Anda di halaman 1dari 14

Peripheral Artery Disease Compendium

Circulation Research Compendium on Peripheral Artery Disease


Epidemiology of Peripheral Artery Disease
Pathogenesis of the Limb Manifestations and Exercise Limitations in Peripheral Artery Disease
Lower Extremity Manifestations of Peripheral Artery Disease: The Pathophysiologic and Functional Implications of Leg Ischemia
The Genetic Basis of Peripheral Arterial Disease: Current Knowledge, Challenges and Future Directions
Modulating the Vascular Response to Limb Ischemia: Angiogenic and Cell Therapies
Pharmacological Treatment and Current Management of Peripheral Artery Disease
Endovascular Intervention for Peripheral Artery Disease
Surgical Intervention for Peripheral Arterial Disease
John Cooke, Guest Editor

Pathogenesis of the Limb Manifestations and Exercise


Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

Limitations in Peripheral Artery Disease


William R. Hiatt, Ehrin J. Armstrong, Christopher J. Larson, Eric P. Brass

Abstract: Patients with peripheral artery disease have a marked reduction in exercise performance and daily
ambulatory activity irrespective of their limb symptoms of classic or atypical claudication. This review will evaluate
the multiple pathophysiologic mechanisms underlying the exercise impairment in peripheral artery disease based
on an evaluation of the current literature and research performed by the authors. Peripheral artery disease results
in atherosclerotic obstructions in the major conduit arteries supplying the lower extremities. This arterial disease
process impairs the supply of oxygen and metabolic substrates needed to match the metabolic demand generated
by active skeletal muscle during walking exercise. However, the hemodynamic impairment associated with the
occlusive disease process does not fully account for the reduced exercise impairment, indicating that additional
pathophysiologic mechanisms contribute to the limb manifestations. These mechanisms include a cascade of
pathophysiological responses during exercise-induced ischemia and reperfusion at rest that are associated with
endothelial dysfunction, oxidant stress, inflammation, and muscle metabolic abnormalities that provide opportunities
for targeted therapeutic interventions to address the complex pathophysiology of the exercise impairment in
peripheral artery disease.   (Circ Res. 2015;116:1527-1539. DOI: 10.1161/CIRCRESAHA.116.303566.)
Key Words: exercise ■ metabolism ■ peripheral artery disease ■ peripheral vascular diseases
■ physiopathology ■ therapeutics

P eripheral artery disease (PAD) is initiated by athero-


genic mechanisms common to all major vascular terri-
tories but encompasses several distinctly important clinical
exercise limitation in the majority of patients, whereas the
classic symptoms of intermittent claudication, ischemic
rest pain, and ischemic ulceration are less common mani-
sequelae.1 The global burden of PAD is large and rising, festations.3 Patients with PAD are also at increased risk of
with substantial morbidity and mortality found in high-, myocardial infarction, ischemic stroke, heart failure, re-
middle-, and low-income countries.2 The atherosclerotic novascular hypertension, and vascular death reflecting the
occlusive disease underlying PAD is associated with an systemic atherosclerotic burden.4–6 The epidemiology of

Original received September 18, 2014; revision received December 8, 2014; accepted December 17, 2014. In February 2015, the average time from
submission to first decision for all original research papers submitted to Circulation Research was 13.9 days.
From the Division of Cardiology, Department of Medicine (W.R.H., E.J.A.), CPC Clinical Research (W.R.H.), University of Colorado School of
Medicine, Aurora; Cardiovascular & Metabolic Diseases Drug Discovery Unit, Takeda Pharmaceuticals, San Diego, CA (C.J.L.); and Department of
Medicine, Harbor-UCLA Center for Clinical Pharmacology, Torrance, CA (E.P.B.).
Correspondence to William R. Hiatt, MD, Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, C/O CPC
Clinical Research, 13199 E Montview Blvd, Suite 200, Aurora, CO 80045. E-mail Will.Hiatt@UCDenver.edu
© 2015 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.116.303566

1527
1528  Circulation Research  April 24, 2015

Nonstandard Abbreviations and Acronyms


Limb Symptoms
Patients with PAD may present with a range of limb symp-
ABI ankle-brachial index toms. Most patients are either asymptomatic or have atypical
ERR estrogen related receptor limb symptoms during exercise, whereas only a third have
ETA endothelin receptor A typical symptoms of claudication.18 The clinical character-
ETB endothelin receptor B istics of PAD have been characterized in a longitudinal co-
FFR fractional flow reserve hort study that defined typical and atypical limb symptoms.
PAD peripheral artery disease Typical claudication is defined by exercise-induced calf dis-
PPAR peroxisome proliferator–activated receptor comfort that is relieved by rest.19 Atypical limb symptoms
can be characterized as exertional calf symptoms that do not
begin at rest but are otherwise not consistent with classical
claudication, such as muscle symptoms that do not include
PAD is linked to that of the other common atherosclerotic
the calves, and pain at rest and pain with exertion.18 These
processes, such as coronary and cerebral artery diseases,
symptoms should not be confused with other causes of atypi-
and affects 15% of the adult population aged >45 years.7
cal limb pain, including statin-induced myalgias, spinal ste-
Predominant risk factors for PAD include advancing age,
nosis, and other orthopedic conditions. The pathogenesis can
smoking, and diabetes mellitus; hypertension and lipid
usually be differentiated with a careful history and physical
disorders contribute less risk when compared with their
examination, although some patients may have PAD concom-
association with ischemic stroke and myocardial infarc-
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

itant orthopedic disease.


tion.2,8 Patients with PAD have cardiovascular outcomes
Atypical exertional leg symptoms are more common than
similar to patients with coronary artery disease or isch-
classic claudication and are also associated with decreased
emic stroke, with the risk of myocardial infarction, stroke,
overall functional exercise capacity.20 Patients with hemo-
or vascular death >5% per year.9–15
dynamic evidence of PAD in the peripheral circulation (eg,
The diagnosis of PAD can be ascertained by simple hemo-
low ABI) consistently demonstrate a marked reduction in
dynamic measurements, in particular, the ankle-brachial index
peak exercise performance and daily ambulatory activity.21–23
(ABI), a test performed by measuring the systolic blood pres-
Thus, the pathophysiology, which results in reduced exercise
sure in each arm and the dorsalis pedis and posterior tibial
performance in PAD, affects a broad spectrum of patients ir-
arteries of each ankle. The highest of the dorsalis pedis or
respective of their limb symptoms. In this context, although
posterior tibial artery pressures is the numerator of the ABI
patients with typical intermittent claudication have been the
specific to each leg, whereas the highest arm pressure on ei-
best studied and the term is in common usage, the mecha-
ther side is the denominator. A ratio of <0.90 in either leg is
nisms underlying the exercise limitation are likely common
considered hemodynamic evidence of PAD.16 When imaged,
to all patients regardless of the nature of their limb symptoms.
PAD can be characterized as one or more arterial stenoses or
Therefore, using the term claudication to describe all symp-
occlusions involving the distal aorta, iliac, femoral, popliteal,
tomatic patients with PAD is inappropriate, and in this review,
or tibial arteries.17
the functional deficit associated with PAD will be referred to
The underlying mechanisms responsible for the functional
as an exercise limitation and includes otherwise asymptomatic
limitations in PAD are hemodynamic in origin, which affects
patients who also have exercise impairment.
the supply of oxygen and substrates to metabolically active
tissue: skeletal muscle in the case of intermittent claudication Severity of the Exercise Impairment
and skin and subcutaneous tissues in the case of critical leg The severity of the functional limitation in PAD has been
ischemia. However, additional pathophysiologic mechanisms well characterized. On a population basis, patients with PAD
contribute to the limb manifestations, where a reduction in have an ≈50% reduction in peak exercise performance when
blood flow alone does not fully account for the exercise limita- compared with an age-matched healthy cohort.21 This exer-
tion. Recent research into the pathophysiology of this exercise cise limitation is associated with marked impairments in daily
limitation has identified several important sequelae resultant physical activity as assessed by questionnaire and daily ener-
from the chronic hemodynamic deficit. These changes affect- gy expenditure.24,25 Patients with PAD also have an accelerated
ing the vasculature and skeletal muscle distal to the primary functional decline over time that is related to the underlying
hemodynamic lesions likely contribute to the mechanisms of hemodynamic severity of the disease in the leg.26 The primary
exercise limitation in PAD. These advances are reviewed here treatment goal for all patients with PAD is cardiovascular risk
with an additional focus on identifying possible future thera- reduction, whereas on the basis of the above considerations,
peutic strategies. an additional goal is to improve exercise performance, daily
functional activity, and quality of life.
Symptomatic Manifestations and Exercise
Limitations in PAD Arterial Hemodynamics in PAD
In the coronary circulation, plaque rupture leading to acute The reduction in blood flow to the lower extremity as a conse-
ischemic events is a common clinical presentation; in PAD, quence of the arterial occlusive disease process is a contribu-
progressive atherosclerosis leading to chronic stenosis and tor to the exercise impairment in PAD. Ischemia with exercise
occlusion, often in series, in the arteries supplying the lower followed by reperfusion is also an important initiator of the
extremities dominates the symptomatic manifestations.6 subsequent pathophysiology in skeletal muscle.
Hiatt et al   Exercise Limitation in Peripheral Artery Disease   1529

