters T, Janssens W. Vitamin Dsupplementation during rehabilitation in COPD: a secondaryanalysis of a randomized trial. Respir Res 2012;13:84.

184. Hopkinson NS, Li KW, Kehoe A, Humphries SE, Roughton M,Moxham J, Montgomery H, Polkey MI. Vitamin D receptorgenotypes influence quadriceps strength in chronic obstructivepulmonary disease. Am J Clin Nutr 2008;87:385–390.

185. De Paepe B, Brusselle GG, Maes T, Creus KK, D’hose S, D’Haese N,Bracke KR, D’hulst AI, Joos GF, De Bleecker JL. TNF alphareceptor genotype influences smoking-induced muscle-fibre-typeshift and atrophy in mice. Acta Neuropathol 2008;115:675–681.

186. Gosker HR, Langen RC, Bracke KR, Joos GF, Brusselle GG, Steele C,Ward KA, Wouters EF, Schols AM. Extrapulmonary manifestationsof chronic obstructive pulmonary disease in a mouse model ofchronic cigarette smoke exposure. Am J Respir Cell Mol Biol 2009;40:710–716.

187. Tang K, Wagner PD, Breen EC. TNF-alpha-mediated reduction inPGC-1alpha may impair skeletal muscle function after cigarettesmoke exposure. J Cell Physiol 2010;222:320–327.

188. Rinaldi M, Maes K, De Vleeschauwer S, Thomas D, Verbeken EK,Decramer M, Janssens W, Gayan-Ramirez GN. Long-term nose-onlycigarette smoke exposure induces emphysema and mild skeletalmuscle dysfunction in mice. Dis Model Mech 2012;5:333–341.

189. Montes de Oca M, Loeb E, Torres SH, De Sanctis J, Hernandez N,Talamo C. Peripheral muscle alterations in non-COPD smokers.Chest 2008;133:13–18.

190. Orlander J, Kiessling KH, Larsson L. Skeletal muscle metabolism,morphology and function in sedentary smokers and nonsmokers.Acta Physiol Scand 1979;107:39–46.

191. Larsson L, Orlander J. Skeletal muscle morphology, metabolism andfunction in smokers and non-smokers. A study on smoking-discordantmonozygous twins. Acta Physiol Scand 1984;120:343–352.

192. Steiner MC, Barton RL, Singh SJ, Morgan MD. Bedside methodsversus dual energy X-ray absorptiometry for body compositionmeasurement in COPD. Eur Respir J 2002;19:626–631.

193. Robles PG, Mathur S, Janaudis-Fereira T, Dolmage TE, Goldstein RS,Brooks D. Measurement of peripheral muscle strength in individualswith chronic obstructive pulmonary disease: a systematic review. JCardiopulm Rehabil Prev 2011;31:11–24.

194. Stone CA, Nolan B, Lawlor PG, Kenny RA. Hand-held dynamometry:tester strength is paramount, even in frail populations. J RehabilMed 2011;43:808–811.

195. Kaelin ME, Swank AM, Adams KJ, Barnard KL, Berning JM, Green A.Cardiopulmonary responses, muscle soreness, and injury duringthe one repetition maximum assessment in pulmonary rehabilitationpatients. J Cardiopulm Rehabil 1999;19:366–372.

196. Casaburi R, Bhasin S, Cosentino L, Porszasz J, Somfay A, Lewis MI,Fournier M, Storer TW. Effects of testosterone and resistancetraining in men with chronic obstructive pulmonary disease. Am JRespir Crit Care Med 2004;170:870–878.

197. Mador MJ, Bozkanat E, Aggarwal A, Shaffer M, Kufel TJ. Enduranceand strength training in patients with COPD. Chest 2004;125:2036–2045.

198. Mathur S, Makrides L, Hernandez P. Test-retest reliability of isometricand isokinetic torque in patients with chronic obstructive pulmonarydisease. Physiother Can 2004;56:94–101.

199. Maffiuletti NA, Bizzini M, Desbrosses K, Babault N, Munzinger U.Reliability of knee extension and flexion measurements using theCon-Trex isokinetic dynamometer. Clin Physiol Funct Imaging2007;27:346–353.

200. Spruit MA, Gosselink R, Troosters T, De Paepe K, Decramer M.Resistance versus endurance training in patients with COPD andperipheral muscle weakness. Eur Respir J 2002;19:1072–1078.

201. Bernard S, Whittom F, Leblanc P, Jobin J, Belleau R, Berube C,Carrier G, Maltais F. Aerobic and strength training in patients withchronic obstructive pulmonary disease. Am J Respir Crit Care Med1999;159:896–901.