Resting Hemodynamics indicating that under the conditions of low muscle metabolic
Volumetric blood flow in the major conduit arteries delivering demand at rest, flow can be maintained by a corresponding
oxygen to the lower extremities is determined by the driving decrease in peripheral resistance to compensate for the drop in
pressure, geometry of the vessel and associated stenosis, and arterial pressure across the stenosis.31
blood viscosity.27 Poiseuille first described these fundamental
Exercise Hemodynamics
relationships by showing that flow was inversely proportional
In normal subjects during exercise, the increase in limb oxy-
to the length of a tube, directly proportional to the pressure
gen uptake is driven by an increase in metabolic demand.33 In
gradient, and directly proportional to the fourth power of the
healthy adults, the system is dynamic in the rest-to-exercise
tube diameter.28 Under normal conditions, flow is laminar and
transition, with blood flow increasing 10-fold at low levels of
there is minimal pressure drop from the heart to the distal ar- exercise and 40-fold at high levels.34 At steady-state exercise,
terial circulation. However, with an arterial stenosis, there is this leads to a close coupling of ventilatory oxygen update in
a drop in pressure and flow across the stenosis. This pressure the lungs, delivery of oxygen by the heart and arterial circula-
drop is accentuated by a loss of kinetic energy because of tur- tion to active skeletal muscle, and muscle mitochondrial oxy-
bulent flow induced by the stenosis (Figure 1). In the lower gen uptake at the tissue level.35 In patients with PAD, there is a
extremity, a series of arterial stenoses, for example, a stenosis markedly blunted flow response in the rest-to-exercise transi-
or occlusion in the external iliac, superficial femoral arteries, tion that plateaus at flows well below that obtained in healthy
and popliteal arteries, will be hemodynamically additive with subjects. This flow limitation is the result of the fixed stenoses
the overall flow limitation of the sum of the individual compo- becoming flow limiting (a concept termed critical arterial ste-
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

nents.29,30 These hemodynamic manifestations of PAD can be nosis). Furthermore, the vasodilator response in the peripheral
assessed with the ABI, which reflects the total hemodynamic circulation is also abnormal and is unable to fully compensate
burden of stenoses and occlusions from the central circulation for the fixed resistance lesions.36,37
to the ankle. Except for patients with noncompressible tibial A related approach to understanding the change in arterial
vessels where the ABI does not reflect the intra-arterial pres- hemodynamics with exercise is the fractional flow reserve
sure, patients with PAD will typically have an ABI <0.90 at (FFR), which is the ratio of the flow distal to a stenosis di-
rest. However, these patients have no limb symptoms at rest in vided by the pressure proximal to the stenosis under condi-
the absence of more severe hemodynamic compromise caus- tions of maximal vasodilation. For example, an FFR of 0.80
ing critical leg ischemia. Under conditions of rest, skeletal indicates that the stenotic vessel can only deliver 80% of the
muscle oxygen demand is low and limb oxygen delivery is ad- flow of a normal vessel when the distal arteriolar bed is maxi-
equate despite the drop in systolic pressure from the arm to the mally vasodilated. The vasodilator response in PAD can also
ankle. Resting calf muscle blood flow in patients with claudi- be impaired by endothelial dysfunction, as discussed below. In
cation is similar to that in an age-matched control population, humans with coronary artery disease, these relationships have

Figure 1. Hemodynamic alterations in peripheral artery disease. Top, A healthy arterial bed with normal large vessel and
microcirculatory flow characteristics. Bottom, A peripheral artery with atherosclerotic occlusive disease. ABI indicates ankle-brachial
index. Reprinted from Hiatt and Brass.32
1530  Circulation Research  April 24, 2015

been confirmed to have physiological significance for quanti- impaired vasodilation in patients with PAD is likely a sequela
fying coronary ischemia.38 In the coronary circulation, clinical of the obstruction, and improvement in physiological vasodi-
studies have shown that an FFR of <0.80 is physiologically lation with exercise offers a potential therapeutic target.
significant and that this FFR value identifies a clinically sig- Microcirculatory dysfunction and local skeletal muscle
nificant cutoff point where symptomatic patients benefit from flow can also be assessed by novel noninvasive imaging tech-
coronary revascularization.39 In patients with claudication, niques, including arterial spin–labeling MRI and contrast-en-
initial studies have shown that FFR measurements correlate hanced ultrasound. In a pilot study, arterial spin–labeling MRI
with the lesion peak systolic velocity, a noninvasive measure- demonstrated that microvascular flow was relatively preserved
ment of lesion severity.40 Further studies will be necessary to among patients with mild PAD (ABI>0.50), but more severe
correlate the clinical use of FFR measurements to potentially PAD (ABI<0.50) was associated with a significant decrement
guide revascularization or other treatment strategies in PAD. in microvascular blood flow.55 This technique can also be used
A complementary approach to determining the physiologi- to image calf muscle flow at peak exercise and may be a useful
cal significance of flow limitations in patients with PAD is tool to assess changes in calf blood flow after therapeutic in-
the assessment of tissue oxygenation. Blood-oxygen-level– terventions.56 In another small study, perfusion imaging of the
dependent MRI imaging techniques can measure the extent skeletal muscle microcirculation was performed by contrast-
of tissue deoxygenation during postocclusive reactive hyper- enhanced ultrasound at rest and with exercise. In a multivari-
emia.41 Near-infrared spectroscopy has also been used to make ate model, the treadmill time to the onset of claudication was
dynamic assessments of tissue oxygenation in patients with correlated with microcirculatory exercise flow and the differ-
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

PAD. For example, the phosphodiestersase-5 inhibitor silde- ence between resting and exercise microcirculatory flow.57
nafil improved muscle oxygenation as assessed by near-infra- Contrast-enhanced MRI measures of peak exercise blood flow
red spectroscopy without improving walking performance in a have also been shown to correlate with exercise performance
small study of patients with PAD.42 These approaches have the and 6-minute walk times.58
potential to differentiate the effect of oxygen delivery versus
oxygen use on muscle dysfunction. Rheological Factors
Alterations in blood hemorheology can also impair arte-
Endothelial and Microcirculatory Dysfunction rial flow. Changes in the blood concentration of fibrinogen
As described above, optimizing muscle blood flow during ex- and von Willebrand factor can increase blood viscosity, par-
ercise is dependent on vasodilation in the limb to minimize ticularly at the microcirculatory level, which according to
the overall resistance to flow delivery to active muscle. This Pousseille law could contribute to reduced arterial flow—but
vasodilation is an active process with several important vaso- less so than a change of equal magnitude in the percent steno-
active mediators, including nitric oxide.43 Patients with PAD sis.59,60 Activated leukocytes may also promote microvascular
have significant abnormalities in endothelium-dependent va- plugging and thrombosis, thereby further increasing the re-
sodilation.44,45 Oxidant stress leads to generation of superox- sistance to arterial flow. In a large cohort of patients from the
ide anion and other mediators of endothelial dysfunction.10,46 Edinburgh Artery Study, blood viscosity and fibrinogen were
Exercise-induced release of endothelin-1, a potent vasocon- independently associated with hemodynamic disease severity
strictor, can also antagonize exercise-induced skeletal muscle of PAD, and fibrin degradation products were also associated
vasodilation.47,48 Through these mechanisms, impaired endo- with progression of PAD.61,62 These findings suggest that in-
thelial function as assessed by flow-mediated dilation has been creased blood viscosity may be a component of the functional
correlated with the clinical severity of PAD.49,50 Furthermore, limitation or simply secondary to the systemic atherosclerotic
in studies where leg blood flow was measured by using a disease process.
thermodilution catheter, there was a correlation (r=0.71) be-
tween maximal leg blood flow and performance on a bicycle Existing Medical Therapy for the Exercise
ergometer.31 Limitation in PAD
Interestingly, endothelial dysfunction was recently associat- Current medical therapy for the exercise limitation in PAD is
ed with walking impairment independent of the ABI, suggest- limited. Reasonable evidence exists for the use of phospho-
ing that endothelial dysfunction may contribute to the exercise diesterase type 3 inhibition, particularly cilostazol. Several
impairment in PAD.49 Other studies have suggested a correla- meta-analyses have demonstrated a consistent clinical benefit
tion between ABI and endothelial dysfunction as assessed by of this medication despite a lack of detailed understanding of
flow-mediated dilation.51 Exercise training, which improves its mechanism of disease benefit.63 Specifically, it is not clear
functional status in patients with PAD, also improves flow- why either the inhibition of platelet aggregation or the stimu-
mediated dilation.52 A recent study of patients undergoing a lation of vasodilation—both mediated by phosphodiesterase
12-week exercise training program demonstrated an improve- type 3 inhibition—would translate into functional benefit in
ment in flow-mediated dilation compared with baseline, sug- patients with PAD. As noted above, a nonspecific vasodilator,
gesting that improvements in endothelial function may be one such as cilostazol, has the potential for steal of flow by health-
mechanism mediating reduced symptoms of claudication after ier, more responsive vessels. Also, no convincing explanation
exercise training.53 In addition, in an animal model of chronic exists for why the clinical benefits of phosphodiesterase type
atherosclerosis that simulated some features of PAD, a phar- 3 inhibition continue to increase during 24 weeks of treat-
macological intervention that improved exercise-induced ment when the direct pharmacological effects—vasodilation
blood flow also improved the animals’ functional status.54 The and inhibition of platelet aggregation—are immediate.64 This
Hiatt et al   Exercise Limitation in Peripheral Artery Disease   1531

time-dependent improvement suggests a complex physiologi- indicates that factors other than changes in limb vascular re-
cal response to treatment. Although modest improvements in sistance contribute to the exercise impairment in patients with
ABI have been observed with phosphodiesterase inhibition, it PAD. Underlying mechanisms that may induce factors that
is unlikely that this effect alone accounts for the mechanism of are linked to the functional impairment include inflammation
benefit.65 The efficacy signals from 2 additional phosphodies- and oxidant stress with subsequent endothelial and microcir-
terase type 3 inhibitors support a disease benefit for the class, culatory dysfunction, skeletal muscle structural abnormalities,
albeit one that is not completely understood.66,67 altered oxygen coupling and mitochondrial respiration, and
Older medications, such as pentoxifylline and naftidrofuryl, skeletal muscle metabolic abnormalities. A better understand-
are approved for use in patients with PAD within and outside ing of these pathways may help guide novel medical therapies
of the United States, respectively. The overall data supporting for the treatment of PAD and improve functional outcomes in
any functional benefit for treatment of patients with PAD with patients with claudication.
pentoxifylline are weak.68,69 The limited benefit of naftidro-
furyl in patients with PAD also cannot be linked to any well- Endothelial and Microcirculatory Dysfunction
defined mechanism of action.69–71 As described above, patients with PAD have significant ab-
normalities in endothelium-dependent vasodilation.44,45 A
Exercise training has been a mainstay of treatment for PAD,
potential therapeutic strategy with respect to endothelial and
with a well-established benefit during a typical 12-week train-
microcirculatory dysfunction is antagonism of the release or
ing program.72 Exercise training may affect several pathways
action of endothelin. Endothelin can bind to either endothelin
associated with clinical benefit, including improved skeletal
receptor A (ETA) or endothelin receptor B (ETB) subtypes
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

muscle metabolism, endothelial function, and the biomechan-


1 and 2. ETA receptors are located on the smooth muscle
ics of gait.73,74 Given the established efficacy of exercise train-
cells of vascular tissue, and endothelin binding causes vaso-
ing, it would be logical to use pharmacotherapy as an adjunct
constriction through agonist-induced receptor-mediated sig-
to structured exercise rehabilitation in patients with PAD.
naling.80 ETB receptor action is more complex, with ETB1
Candidates for pharmacotherapy might include drugs with
mediating vasodilation through nitric oxide release, whereas
mechanisms that augment or complement the beneficial ef-
ETB2 causes vasoconstriction. Thus, antagonism of vasocon-
fects of training. In part to due to lack of good candidates and
striction mediated by ETA represents a potential therapeutic
the complexity of studying drug therapy against a background
strategy to improve microcirculatory flow in PAD. However,
of exercise training, there is a paucity of data addressing this
the resultant vasodilator effect will be of benefit only if it im-
hypothesis. A randomized trial of exercise training added to
proves microvascular flow to ischemic muscle and does not
optimal medical therapy, including cilostazol, demonstrated
induce a steal phenomenon. Several selective ETA receptor
a clear additive benefit of training, but the design did not
antagonists are available for clinical testing. Ambrisentan is
address if cilostazol added benefit compared with exercise
an ETA selective antagonist approved in the United States and
training without the drug.75 In addition, a study combining
Europe for the treatment of pulmonary arterial hypertension.81
treatment with propionyl-l-carnitine and exercise training
Two other candidates for testing would be the ETA and ETB
failed to show significantly better improvement in walking
dual antagonists macitentan and bosentan, which are both also
performance when compared with exercise training alone.76
approved for the treatment of pulmonary artery hypertension.
Endovascular revascularization is a commonly used inter-
Macitentan represents an intermediate pharmacological pro-
vention to improve skeletal muscle blood flow. When provided
file between bosentan and ambrisentan, with a 50-fold selec-
on a background of best medical therapy, revascularization of
tivity for the ETA subtype compared with the ETB subtype.82,83
aortoiliac disease results in improved peak walking time and
Given the high prevalence of diabetes mellitus in patients
associated parameters of quality of life, and the magnitude of
with PAD, it is interesting to note that studies have also sug-
benefit is similar to that achieved with supervised exercise.75
gested that pioglitazone could improve vascular function.84–86
The relevance of these changes to functional status in patients
Cellular and Molecular Pathophysiology of the with PAD remains unknown.
Exercise Impairment in PAD
Although the primary pathophysiology of the exercise limita- Inflammation and Oxidant Stress
tion in PAD is initiated by the underlying arterial occlusive Inflammation is a key mediator of the atherosclerotic process
disease and associated reduction in exercise blood flow and and likely contributes to the limb manifestations of PAD as
oxygen delivery, the correlation is not strong between the flow well.9,10 In addition, the mismatch between oxygen demand
limitation versus the observed decrease in exercise perfor- and oxygen delivery during exercise in patients with PAD
mance. Studies examining the resting ABI showed a weak to induces an inflammatory response.10 In PAD, exercise is as-
no correlation with peak treadmill exercise performance.22,77,78 sociated with an increase in plasma levels of numerous in-
In addition, interventions, such as exercise training, that lead flammatory mediators, including thiobarbituric acid–reactive
to a large increase in peak exercise performance have little ef- substances (formed as a byproduct of lipid peroxidation),
fect on skeletal muscle blood flow or ABI.79 Thus, although the thromboxane, interleukin-8, tumor necrosis factor-α, soluble
occlusive atherosclerotic disease process is associated with a intercellular adhesion molecule-1, vascular cell adhesion mol-
well-defined limitation of blood flow and oxygen delivery to ecule-1, von Willebrand factor, E-selectin, and thrombomodu-
exercising skeletal muscle, the absence of a strong correla- lin.87–93 Inflammatory mediators can aggravate endothelial
tion between the arterial obstruction and exercise performance dysfunction, and markers, such as interleukin-6, are inversely
1532  Circulation Research  April 24, 2015

correlated with maximum treadmill performance.94 When ex- antibody, has completed safety, tolerability, pharmacokinetic,
ercise impairment was evaluated by a series of walking tests, and pharmacodynamic studies showing reduction in myo-
higher serum C-reactive protein and serum amyloid A concen- cardial damage after percutaneous coronary intervention for
trations were associated with reductions in 6-minute-walk dis- non–ST-segment–elevation myocardial infarction.113 Whether
tance, 4-meter walking velocity, and a summary performance modulating P-selectin in PAD has longer term clinical benefit
score that combined performance in walking speed, stand- has not yet been studied. In addition, inclacumab has been
ing balance, and time for 5 repeated chair rises.95 Elevated demonstrated to reduce elevated circulating platelet–leuko-
C-reactive protein levels were also associated with a greater cyte aggregate levels in patients with PAD, thus providing
annual decline in 6-minute walk performance for a period of 3 pharmacodynamic data useful for hypothesis generation.114
years and peak walking time.96,97 The angiotensin-converting enzyme inhibitor ramipril re-
During ischemia, skeletal muscle mitochondria release free cently has been reported to improve placebo-corrected walk-
radicals, including superoxide and other reactive oxygen spe- ing distance.115 However, the placebo group in this study
cies, that are derived from the oxidation–reduction cascade.98 did not show the usual walking performance improvements
Reperfusion of ischemic muscle after exercise may also lead typically seen in these trials, which may have inflated the
to an increase in oxidant stress.99,100 These reactive oxygen net benefit of the drug over placebo. Potential mechanisms
species have the potential to trigger numerous pathophysi- of ramipril-mediated improvement in walking performance
ologic pathways, including endothelial dysfunction and cova- include associations with an increase in angiogenic biomark-
lent modification of muscle macromolecules. Over time, this ers and reduction in the markers of thrombosis, inflammation,
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

oxidant stress can also lead to mitochondrial DNA injury as and leukocyte adhesion.116 These biomarker changes may re-
demonstrated in patients with PAD.101,102 Mitochondrial DNA flect modulation of more fundamental processes. As a meta-
injury can also be detected in the less affected limbs of pa- analysis of angiotensin-converting inhibition did not find any
tients with unilateral PAD, suggesting that the mitochondrial benefit on exercise performance recently,117 it seems unlikely
injury may reflect systemic inflammation and not simply local that renin–angiotensin–aldosterone system pathway interven-
skeletal muscle ischemia. Although exercise acutely induces tion provides consistent functional benefit.
oxidant stress in patients with PAD, exercise training has con- Metabolic agents, such as propionyl-l-carnitine, have also
sistently been shown to improve symptoms among patients shown a signal of benefit but have not been fully developed
with PAD. Although this may seem to be contradictory, it is as an approved pharmacological therapy.118 Although several
important to differentiate the effects of acute versus training large studies have demonstrated the atherosclerotic disease–
exercise paradigms.103 For example, in model systems, exercise modifying benefit of statin treatment, which may include
training has been shown to upregulate modulators of oxidative actions through anti-inflammatory pathways, inconsistent
stress, including superoxide dismutase, inducible nitric oxide benefits on exercise performance have been observed in
synthase, and thioredoxin.104–106 This modulation of oxidative PAD.119–121
stress defense mechanisms may be an important mechanism Skeletal Muscle Structural Abnormalities
for the benefit of exercise training in patients with PAD. Numerous pathological changes have been identified in the
Inflammatory mediators may also have proangiogenic and skeletal muscle of patients with PAD, including muscle apop-
antiangiogenic effects, potentially modulating the endogenous tosis and atrophy, increased fiber type switching, altered
response to ischemia.107 For example, circulating concentra- myosin heavy-chain expression, and muscle fiber denerva-
tions of vascular endothelial growth factor are decreased in tion.122–125 These structural changes may be mediated, in part,
patients with PAD when compared with controls with other by higher levels of inflammatory mediators in PAD.126 On the
comorbid conditions.93 basis of biopsy specimens from the gastrocnemius muscles of
Innovative anti-inflammatory therapies developed for other patients with PAD, ≈4% of gastrocnemius cells are apoptotic,
indications may have relevance for PAD. For example, inter- and caspase-3 levels are twice as high as in patients without
cellular adhesion molecules and vascular cell adhesion mol- PAD.125 Selective fiber type switching from type I (aerobic)
ecules play important roles in intracellular signaling and the to type II (glycolytic) fibers may impair skeletal muscle per-
immune and inflammatory responses and are elevated in pa- formance and has been associated with decreased exercise
tients with PAD.108 Antagonists of vascular cell adhesion mol- tolerance.123 Decreased expression of myosin heavy-chain iso-
ecule and intercellular adhesion molecule function have been form II in patients with PAD may also result in altered muscle
developed that could be tested for efficacy in PAD. In particu- contraction kinetics and decreased cellular efficiency.127 The
lar, antagonists of very late antigen-4 have shown promise in observation of muscle fiber denervation among patients with
treating inflammatory disorders in several animal models and PAD has also led to the hypothesis that arterial insufficiency
could be advanced into clinical trials for this indication.109–112 coexists with a distal motor neuron neuropathy that worsens
Selectins, also elevated in PAD populations, are a set of muscle weakness independent of arterial flow.128 Consistent
cell adhesion molecules consisting of 3 subsets: E-selectins with this hypothesis, impaired peroneal nerve conduction
expressed on endothelial cells in skin microvessels after ex- velocity was associated with decreased calf muscle area and
posure to cytokines, L-selectins basally expressed on leuko- lower 6-minute walk distance among patients with PAD.129
cytes, such as granulocytes and monocytes, and P-selectins Numerous studies have correlated changes in calf muscle
basally expressed on platelets and endothelial cells. Recently, ultrastructure and overall muscle strength with subsequent car-
inclacumab, a potent and selective P-selectin–neutralizing diovascular outcomes in patients with claudication. Computed
Hiatt et al   Exercise Limitation in Peripheral Artery Disease   1533