202. O’Donnell DE, McGuire M, Samis L, Webb KA. General exercisetraining improves ventilatory and peripheral muscle strength andendurance in chronic airflow limitation. Am J Respir Crit Care Med1998;157:1489–1497.

203. Ortega F, Toral J, Cejudo P, Villagomez R, Sanchez H, Castillo J,Montemayor T. Comparison of effects of strength and endurancetraining in patients with chronic obstructive pulmonary disease. AmJ Respir Crit Care Med 2002;166:669–674.

204. Vogiatzis I, Terzis G, Nanas S, Stratakos G, Simoes DC, GeorgiadouO, Zakynthinos S, Roussos C. Skeletal muscle adaptations tointerval training in patients with advanced COPD. Chest 2005;128:3838–3845.

205. Vogiatzis I, Stratakos G, Simoes DC, Terzis G, Georgiadou O,Roussos C, Zakynthinos S. Effects of rehabilitative exercise onperipheral muscle TNFalpha, IL-6, IGF-I and MyoD expression inpatients with COPD. Thorax 2007;62:950–956.

206. Vogiatzis I, Simoes DC, Stratakos G, Kourepini E, Terzis G, Manta P,Athanasopoulos D, Roussos C, Wagner PD, Zakynthinos S. Effectof pulmonary rehabilitation on muscle remodelling in cachecticpatients with COPD. Eur Respir J 2010;36:301–310.

207. O’Shea SD, Taylor NF, Paratz JD. Progressive resistance exerciseimproves muscle strength and may improve elements ofperformance of daily activities for people with COPD: a systematicreview. Chest 2009;136:1269–1283.

208. Gouzi F, Prefaut C, Abdellaoui A, Roudier E, de Rigal P, Molinari N,Laoudj-Chenivesse D, Mercier J, Birot O, Hayot M. Blunted muscleangiogenic training-response in COPD patients versus sedentarycontrols. Eur Respir J 2013;41:806–814.

209. Constantin D, Menon MK, Houchen-Wolloff L, Morgan MD, Singh SJ,Greenhaff P, Steiner MC. Skeletal muscle molecular responses toresistance training and dietary supplementation in COPD. Thorax2013;68:625–633.

210. Radom-Aizik S, Kaminski N, Hayek S, Halkin H, Cooper DM, Ben-DovI. Effects of exercise training on quadriceps muscle geneexpression in chronic obstructive pulmonary disease. J ApplPhysiol (1985) 2007;102:1976–1984.

211. Rabinovich RA, Ardite E, Mayer AM, Polo MF, Vilaro J, Argiles JM,Roca J. Training depletes muscle glutathione in patients withchronic obstructive pulmonary disease and low body mass index.Respiration 2006;73:757–761.

212. Troosters T, Gosselink R, Decramer M. Exercise training in COPD:how to distinguish responders from nonresponders. J CardiopulmRehabil 2001;21:10–17.

213. Bouchard C, Sarzynski MA, Rice TK, Kraus WE, Church TS, Sung YJ,Rao DC, Rankinen T. Genomic predictors of the maximal O2 uptakeresponse to standardized exercise training programs. J ApplPhysiol (1985) 2011;110:1160–1170.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1135

Page 16: Upper Limb Muscle Dysfunction Executive Summary

214. Folland J, Leach B, Little T, Hawker K, Myerson S, Montgomery H,Jones D. Angiotensin-converting enzyme genotype affects theresponse of human skeletal muscle to functional overload. ExpPhysiol 2000;85:575–579.

215. Burtin C, Saey D, Saglam M, Langer D, Gosselink R, Janssens W,Decramer M, Maltais F, Troosters T. Effectiveness of exercisetraining in patients with COPD: the role of muscle fatigue. EurRespir J 2012;40:338–344.

216. Pitta F, Troosters T, Probst VS, Langer D, Decramer M, Gosselink R.Are patients with COPD more active after pulmonary rehabilitation?Chest 2008;134:273–280.

217. Burtin C, Decramer M, Gosselink R, Janssens W, Troosters T.Rehabilitation and acute exacerbations. Eur Respir J 2011;38:702–712.

218. Vivodtzev I, Lacasse Y, Maltais F. Neuromuscular electricalstimulation of the lower limbs in patients with chronic obstructivepulmonary disease. J Cardiopulm Rehabil Prev 2008;28:79–91.

219. Sillen MJ, Speksnijder CM, Eterman RM, Janssen PP, Wagers SS,Wouters EF, Uszko-Lencer NH, Spruit MA. Effects ofneuromuscular electrical stimulation of muscles of ambulation inpatients with chronic heart failure or COPD: a systematic review ofthe English-language literature. Chest 2009;136:44–61.