tomographic measurements of the content of muscle and fat PAD.141 Studies have also been performed using 31P magnetic
in the calf have shown that calf muscle content is inversely resonance spectroscopy in PAD. When assessed at the onset
associated with loss of mobility, even after adjusting for other of exercise, changes in muscle pH and phosphocreatine con-
risk factors and comorbidities.130 During long-term follow-up, centration suggested inefficient oxidative metabolism neces-
a reduced calf muscle content was associated with increased sitating a higher rate of ATP turnover for given power output
all-cause and cardiovascular mortality independent of the as observed in control muscle.142 Taken together, these obser-
ABI.131 Overall leg strength has also been shown to associate vations demonstrate increased metabolic inertia in patients
with mortality in men, but not women, with PAD.132 Taken to- with PAD and provide evidence of impaired muscle metabolic
gether, these findings suggest that the morphological changes function in PAD.
in calf muscle predispose to significant functional decline and Other evidence supports impaired mitochondrial function
may be an additional marker for increased risk among patients as an important factor in functional impairment in PAD. The
with symptomatic PAD. electron transport chain in mitochondria oxidizes nicotinamide
One approach for modulating muscle apoptosis and atrophy adenine dinucleotide in a series of enzymatic steps that trans-
is through exploitation of the myostatin pathway. Multiple fer electrons to oxygen (Figure 2). Key mitochondrial path-
new pharmacological agents that target this pathway are al- ways, including the electron transport chain, are vulnerable to
ready in clinical development, including the soluble ActRIIB- free-radical injury.143 Skeletal muscle from the affected leg in
Fc fusion decoy receptor ramatercept,133,134 antimyostatin patients with PAD has reduced the activities of mitochondrial
antibody MYO-029, and the ActRIIB antibody BYM338.135–137 nicotinamide adenine dinucleotide dehydrogenase of complex
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

However, previous studies have shown that although endur- I and ubiquinol-cytochrome c oxidoreductase (complex III).144
ance training improves function in patients with PAD, strength The impairment in complex III of the electron transport chain
training does not result in the same degree of improvement. may result in a higher redox potential to maintain electron
This discordance between the strong predictive value of en- flux, which may in turn lead to free-radical leakage in the mi-
dogenous muscle mass and the poor response to interven- tochondria. Catalase and superoxide dismutase minimize the
tion may mean that endogenous muscle mass is an integrated deleterious effects of hydrogen peroxide and superoxide, re-
marker of the complex limb changes occurring with chronic spectively, by catalyzing the conversion of these species into
PAD. Thus, the likelihood of success of a muscle mass–based oxygen and hydrogen peroxide, which is subsequently con-
strategy alone may be low.74 Furthermore, this strategy is like- verted to water. Damage ensues when reactive oxygen species
ly to be associated with systemic adverse events as exempli- levels exceed the reductive capacity of the cell.145 These ob-
fied by the epistaxis, gum bleeding, and skin vessel dilation servations suggest that electron transport chain activity is im-
observed in patients treated with ramatercept.133 paired, likely due to the ischemia-reperfusion injury in PAD,
which may contribute to propagation of oxidative injury and
Alterations in Oxygen Coupling and Mitochondrial metabolic dysfunction. Future strategies to modulate mito-
Respiration chondrial respiratory function and efficiency could represent
There is a tight coupling between pulmonary oxygen uptake, novel therapeutic targets for patients with PAD.
circulatory delivery of oxygen, and muscle mitochondrial
respiration in healthy subjects during steady-state exercise. Skeletal Muscle Metabolic Abnormalities
The transition from rest-to-exercise causes a rapid increase Numerous alterations in metabolic pathways have been stud-
in the cellular degradation of ATP to ADP to support mus- ied in the skeletal muscle of patients with PAD. A commonly
cle contraction. Regeneration of ATP is initially supported used experimental paradigm has been used to identify patients
by nonoxidative mechanisms, including a rapid depletion of with predominant evidence of hemodynamic abnormalities in
phosphocreatine. Oxidative resynthesis of ATP occurs more one leg with no ABI abnormality or claudication symptoms in
slowly and requires delivery of oxygen to exercising skeletal the contralateral leg. This allowed comparisons between the
muscle. The delay in oxidative adjustment to a new metabolic affected leg, an apparently unaffected leg in the same patient,
demand has been termed metabolic inertia.138 and an age-matched control population. These studies have
The transition to increased oxygen consumption can be identified many key metabolic abnormalities in PAD.
characterized by several noninvasive measurements, including Early observations focused on alterations in muscle car-
the kinetics of pulmonary oxygen consumption at the onset of nitine metabolism. Under normal metabolic conditions, fuel
exercise and the rate of hemoglobin desaturation in exercising substrates, such as fatty acids, proteins, and carbohydrates,
skeletal muscle. In healthy subjects at the onset of exercise, are converted into acyl-CoAs as intermediates in their com-
these kinetic assessments are an attempt to characterize rates plete oxidation. These coenzyme A–coupled intermediates
of mitochondrial oxidative metabolism rather than overall are linked to the cellular carnitine pool through the reversible
oxygen delivery.138 In patients with PAD, a profound prolon- transfer of acyl groups between carnitine and coenzyme A.146
gation of the kinetic rates of pulmonary oxygen consumption One of the functions of carnitine is to serve as a buffer for the
and tissue hemoglobin desaturation have been described at acyl-CoA pool through the formation of acylcarnitines. Under
the onset of exercise relative to healthy, age-matched con- conditions of metabolic stress, incomplete oxidation or incom-
trols.139,140 This prolonged kinetic response associated with plete use of acyl-CoAs leads to their accumulation. Transfer
the reduced exercise performance in subjects with PAD sug- of the acyl group to carnitine results in the accumulation of the
gests that alterations in skeletal muscle mitochondrial respira- corresponding acylcarnitine, which can be measured.147 Thus,
tion may contribute to the decreased exercise performance in an increase in the concentration of acylcarnitines in plasma or
1534  Circulation Research  April 24, 2015

Figure 2. Metabolic and mitochondrial


abnormalities in peripheral artery
disease. FAD indicates flavin adenine
dinucleotide; and NAD, nicotine adenine
dinucleotide. Reprinted from Hiatt and
Brass.32
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

muscle in PAD is consistent with abnormal, incomplete oxida- pool from acylcarnitine to carnitine, indicating an improve-
tive metabolism of fuel substrates. ment in complete substrate oxidation.73
In patients with PAD, initial observations demonstrated ac- Peroxisome proliferator–activated receptor (PPAR)-α and
cumulation of short-chain acylcarnitines in plasma that was PPAR-δ both modulate mitochondrial expression, and their
inversely correlated with exercise performance.148 Subsequent levels correlate well with type I fiber levels and endurance
studies evaluated changes in muscle carnitine metabolism us- training and capacity.153,154 Although agonists of PPAR-δ, such
ing gastrocnemius biopsy specimens. These studies confirmed as L165041 and GW501516, have not advanced substantially
the accumulation of acylcarnitines in the affected skeletal through clinical development because of concerns of neoplas-
muscle with no evidence of accumulation in the unaffected tic risk,155 the PPAR-α agonist fibrates have been marketed
muscle.149 Acylcarnitine accumulation was also associated for many years. Fibrates could be used to test the therapeutic
with reduced peak treadmill exercise performance and was hypothesis that a PPAR-α agonist in patients with PAD would
a stronger predictor of exercise performance than the ABI.149 improve skeletal muscle in type I oxidative capacity and that
Muscle lactate levels are also significantly increased in the this enrichment will translate into improved walking perfor-
skeletal muscle of patients with PAD as a result of incomplete mance in PAD.
oxidation of glucose. Lactate accumulation likely reflects high Several molecular interventions that modulate a subset of
nicotinamide adenine dinucleotide content (because of im- targets for mitochondrial biogenesis beyond the PPAR family
paired entry into the electron transport chain) and decreased are in preclinical development. These include the sirtuins and
pyruvate dehydrogenase activity.150 5′ AMP-activated protein kinase pathway,156 estrogen related
Pharmacological approaches to restoring mitochondrial receptor (ERR)-α,157 estrogen related receptor (ERR)-γ,158
function have been reviewed recently.151 These approaches and nuclear respiratory factor 1.159 Although the use of the 5′
could include modifying mitochondrial biogenesis, mito- AMP-activated protein kinase activator 5-aminoimidazole-
chondrial dynamics, mitophagy, or the mitochondrial unfold- 4-carboxamide ribonucleotide for doping in cycling and other
ed protein response. Many therapeutic hypotheses focused professional sports is suspect, clinical trial results assessing
on mitochondrial function can be formulated and tested in its effect on performance have been generally disappoint-
PAD based on the established efficacy of exercise training, ing.160 However, lack of benefit in healthy subjects may not
as training likely affects mitochondrial function in PAD. exclude effectiveness in patients with PAD. The indirect 5′
Mitochondria from subjects with PAD who undergo exercise AMP-activated protein kinase activator R118 has improved
training demonstrate improved oxidation of pyruvate,152 and exercise performance and vascular insufficiency in high-fat
exercise training results in a redistribution of the carnitine fed mice.54 Although the overlapping metabolic functions of
Hiatt et al   Exercise Limitation in Peripheral Artery Disease   1535

Table 1.  Mechanisms of Exercise Impairment in PAD underlying the reduced exercise performance in these patients
Healthy Physiology PAD Pathophysiology
and potential new targets for therapeutic interventions.
Arterial flow Normal at rest, inadequate increment
with exercise to meet metabolic
Disclosures
demand Dr Hiatt reports grant awards in the past 2 years to CPC Clinical
Research (a nonprofit academic research organization and affiliated
Endothelial and microvasculature Impaired endothelium-dependent to the University of Colorado) from the following sponsors: Aastrom,
dysfunction vasodilation on exercise challenge AstraZeneca, Bayer, National Institutes of Health, CSI, Cytokinetics,
Inflammation Increase in plasma levels of numerous DNAVEC, Kowa, Kyushu University, Merck, Pluristem, Regeneron,
inflammatory mediators; inflammation- Rigel, and Takeda. Dr Armstrong reports being a consultant in the
impaired actions of progenitor/satellite past 2 years to Pfizer, Abbott Vascular, and Spectranetics. Dr Larson
cell differentiation and responses and is an employee of Takeda Pharmaceuticals Company, Inc. Dr Brass
growth factors reports being a consultant in the past 2 years to GlaxoSmithKline,
Novartis, McNeil Consumer Pharmaceuticals, Novo Nordisk,
Reactive oxygen species and During ischemia, skeletal muscle
3D Communications, Catabasis Pharmaceuticals, Allergan,
oxidative/reductive stress mitochondria release free radicals,
NovaDigm Therapeutics, Bayer, Endo Pharmaceuticals, Amgen,
including superoxide and other reactive Boston Scientific, World Self-Medication Institute, Merck, NPS
oxygen species that are derived from Pharmaceuticals, HeartWare International, Takeda Pharmaceuticals,
the oxidation–reduction cascade Genzyme, Consumer Healthcare Products Association, DepoMed,
Muscle structural abnormalities Muscle apoptosis and atrophy; fiber Cangene, Aveo Oncology, BioMarin, Galderma, QRx Pharma,
type switching; altered myosin heavy University of Washington, Amarin, EnteroMedics, Acerta, Cerexa,
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

chain expression; fiber denervation Kythera Biopharmaceuticals, and Trius Therapeutics. Dr Brass has
Muscle metabolic abnormalities Altered oxygen coupling and equity in Calistoga Pharmaceuticals and Catabasis Pharmaceuticals.
mitochondrial respiration: in patients
None of the authors received financial support for this article.
with PAD, prolongation of the kinetic
rates of oxygen consumption, and References
tissue hemoglobin desaturation has 1. Libby P. Mechanisms of acute coronary syndromes and their implica-
been described at the onset of exercise tions for therapy. N Engl J Med. 2013;368:2004–2013. doi: 10.1056/
NEJMra1216063.
PAD indicates peripheral artery disease. 2. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott
MM, Norman PE, Sampson UK, Williams LJ, Mensah GA, Criqui MH.
ERRα and ERRγ suggest potential benefit for patients with Comparison of global estimates of prevalence and risk factors for periph-
PAD, breast cancer association studies suggest that ERRα eral artery disease in 2000 and 2010: a systematic review and analysis.
Lancet. 2013;382:1329–1340. doi: 10.1016/S0140-6736(13)61249-0.
and ERRγ have differing effects on cancer progression, with 3. McDermott MM, Greenland P, Liu K, Guralnik JM, Criqui MH, Dolan
high levels of ERRα associated with expression of ErbB2, in- NC, Chan C, Celic L, Pearce WH, Schneider JR, Sharma L, Clark E,
creased risk of recurrence, and adverse clinical outcomes.161,162 Gibson D, Martin GJ. Leg symptoms in peripheral arterial disease: as-
sociated clinical characteristics and functional impairment. JAMA.
Thus, careful choices of pharmacological agents will need to 2001;286:1599–1606.
be made if mitochondrial biogenesis in patients with PAD will 4. Golomb BA, Dang TT, Criqui MH. Peripheral arterial disease: morbidity
be attempted through ERR modulation. At this time, there is and mortality implications. Circulation. 2006;114:688–699. doi: 10.1161/
CIRCULATIONAHA.105.593442.
minimal literature reporting ERRγ modulators likely to be
5. Hirsch AT, Allison MA, Gomes AS, Corriere MA, Duval S, Ershow
useful for experimental medicine hypothesis testing, leaving AG, Hiatt WR, Karas RH, Lovell MB, McDermott MM, Mendes DM,
only small molecules associated with the unclear benefit/risk Nussmeier NA, Treat-Jacobson D; American Heart Association Council on
ratio of ERRα modulation available for use in the near future. Peripheral Vascular Disease; Council on Cardiovascular Nursing; Council
on Cardiovascular Radiology and Intervention; Council on Cardiovascular
Likewise, no facile methods to clinically modulate the impor- Surgery and Anesthesia; Council on Clinical Cardiology; Council on
tant regulator of mitochondrial expression nuclear respiratory Epidemiology and Prevention. A call to action: women and peripheral ar-
factor 1 exist at this time. Thus, both basic research and drug tery disease: a scientific statement from the American Heart Association.
Circulation. 2012;125:1449–1472. doi: 10.1161/CIR.0b013e31824c39ba.
discovery in the area of mitochondrial biogenesis are newly 6. Goldschmidt-Clermont PJ, Creager MA, Losordo DW, Lorsordo DW,
needed to generate hypotheses and new targets for clinical Lam GK, Wassef M, Dzau VJ. Atherosclerosis 2005: recent discoveries
testing. and novel hypotheses. Circulation. 2005;112:3348–3353. doi: 10.1161/
CIRCULATIONAHA.105.577460.
7. Bhatt DL, Steg PG, Ohman EM, Hirsch AT, Ikeda Y, Mas JL, Goto S, Liau
Conclusions CS, Richard AJ, Röther J, Wilson PW; REACH Registry Investigators.
PAD represents a complex pathophysiology that results in International prevalence, recognition, and treatment of cardiovascular risk
significant functional limitations in exercise performance. A factors in outpatients with atherothrombosis. JAMA. 2006;295:180–189.
doi: 10.1001/jama.295.2.180.
dominant mechanism in the initiation of the exercise limita- 8. Berger JS, Hochman J, Lobach I, Adelman MA, Riles TS, Rockman CB.
tion is a reduction in exercise blood flow and oxygen delivery Modifiable risk factor burden and the prevalence of peripheral artery dis-
to exercising skeletal muscle in the lower extremity. This flow ease in different vascular territories. J Vasc Surg. 2013;58:673–81.e1. doi:
10.1016/j.jvs.2013.01.053.
limitation is also associated with skeletal muscle ischemia with 9. Grenon SM, Vittinghoff E, Owens CD, Conte MS, Whooley M, Cohen
walking exercise and reperfusion with rest that set up a cas- BE. Peripheral artery disease and risk of cardiovascular events in patients
cade of pathophysiological responses leading to further tissue with coronary artery disease: insights from the Heart and Soul Study. Vasc
injury, endothelial dysfunction, muscle metabolic abnormali- Med. 2013;18:176–184. doi: 10.1177/1358863X13493825.
10. Brevetti G, Giugliano G, Brevetti L, Hiatt WR. Inflammation in periph-
ties, and other changes that further reduce exercise capacity. eral artery disease. Circulation. 2010;122:1862–1875. doi: 10.1161/
Understanding these sequelae offers insights into mechanisms CIRCULATIONAHA.109.918417.
1536  Circulation Research  April 24, 2015

11. Steg PG, Bhatt DL, Wilson PW, D’Agostino R Sr, Ohman EM, Röther 30. Karayannacos PE, Talukder N, Nerem RM, Roshon S, Vasko JS. The role
J, Liau CS, Hirsch AT, Mas JL, Ikeda Y, Pencina MJ, Goto S; REACH of multiple noncritical arterial stenoses in the pathogenesis of ischemia. J
Registry Investigators. One-year cardiovascular event rates in outpa- Thorac Cardiovasc Surg. 1977;73:458–469.
tients with atherothrombosis. JAMA. 2007;297:1197–1206. doi: 10.1001/ 31. Sørlie D, Myhre K. Lower leg blood flow in intermittent claudication.
jama.297.11.1197. Scand J Clin Lab Invest. 1978;38:171–179.
12. Pande RL, Perlstein TS, Beckman JA, Creager MA. Secondary prevention 32. Hiatt WR, Brass EP. Pathophysiology of intermittent claudication. In:
and mortality in peripheral artery disease: National Health and Nutrition Creager MA, Dzau VJ, Loscalzo J, eds. Vascular Medicine: A Companion
Examination Study, 1999 to 2004. Circulation. 2011;124:17–23. doi: to Braunwald’s Heart Disease. 2nd ed. Philadelphia: Saunders Elsevier;
10.1161/CIRCULATIONAHA.110.003954. 2013;223–230.
13. Hirsch AT, Criqui MH, Treat-Jacobson D, Regensteiner JG, Creager
33. Hansen JE, Sue DY, Oren A, Wasserman K. Relation of oxygen uptake
MA, Olin JW, Krook SH, Hunninghake DB, Comerota AJ, Walsh ME, to work rate in normal men and men with circulatory disorders. Am J
McDermott MM, Hiatt WR. Peripheral arterial disease detection, aware- Cardiol. 1987;59:669–674.
ness, and treatment in primary care. JAMA. 2001;286:1317–1324. 34. Hiatt WR, Marsh RC, Brammell HL, Fee C, Horwitz LD. Effect of aerobic
14. 2011 Writing Group Members; 2005 Writing Committee Members;
conditioning on the peripheral circulation during chronic beta-adrenergic
ACCF/AHA Task Force Members. 2011 ACCF/AHA Focused Update blockade. J Am Coll Cardiol. 1984;4:958–963.
of the Guideline for the Management of Patients With Peripheral Artery 35. Wasserman K. Coupling of external to cellular respiration during exercise:
Disease (Updating the 2005 Guideline): a report of the American College the wisdom of the body revisited. Am J Physiol. 1994;266:E519–E539.
of Cardiology Foundation/American Heart Association Task Force on 36. Bragadeesh T, Sari I, Pascotto M, Micari A, Kaul S, Lindner JR. Detection
practice guidelines. Circulation. 2011;124:2020–2045. doi: 10.1161/ of peripheral vascular stenosis by assessing skeletal muscle flow reserve. J
CIR.0b013e31822e80c3. Am Coll Cardiol. 2005;45:780–785. doi: 10.1016/j.jacc.2004.11.045.
15. Armstrong EJ, Chen DC, Westin GG, Singh S, McCoach CE, Bang H, 37. Young DF, Cholvin NR, Kirkeeide RL, Roth AC. Hemodynamics of arte-
Yeo KK, Anderson D, Amsterdam EA, Laird JR. Adherence to guide- rial stenoses at elevated flow rates. Circ Res. 1977;41:99–107.
line-recommended therapy is associated with decreased major adverse 38. Opolski MP, Pregowski J, Kruk M, Kepka C, Staruch AD, Witkowski
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