220. Sillen MJ, Wouters EF, Franssen FM, Meijer K, Stakenborg KH, SpruitMA. Oxygen uptake, ventilation, and symptoms during low-frequency versus high-frequency NMES in COPD: a pilot study.Lung 2011;189:21–26.

221. Neder JA, Sword D, Ward SA, Mackay E, Cochrane LM, Clark CJ.Home based neuromuscular electrical stimulation as a newrehabilitative strategy for severely disabled patients with chronicobstructive pulmonary disease (COPD). Thorax 2002;57:333–337.

222. Abdellaoui A, Prefaut C, Gouzi F, Couillard A, Coisy-Quivy M, HugonG, Molinari N, Lafontaine T, Jonquet O, Laoudj-Chenivesse D, et al.Skeletal muscle effects of electrostimulation after COPDexacerbation: a pilot study. Eur Respir J 2011;38:781–788.

223. Dal Corso S, Napolis L, Malaguti C, Gimenes AC, Albuquerque A,Nogueira CR, De Fuccio MB, Pereira RD, Bulle A, McFarlane N,et al. Skeletal muscle structure and function in response toelectrical stimulation in moderately impaired COPD patients. RespirMed 2007;101:1236–1243.

224. Vivodtzev I, Debigare R, Gagnon P, Mainguy V, Saey D, Dube A, PareME, Belanger M, Maltais F. Functional and muscular effects ofneuromuscular electrical stimulation in patients with severe COPD:a randomized clinical trial. Chest 2012;141:716–725.

225. Efthimiou J, Fleming J, Gomes C, Spiro SG. The effect of supplementaryoral nutrition in poorly nourished patients with chronic obstructivepulmonary disease. Am Rev Respir Dis 1988;137:1075–1082.

226. Weekes CE, Emery PW, Elia M. Dietary counselling and food fortificationin stable COPD: a randomised trial. Thorax 2009;64:326–331.

227. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss isa reversible factor in the prognosis of chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 1998;157:1791–1797.

228. Steiner MC, Barton RL, Singh SJ, Morgan MD. Nutritional enhancementof exercise performance in chronic obstructive pulmonary disease:a randomised controlled trial. Thorax 2003;58:745–751.

229. Creutzberg EC, Wouters EF, Mostert R, Weling-Scheepers CA, ScholsAM. Efficacy of nutritional supplementation therapy in depletedpatients with chronic obstructive pulmonary disease. Nutrition2003;19:120–127.

230. Saudny-Unterberger H, Martin JG, Gray-Donald K. Impact ofnutritional support on functional status during an acuteexacerbation of chronic obstructive pulmonary disease. Am JRespir Crit Care Med 1997;156:794–799.

231. Ferreira IM, Brooks D, White J, Goldstein R. Nutritionalsupplementation for stable chronic obstructive pulmonary disease.Cochrane Database Syst Rev 2012;12:CD000998.

232. Schols AM, Soeters PB, Mostert R, Pluymers RJ, Wouters EF. Physiologiceffects of nutritional support and anabolic steroids in patients withchronic obstructive pulmonary disease. A placebo-controlledrandomized trial. Am J Respir Crit Care Med 1995;152:1268–1274.

233. Ferreira IM, Verreschi IT, Nery LE, Goldstein RS, Zamel N, Brooks D,Jardim JR. The influence of 6 months of oral anabolic steroids onbody mass and respiratory muscles in undernourished COPDpatients. Chest 1998;114:19–28.

234. Casaburi R, Make B, Piquette C, Kramer T, Ries A. Oxandroloneincreases weight and muscle mass in underweight COPD patients[abstract]. Chest 2002;122:73S.

235. Creutzberg EC, Wouters EF, Mostert R, Pluymers RJ, Schols AM. Arole for anabolic steroids in the rehabilitation of patients withCOPD? A double-blind, placebo-controlled, randomized trial. Chest2003;124:1733–1742.

236. Svartberg J, Aasebø U, Hjalmarsen A, Sundsfjord J, Jorde R.Testosterone treatment improves body composition and sexualfunction in men with COPD, in a 6-month randomized controlledtrial. Respir Med 2004;98:906–913.

237. Pape GS, Friedman M, Underwood LE, Clemmons DR. The effect ofgrowth hormone on weight gain and pulmonary function in patientswith chronic obstructive lung disease. Chest 1991;99:1495–1500.

238. Gullett NP, Hebbar G, Ziegler TR. Update on clinical trials of growthfactors and anabolic steroids in cachexia and wasting. Am J ClinNutr 2010;91:1143S–1147S.