cardiovascular events and major adverse limb events among patients A. Advanced 2-dimensional quantitative coronary angiographic analy-
with peripheral arterial disease. J Am Heart Assoc. 2014;3:e000697. doi: sis for prediction of fractional flow reserve in intermediate coro-
10.1161/JAHA.113.000697. nary stenoses. J Thorac Imaging. 2014;29:217–223. doi: 10.1097/
16. Aboyans V, Criqui MH, Abraham P, et al; American Heart Association RTI.0000000000000058.
Council on Peripheral Vascular Disease; Council on Epidemiology and 39. Tonino PA, De Bruyne B, Pijls NH, Siebert U, Ikeno F, van’ t Veer
Prevention; Council on Clinical Cardiology; Council on Cardiovascular M, Klauss V, Manoharan G, Engstrøm T, Oldroyd KG, Ver Lee PN,
Nursing; Council on Cardiovascular Radiology and Intervention, and MacCarthy PA, Fearon WF; FAME Study Investigators. Fractional flow
Council on Cardiovascular Surgery and Anesthesia. Measurement and reserve versus angiography for guiding percutaneous coronary interven-
interpretation of the ankle-brachial index: a scientific statement from tion. N Engl J Med. 2009;360:213–224. doi: 10.1056/NEJMoa0807611.
the American Heart Association. Circulation. 2012;126:2890–2909. doi: 40. Lotfi AS, Sivalingam SK, Giugliano GR, Ashraf J, Visintainer P. Use
10.1161/CIR.0b013e318276fbcb. of fraction flow reserve to predict changes over time in management
17. von Ziegler F, Costa MA. The role of CT and MRI in the assessment of of superficial femoral artery. J Interv Cardiol. 2012;25:71–77. doi:
peripheral vascular disease. Curr Cardiol Rep. 2007;9:412–419. 10.1111/j.1540-8183.2011.00691.x.
18. McDermott MM, Mehta S, Greenland P. Exertional leg symptoms other 41. Ledermann HP, Schulte AC, Heidecker HG, Aschwanden M, Jäger KA,
than intermittent claudication are common in peripheral arterial disease. Scheffler K, Steinbrich W, Bilecen D. Blood oxygenation level-dependent
Arch Intern Med. 1999;159:387–392. magnetic resonance imaging of the skeletal muscle in patients with pe-
19. Smith GD, Shipley MJ, Rose G. Intermittent claudication, heart disease risk ripheral arterial occlusive disease. Circulation. 2006;113:2929–2935. doi:
factors, and mortality. The Whitehall Study. Circulation. 1990;82:1925–1931. 10.1161/CIRCULATIONAHA.105.605717.
20. McDermott MM, Guralnik JM, Ferrucci L, Tian L, Liu K, Liao Y,
42. Roseguini BT, Hirai DM, Alencar MC, Ramos RP, Silva BM, Wolosker
Green D, Sufit R, Hoff F, Nishida T, Sharma L, Pearce WH, Schneider N, Neder JA, Nery LE. Sildenafil improves skeletal muscle oxygenation
JR, Criqui MH. Asymptomatic peripheral arterial disease is associ- during exercise in men with intermittent claudication. Am J Physiol
ated with more adverse lower extremity characteristics than intermit- Regul Integr Comp Physiol. 2014;307:R396–R404. doi: 10.1152/
tent claudication. Circulation. 2008;117:2484–2491. doi: 10.1161/ ajpregu.00183.2014.
CIRCULATIONAHA.107.736108. 43. Hickner RC, Fisher JS, Ehsani AA, Kohrt WM. Role of nitric oxide in
21. Hiatt WR, Nawaz D, Regensteiner JG, Hossack KF. The evaluation of skeletal muscle blood flow at rest and during dynamic exercise in humans.
exercise performance in patients with peripheral vascular disease. J Am J Physiol. 1997;273:H405–H410.
Cardiopulmonary Rehabil. 1988;12:525–532. 44. Liao JK, Bettmann MA, Sandor T, Tucker JI, Coleman SM, Creager
22. Gardner AW. Claudication pain and hemodynamic responses to exercise MA. Differential impairment of vasodilator responsiveness of peripheral
in younger and older peripheral arterial disease patients. J Gerontol. resistance and conduit vessels in humans with atherosclerosis. Circ Res.
1993;48:M231–M236. 1991;68:1027–1034.
23. Gardner AW, Killewich LA, Katzel LI, Womack CJ, Montgomery PS, 45. Schellong SM, Böger RH, Burchert W, Bode-Böger SM, Galland A,
Otis RB, Fonong T. Relationship between free-living daily physical activ- Frölich JC, Hundeshagen H, Alexander K. Dose-related effect of intrave-
ity and peripheral circulation in patients with intermittent claudication. nous L-arginine on muscular blood flow of the calf in patients with periph-
Angiology. 1999;50:289–297. eral vascular disease: a H215O positron emission tomography study. Clin
24. Gardner AW, Poehlman ET. Assessment of free-living daily physical activ- Sci (Lond). 1997;93:159–165.
ity in older claudicants: validation against the doubly labeled water tech- 46. Tsao PS, Buitrago R, Chang H, Chen UDI, Reaven GM. Effects of dia-
nique. J Gerontol A Biol Sci Med Sci. 1998;53:M275–M280. betes on monocyte-endothelial interactions and endothelial superoxide
25. McDermott MM, Greenland P, Ferrucci L, Criqui MH, Liu K, Sharma L, production in fructose-induced insulin-resistant and hypertensive rats.
Chan C, Celic L, Priyanath A, Guralnik JM. Lower extremity performance Circulation. 1995;92:A2666.
is associated with daily life physical activity in individuals with and with- 47. Mangiafico RA, Malatino LS, Spada RS, Santonocito M, Messina R,
out peripheral arterial disease. J Am Geriatr Soc. 2002;50:247–255. Dell’Arte S, Attinà T. Treadmill exercise-induced release of endothelin-1
26. McDermott MM, Liu K, Greenland P, Guralnik JM, Criqui MH, Chan in patients with peripheral arterial occlusive disease at Fontaine stage IIb.
C, Pearce WH, Schneider JR, Ferrucci L, Celic L, Taylor LM, Vonesh Int Angiol. 2000;19:14–17.
E, Martin GJ, Clark E. Functional decline in peripheral arterial disease: 48. Brevetti G, Silvestro A, Di Giacomo S, Bucur R, Di Donato A, Schiano
associations with the ankle brachial index and leg symptoms. JAMA. V, Scopacasa F. Endothelial dysfunction in peripheral arterial disease is
2004;292:453–461. doi: 10.1001/jama.292.4.453. related to increase in plasma markers of inflammation and severity of pe-
27. Wheeler EC, Brenner ZR. Peripheral vascular anatomy, physiology, and ripheral circulatory impairment but not to classic risk factors and athero-
pathophysiology. AACN Clin Issues. 1995;6:505–514. sclerotic burden. J Vasc Surg. 2003;38:374–379.
28. Pfitzner J. Poiseuille and his law. Anaesthesia. 1976;31:273–275. 49. Grenon SM, Chong K, Alley H, Nosova E, Gasper W, Hiramoto J,

29. Hlavová J, Linhart J, Prerovský I, Ganz V, Fronĕk A. Leg blood flow at Boscardin WJ, Owens CD. Walking disability in patients with peripheral
rest, during and after exercise in normal subjects and in patients with fem- artery disease is associated with arterial endothelial function. J Vasc Surg.
oral artery occlusion. Clin Sci. 1965;29:555–564. 2014;59:1025–1034. doi: 10.1016/j.jvs.2013.10.084.
Hiatt et al   Exercise Limitation in Peripheral Artery Disease   1537

50. Coutinho T, Rooke TW, Kullo IJ. Arterial dysfunction and functional per- 70. de Backer TL, Vander Stichele R, Lehert P, Van Bortel L. Naftidrofuryl
formance in patients with peripheral artery disease: a review. Vasc Med. for intermittent claudication. Cochrane Database Syst Rev.
2011;16:203–211. doi: 10.1177/1358863X11400935. 2012;12:CD001368. doi: 10.1002/14651858.CD001368.pub4.
51. Silvestro A, Scopacasa F, Ruocco A, Oliva G, Schiano V, Zincarelli C, 71. Barradell LB, Brogden RN. Oral naftidrofuryl. A review of its pharmacol-
Brevetti G. Inflammatory status and endothelial function in asymptomatic ogy and therapeutic use in the management of peripheral occlusive arterial
and symptomatic peripheral arterial disease. Vasc Med. 2003;8:225–232. disease. Drugs Aging. 1996;8:299–322.
52. Brendle DC, Joseph LJ, Corretti MC, Gardner AW, Katzel LI. Effects of 72. Bendermacher BL, Willigendael EM, Teijink JA, Prins MH. Supervised
exercise rehabilitation on endothelial reactivity in older patients with pe- exercise therapy versus non-supervised exercise therapy for intermittent
ripheral arterial disease. Am J Cardiol. 2001;87:324–329. claudication. Cochrane Database Syst Rev. 2006;2:CD005263.
53. Januszek R, Mika P, Konik A, Petriczek T, Nowobilski R, Niżankowski R. 73. Hiatt WR, Regensteiner JG, Wolfel EE, Carry MR, Brass EP. Effect of ex-
Effect of treadmill training on endothelial function and walking abilities in ercise training on skeletal muscle histology and metabolism in peripheral
patients with peripheral arterial disease. J Cardiol. 2014;64:145–151. doi: arterial disease. J Appl Physiol (1985). 1996;81:780–788.
10.1016/j.jjcc.2013.12.002. 74. Hiatt WR, Wolfel EE, Meier RH, Regensteiner JG. Superiority of tread-
54. Baltgalvis KA, White K, Li W, et al. Exercise performance and peripheral mill walking exercise versus strength training for patients with peripheral
vascular insufficiency improve with AMPK activation in high-fat diet-fed arterial disease. Implications for the mechanism of the training response.
mice. Am J Physiol Heart Circ Physiol. 2014;306:H1128–H1145. doi: Circulation. 1994;90:1866–1874.
10.1152/ajpheart.00839.2013. 75. Murphy TP, Cutlip DE, Regensteiner JG, et al; CLEVER Study Investigators.
55. Wu WC, Mohler E 3rd, Ratcliffe SJ, Wehrli FW, Detre JA, Floyd TF. Skeletal Supervised exercise versus primary stenting for claudication resulting from
muscle microvascular flow in progressive peripheral artery disease: assessment aortoiliac peripheral artery disease: six-month outcomes from the claudication:
with continuous arterial spin-labeling perfusion magnetic resonance imaging. exercise versus endoluminal revascularization (CLEVER) study. Circulation.
J Am Coll Cardiol. 2009;53:2372–2377. doi: 10.1016/j.jacc.2009.03.033. 2012;125:130–139. doi: 10.1161/CIRCULATIONAHA.111.075770.
56. Pollak AW, Meyer CH, Epstein FH, Jiji RS, Hunter JR, Dimaria JM, 76. Hiatt WR, Creager MA, Amato A, Brass EP. Effect of propionyl-L-car-
Christopher JM, Kramer CM. Arterial spin labeling MR imaging repro- nitine on a background of monitored exercise in patients with claudica-
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

ducibly measures peak-exercise calf muscle perfusion: a study in patients tion secondary to peripheral artery disease. J Cardiopulm Rehabil Prev.
with peripheral arterial disease and healthy volunteers. JACC Cardiovasc 2011;31:125–132. doi: 10.1097/HCR.0b013e3181f1fd65.
Imaging. 2012;5:1224–1230. doi: 10.1016/j.jcmg.2012.03.022. 77. Pernow B, Zetterquist S. Metabolic evaluation of the leg blood flow in
57. Lindner JR, Womack L, Barrett EJ, Weltman J, Price W, Harthun NL, Kaul claudicating patients with arterial obstructions at different levels. Scand J
S, Patrie JT. Limb stress-rest perfusion imaging with contrast ultrasound Clin Lab Invest. 1968;21:277–287.
for the assessment of peripheral arterial disease severity. JACC Cardiovasc 78. Szuba A, Oka RK, Harada R, Cooke JP. Limb hemodynamics are not predic-
Imaging. 2008;1:343–350. doi: 10.1016/j.jcmg.2008.04.001. tive of functional capacity in patients with PAD. Vasc Med. 2006;11:155–163.
79. Hiatt WR, Regensteiner JG, Hargarten ME, Wolfel EE, Brass EP. Benefit
58. Anderson JD, Epstein FH, Meyer CH, Hagspiel KD, Wang H, Berr

of exercise conditioning for patients with peripheral arterial disease.
SS, Harthun NL, Weltman A, Dimaria JM, West AM, Kramer CM.
Circulation. 1990;81:602–609.
Multifactorial determinants of functional capacity in peripheral arterial
80. Hynynen MM, Khalil RA. The vascular endothelin system in hyperten-
disease: uncoupling of calf muscle perfusion and metabolism. J Am Coll
sion–recent patents and discoveries. Recent Pat Cardiovasc Drug Discov.
Cardiol. 2009;54:628–635. doi: 10.1016/j.jacc.2009.01.080.
2006;1:95–108.
59. Woodburn KR, Lowe GD, Rumley A, Love J, Pollock JG. Relation of hae-
81. Vatter H, Seifert V. Ambrisentan, a non-peptide endothelin receptor antagonist.
mostatic, fibrinolytic, and rheological variables to the angiographic extent
Cardiovasc Drug Rev. 2006;24:63–76. doi: 10.1111/j.1527-3466.2006.00063.x.
of peripheral arterial occlusive disease. Int Angiol. 1995;14:346–352.
82. Hong IS, Coe HV, Catanzaro LM. Macitentan for the treatment of pul-
60. Dormandy JA, Hoare E, Colley J, Arrowsmith DE, Dormandy TL.

monary arterial hypertension. Ann Pharmacother. 2014;48:538–547. doi:
Clinical, haemodynamic, rheological, and biochemical findings in 126
10.1177/1060028013518900.
patients with intermittent claudication. Br Med J. 1973;4:576–581.
83. Iglarz M, Binkert C, Morrison K, Fischli W, Gatfield J, Treiber A, Weller
61. Lowe GD, Fowkes FG, Dawes J, Donnan PT, Lennie SE, Housley E.
T, Bolli MH, Boss C, Buchmann S, Capeleto B, Hess P, Qiu C, Clozel M.
Blood viscosity, fibrinogen, and activation of coagulation and leukocytes
Pharmacology of macitentan, an orally active tissue-targeting dual endo-
in peripheral arterial disease and the normal population in the Edinburgh thelin receptor antagonist. J Pharmacol Exp Ther. 2008;327:736–745. doi:
Artery Study. Circulation. 1993;87:1915–1920. 10.1124/jpet.108.142976.
62. Fowkes FG, Lowe GD, Housley E, Rattray A, Rumley A, Elton RA,
84. Forst T, Lübben G, Hohberg C, Kann P, Sachara C, Gottschall V, Friedrich
MacGregor IR, Dawes J. Cross-linked fibrin degradation products, progres- C, Rosskopf R, Pfützner A. Influence of glucose control and improvement
sion of peripheral arterial disease, and risk of coronary heart disease. Lancet. of insulin resistance on microvascular blood flow and endothelial function
1993;342:84–86. in patients with diabetes mellitus type 2. Microcirculation. 2005;12:543–
63. Pande RL, Hiatt WR, Zhang P, Hittel N, Creager MA. A pooled analy- 550. doi: 10.1080/10739680500253402.
sis of the durability and predictors of treatment response of cilostazol in 85. Martens FM, Visseren FL, de Koning EJ, Rabelink TJ. Short-term piogli-
patients with intermittent claudication. Vasc Med. 2010;15:181–188. doi: tazone treatment improves vascular function irrespective of metabolic changes
10.1177/1358863X10361545. in patients with type 2 diabetes. J Cardiovasc Pharmacol. 2005;46:773–778.
64. Brass EP. Intermittent claudication: new targets for drug development. 86. Tsuchiya K, Akaza I, Yoshimoto T, Hirata Y. Pioglitazone improves endo-
Drugs. 2013;73:999–1014. doi: 10.1007/s40265-013-0078-3. thelial function with increased adiponectin and high-density lipoprotein
65. Money SR, Herd JA, Isaacsohn JL, Davidson M, Cutler B, Heckman J, cholesterol levels in type 2 diabetes. Endocr J. 2009;56:691–698.
Forbes WP. Effect of cilostazol on walking distances in patients with in- 87. Edwards AT, Blann AD, Suarez-Mendez VJ, Lardi AM, McCollum CN.
termittent claudication caused by peripheral vascular disease. J Vasc Surg. Systemic responses in patients with intermittent claudication after tread-
1998;27:267–74; discussion 274. mill exercise. Br J Surg. 1994;81:1738–1741.
66. Brass EP, Anthony R, Cobb FR, Koda I, Jiao J, Hiatt WR. The novel 88. Belch JJ, McLaren M, Khan F, Hickman P, Muir AH, Stonebridge PA.
phosphodiesterase inhibitor NM-702 improves claudication-limited exer- The inflammatory process in intermittent claudication. Eur Heart J.
cise performance in patients with peripheral arterial disease. J Am Coll 2002;4:B31–B34. doi: 10.1016/S1520-765X(02)90014-3.
Cardiol. 2006;48:2539–2545. doi: 10.1016/j.jacc.2006.07.064. 89. Belch JJ, Mackay IR, Hill A, Jennings P, McCollum P. Oxidative stress is
67. Brass EP, Cooper LT, Morgan RE, Hiatt WR. A phase II dose-ranging study present in atherosclerotic peripheral arterial disease and further increased
of the phosphodiesterase inhibitor K-134 in patients with peripheral artery by diabetes mellitus. Int Angiol. 1995;14:385–388.
disease and claudication. J Vasc Surg. 2012;55:381–389.e1. doi: 10.1016/j. 90. Brevetti G, De Caterina M, Martone VD, Ungaro B, Corrado F, Silvestro
jvs.2011.09.004. A, de Cristofaro T, Scopacasa F. Exercise increases soluble adhesion mol-
68. Salhiyyah K, Senanayake E, Abdel-Hadi M, Booth A, Michaels JA.
ecules ICAM-1 and VCAM-1 in patients with intermittent claudication.
Pentoxifylline for intermittent claudication. Cochrane Database Syst Rev. Clin Hemorheol Microcirc. 2001;24:193–199.
2012;1:CD005262. doi: 10.1002/14651858.CD005262.pub2. 91. Signorelli SS, Mazzarino MC, Di Pino L, Malaponte G, Porto C, Pennisi
69. Stevens JW, Simpson E, Harnan S, Squires H, Meng Y, Thomas S, G, Marchese G, Costa MP, Digrandi D, Celotta G, Virgilio V. High cir-
Michaels J, Stansby G. Systematic review of the efficacy of cilostazol, culating levels of cytokines (IL-6 and TNFalpha), adhesion molecules
naftidrofuryl oxalate and pentoxifylline for the treatment of intermittent (VCAM-1 and ICAM-1) and selectins in patients with peripheral arterial
claudication. Br J Surg. 2012;99:1630–1638. doi: 10.1002/bjs.8895. disease at rest and after a treadmill test. Vasc Med. 2003;8:15–19.
1538  Circulation Research  April 24, 2015