239. Scalvini S, Volterrani M, Vitacca M, Clark AL, Solfrini R, Panzali AM,Ferrari R, Levi GF. Plasma hormone levels and haemodynamics inpatients with chronic obstructive lung disease. Monaldi Arch ChestDis 1996;51:380–386.

240. Burdet L, de Muralt B, Schutz Y, Pichard C, Fitting JW. Administrationof growth hormone to underweight patients with chronicobstructive pulmonary disease. A prospective, randomized,controlled study. Am J Respir Crit Care Med 1997;156:1800–1806.

241. Suchner U, Rothkopf MM, Stanislaus G, Elwyn DH, Kvetan V,Askanazi J. Growth hormone and pulmonary disease. Metaboliceffects in patients receiving parenteral nutrition. Arch Intern Med1990;150:1225–1230.

242. Weisberg J, Wanger J, Olson J, Streit B, Fogarty C, Martin T, CasaburiR. Megestrol acetate stimulates weight gain and ventilation inunderweight COPD patients. Chest 2002;121:1070–1078.

243. Fuld JP, Kilduff LP, Neder JA, Pitsiladis Y, Lean ME, Ward SA, CottonMM. Creatine supplementation during pulmonary rehabilitation inchronic obstructive pulmonary disease. Thorax 2005;60:531–537.

244. Faager G, Soderlund K, Skold CM, Rundgren S, Tollback A,Jakobsson P. Creatine supplementation and physical training inpatients with COPD: a double blind, placebo-controlled study. IntJ Chron Obstruct Pulmon Dis 2006;1:445–453.

245. Al-Ghimlas F, Todd DC. Creatine supplementation for patients withCOPD receiving pulmonary rehabilitation: a systematic review andmeta-analysis. Respirology 2010;15:785–795.

246. Deacon SJ, Vincent EE, Greenhaff PL, Fox J, Steiner MC, Singh SJ,Morgan MD. Randomized controlled trial of dietary creatine as anadjunct therapy to physical training in chronic obstructivepulmonary disease. Am J Respir Crit Care Med 2008;178:233–239.

247. Borghi-Silva A, Baldissera V, Sampaio LM, Pires-DiLorenzo VA,Jamami M, Demonte A, Marchini JS, Costa D. L-carnitine as anergogenic aid for patients with chronic obstructive pulmonarydisease submitted to whole-body and respiratory muscle trainingprograms. Braz J Med Biol Res 2006;39:465–474.

248. Ferreira LF, Reid MB. Muscle-derived ROS and thiol regulation inmuscle fatigue. J Appl Physiol (1985) 2008;104:853–860.

249. Barreiro E, Galdiz JB, Mariñan M, Alvarez FJ, Hussain SN, Gea J.Respiratory loading intensity and diaphragm oxidative stress: N-acetyl-cysteine effects. J Appl Physiol (1985) 2006;100:555–563.

250. Laviolette L, Lands LC, Dauletbaev N, Saey D, Milot J, Provencher S,LeBlanc P, Maltais F. Combined effect of dietary supplementationwith pressurized whey and exercise training in chronic obstructivepulmonary disease: a randomized, controlled, double-blind pilotstudy. J Med Food 2010;13:589–598.

251. Lehouck A, Mathieu C, Carremans C, Baeke F, Verhaegen J, VanEldere J, Decallonne B, Bouillon R, Decramer M, Janssens W. Highdoses of vitamin D to reduce exacerbations in chronic obstructivepulmonary disease: a randomized trial. Ann Intern Med 2012;156:105–114.

252. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, IoannidisJP, Clarke M, Devereaux PJ, Kleijnen J, Moher D. The PRISMAstatement for reporting systematic reviews and meta-analyses ofstudies that evaluate health care interventions: explanation andelaboration. J Clin Epidemiol 2009;62:e1–e34.

AMERICAN THORACIC SOCIETY DOCUMENTS

1136 American Journal of Respiratory and Critical Care Medicine Volume 189 Number 9 | May 1 2014

Page 17: Upper Limb Muscle Dysfunction Executive Summary

253. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-CoelloP, Schunemann HJ, Group GW; GRADE Working Group. GRADE:an emerging consensus on rating quality of evidence and strengthof recommendations. BMJ 2008;336:924–926.

254. Semenza GL. Regulation of oxygen homeostasis by hypoxia-induciblefactor 1. Physiology (Bethesda) 2009;24:97–106.

255. Jatta K, Eliason G, Portela-Gomes GM, Grimelius L, Caro O,Nilholm L, Sirjso A, Piehl-Aulin K, Abdel-Halim SM. Overexpressionof von Hippel-Lindau protein in skeletal muscles of patients withchronic obstructive pulmonary disease. J Clin Pathol 2009;62:70–76.

256. Favier FB, Costes F, Defour A, Bonnefoy R, Lefai E, Bauge S,Peinnequin A, Benoit H, Freyssenet D. Downregulation of Akt/mammalian target of rapamycin pathway in skeletal muscle isassociated with increased REDD1 expression in response tochronic hypoxia. Am J Physiol Regul Integr Comp Physiol 2010;298:R1659–R1666.

257. Bhasin S, Storer TW, Berman N, Callegari C, Clevenger B, Phillips J,Bunnell TJ, Tricker R, Shirazi A, Casaburi R. The effects ofsupraphysiologic doses of testosterone on muscle size andstrength in normal men. N Engl J Med 1996;335:1–7.

258. Velloso CP. Regulation of muscle mass by growth hormone and IGF-I.Br J Pharmacol 2008;154:557–568.

259. Goris AH, Vermeeren MA, Wouters EF, Schols AM, Westerterp KR.Energy balance in depleted ambulatory patients with chronicobstructive pulmonary disease: the effect of physical activity andoral nutritional supplementation. Br J Nutr 2003;89:725–731.

260. Boland R. Role of vitamin D in skeletal muscle function. Endocr Rev1986;7:434–448.

261. Gagnon P, Bussieres JS, Ribeiro F, Gagnon SL, Saey D, Gagne N,Provencher S, Maltais F. Influences of spinal anesthesia on exercisetolerance in patients with chronic obstructive pulmonary disease.Am J Respir Crit Care Med 2012;186:606–615.

262. Kutsuzawa T, Shioya S, Kurita D, Haida M, Ohta Y, Yamabayashi H.31P-NMR study of skeletal muscle metabolism in patients withchronic respiratory impairment. Am Rev Respir Dis 1992;146:1019–1024.

263. Remels AH, Schrauwen P, Broekhuizen R, Willems J, Kersten S,Gosker HR, Schols AM. Peroxisome proliferator-activated receptorexpression is reduced in skeletal muscle in COPD. Eur Respir J2007;30:245–252.

264. Picard M, Godin R, Sinnreich M, Baril J, Bourbeau J, Perrault H,Taivassalo T, Burelle Y. The mitochondrial phenotype of peripheralmuscle in chronic obstructive pulmonary disease: disuse ordysfunction? Am J Respir Crit Care Med 2008;178:1040–1047.

265. Lacasse Y, Goldstein R, Lasserson TJ, Martin S. Pulmonaryrehabilitation for chronic obstructive pulmonary disease. CochraneDatabase Syst Rev 2006;4:CD003793.

266. O’Donnell DE, D’Arsigny C, Webb KA. Effects of hyperoxia onventilatory limitation during exercise in advanced chronicobstructive pulmonary disease. Am J Respir Crit Care Med 2001;163:892–898.

267. Maltais F, Simon M, Jobin J, Desmeules M, Sullivan MJ, Belanger M,Leblanc P. Effects of oxygen on lower limb blood flow and O2

uptake during exercise in COPD. Med Sci Sports Exerc 2001;33:916–922.

268. Somfay A, Porszasz J, Lee SM, Casaburi R. Dose-response effect ofoxygen on hyperinflation and exercise endurancein nonhypoxaemic COPD patients. Eur Respir J 2001;18:77–84.

269. Ferreira IM, Brooks D, Lacasse Y, Goldstein RS, White J. Nutritionalsupplementation for stable chronic obstructive pulmonary disease.Cochrane Database Syst Rev 2005;2:CD000998.

270. van Wetering CR, Hoogendoorn M, Broekhuizen R, Geraerts-KeerisGJ, De Munck DR, Rutten-van Molken MP, Schols AM. Efficacy andcosts of nutritional rehabilitation in muscle-wasted patients withchronic obstructive pulmonary disease in a community-basedsetting: a prespecified subgroup analysis of the INTERCOM trial.J Am Med Dir Assoc 2010;11:179–187.

271. Pison CM, Cano NJ, Cherion C, Caron F, Court-Fortune I, AntoniniMT, Gonzalez-Bermejo J, Meziane L, Molano LC, Janssens JP,et al.; IRAD Investigators. Multimodal nutritional rehabilitationimproves clinical outcomes of malnourished patients with chronicrespiratory failure: a randomised controlled trial. Thorax 2011;66:953–960.

AMERICAN THORACIC SOCIETY DOCUMENTS

American Thoracic Society Documents 1137