92. Blann AD, Dobrotova M, Kubisz P, McCollum CN. von Willebrand fac- D, Mann J, Guertin MC, L’Allier PL. Effects of the P-selectin antago-
tor, soluble P-selectin, tissue plasminogen activator and plasminogen ac- nist inclacumab on myocardial damage after percutaneous coronary in-
tivator inhibitor in atherosclerosis. Thromb Haemost. 1995;74:626–630. tervention for non-ST-segment elevation myocardial infarction: results
93. Gardner AW, Parker DE, Montgomery PS, Sosnowska D, Casanegra of the SELECT-ACS trial. J Am Coll Cardiol. 2013;61:2048–2055. doi:
AI, Esponda OL, Ungvari Z, Csiszar A, Sonntag WE. Impaired vas- 10.1016/j.jacc.2013.03.003.
cular endothelial growth factor A and inflammation in patients 114. Kling D, Stucki C, Kronenberg S, Tuerck D, Rhéaume E, Tardif JC,
with peripheral artery disease. Angiology. 2014;65:683–690. doi: Gaudreault J, Schmitt C. Pharmacological control of platelet-leukocyte
10.1177/0003319713501376. interactions by the human anti-P-selectin antibody inclacumab–preclini-
94. Nylaende M, Kroese A, Stranden E, Morken B, Sandbaek G, Lindahl cal and clinical studies. Thromb Res. 2013;131:401–410. doi: 10.1016/j.
AK, Arnesen H, Seljeflot I. Markers of vascular inflammation are associ- thromres.2013.02.020.
ated with the extent of atherosclerosis assessed as angiographic score and 115. Ahimastos AA, Walker PJ, Askew C, Leicht A, Pappas E, Blombery
treadmill walking distances in patients with peripheral arterial occlusive P, Reid CM, Golledge J, Kingwell BA. Effect of ramipril on walking
disease. Vasc Med. 2006;11:21–28. times and quality of life among patients with peripheral artery disease
95. McDermott MM, Greenland P, Green D, Guralnik JM, Criqui MH, Liu and intermittent claudication: a randomized controlled trial. JAMA.
K, Chan C, Pearce WH, Taylor L, Ridker PM, Schneider JR, Martin 2013;309:453–460. doi: 10.1001/jama.2012.216237.
G, Rifai N, Quann M, Fornage M. D-dimer, inflammatory markers, 116. Ahimastos AA, Latouche C, Natoli AK, Reddy-luthmoodoo M,

and lower extremity functioning in patients with and without periph- Golledge J, Kingwell BA. Potential vascular mechanisms of ramipril
eral arterial disease. Circulation. 2003;107:3191–3198. doi: 10.1161/01. induced increases in walking ability in patients with intermittent clau-
CIR.0000074227.53616.CC. dication. Circ Res. 2014;114:1144–1155. doi: 10.1161/CIRCRESAHA.
96. McDermott MM, Liu K, Guralnik JM, Ferrucci L, Green D, Greenland 114.302420.
P, Tian L, Criqui MH, Lo C, Rifai N, Ridker PM, Zheng J, Pearce W. 117. Shahin Y, Mazari F, Chetter I. Do angiotensin converting enzyme in-
Functional decline in patients with and without peripheral arterial dis- hibitors improve walking distance in patients with symptomatic lower
ease: predictive value of annual changes in levels of C-reactive protein limb arterial disease? A systematic review and meta-analysis of ran-
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

and D-dimer. J Gerontol A Biol Sci Med Sci. 2006;61:374–379. domised controlled trials. Int J Surg. 2011;9:209–213. doi: 10.1016/j.
97. Brass EP, Cooper LT, Hanson P, Hiatt WR. Association of clinical at- ijsu.2010.12.006.
tributes and treadmill walking performance in patients with claudication 118. Brass EP, Koster D, Hiatt WR, Amato A. A systematic review and
due to peripheral artery disease. J Vasc Surg. 2013;58:396–403. doi: meta-analysis of propionyl-L-carnitine effects on exercise perfor-
10.1016/j.jvs.2012.12.077. mance in patients with claudication. Vasc Med. 2013;18:3–12. doi:
98. Dröse S, Brandt U. Molecular mechanisms of superoxide production by 10.1177/1358863X12467491.
the mitochondrial respiratory chain. Adv Exp Med Biol. 2012;748:145– 119. LaRosa JC, He J, Vupputuri S. Effect of statins on risk of coronary
169. doi: 10.1007/978-1-4614-3573-0_6. disease: a meta-analysis of randomized controlled trials. JAMA.
99. Hickman P, Harrison DK, Hill A, McLaren M, Tamei H, McCollum PT, 1999;282:2340–2346.
Belch JJ. Exercise in patients with intermittent claudication results in the 120. Hiatt WR, Hirsch AT, Creager MA, Rajagopalan S, Mohler ER,

generation of oxygen derived free radicals and endothelial damage. Adv Ballantyne CM, Regensteiner JG, Treat-Jacobson D, Dale RA, Rooke
Exp Med Biol. 1994;361:565–570. T. Effect of niacin ER/lovastatin on claudication symptoms in pa-
100. Ciuffetti G, Mercuri M, Mannarino E, Ott C, Lombardini R, Pasqualini L, tients with peripheral artery disease. Vasc Med. 2010;15:171–179. doi:
Lupattelli G, Santambrogio L. Free radical production in peripheral vas- 10.1177/1358863X09360579.
cular disease. A risk for critical ischaemia? Int Angiol. 1991;10:81–87. 121. Mohler ER 3rd, Hiatt WR, Creager MA. Cholesterol reduction with
101. Melov S, Shoffner JM, Kaufman A, Wallace DC. Marked increase in the atorvastatin improves walking distance in patients with peripheral ar-
number and variety of mitochondrial DNA rearrangements in aging hu- terial disease. Circulation. 2003;108:1481–1486. doi: 10.1161/01.
man skeletal muscle. Nucleic Acids Res. 1995;23:4122–4126. CIR.0000090686.57897.F5.
102. Bhat HK, Hiatt WR, Hoppel CL, Brass EP. Skeletal muscle mitochon- 122. Regensteiner JG, Wolfel EE, Brass EP, Carry MR, Ringel SP, Hargarten
drial DNA injury in patients with unilateral peripheral arterial disease. ME, Stamm ER, Hiatt WR. Chronic changes in skeletal muscle his-
Circulation. 1999;99:807–812. tology and function in peripheral arterial disease. Circulation.
103. Ji LL. Modulation of skeletal muscle antioxidant defense by exercise: 1993;87:413–421.
role of redox signaling. Free Radic Biol Med. 2008;44:142–152. doi: 123. Askew CD, Green S, Walker PJ, Kerr GK, Green AA, Williams AD,
10.1016/j.freeradbiomed.2007.02.031. Febbraio MA. Skeletal muscle phenotype is associated with exer-
104. Oh-ishi S, Kizaki T, Nagasawa J, Izawa T, Komabayashi T, Nagata N, cise tolerance in patients with peripheral arterial disease. J Vasc Surg.
Suzuki K, Taniguchi N, Ohno H. Effects of endurance training on super- 2005;41:802–807. doi: 10.1016/j.jvs.2005.01.037.
oxide dismutase activity, content and mRNA expression in rat muscle. 124. McGuigan MR, Bronks R, Newton RU, Sharman MJ, Graham JC, Cody
Clin Exp Pharmacol Physiol. 1997;24:326–332. DV, Kraemer WJ. Muscle fiber characteristics in patients with peripheral
105. Vassilakopoulos T, Deckman G, Kebbewar M, Rallis G, Harfouche R, arterial disease. Med Sci Sports Exerc. 2001;33:2016–2021.
Hussain SN. Regulation of nitric oxide production in limb and ventila- 125. Mitchell RG, Duscha BD, Robbins JL, Redfern SI, Chung J, Bensimhon
tory muscles during chronic exercise training. Am J Physiol Lung Cell DR, Kraus WE, Hiatt WR, Regensteiner JG, Annex BH. Increased
Mol Physiol. 2003;284:L452–L457. doi: 10.1152/ajplung.00270.2002. levels of apoptosis in gastrocnemius skeletal muscle in patients
106. Sumida S, Nakamura H, Yodoi J. Thioredoxin induction of peripheral with peripheral arterial disease. Vasc Med. 2007;12:285–290. doi:
blood mononuclear cells in mice in response to a single bout of swim- 10.1177/1358863X07084858.
ming exercise. Gen Physiol Biophys. 2004;23:241–249. 126. McDermott MM, Ferrucci L, Guralnik JM, Tian L, Green D, Liu K, Tan
107. Naldini A, Carraro F. Role of inflammatory mediators in angiogenesis. J, Liao Y, Pearce WH, Schneider JR, Ridker P, Rifai N, Hoff F, Criqui
Curr Drug Targets Inflamm Allergy. 2005;4:3–8. MH. Elevated levels of inflammation, d-dimer, and homocysteine are
108. Gahmberg CG, Tolvanen M, Kotovuori P. Leukocyte adhesion–structure associated with adverse calf muscle characteristics and reduced calf
and function of human leukocyte beta2-integrins and their cellular li- strength in peripheral arterial disease. J Am Coll Cardiol. 2007;50:897–
gands. Eur J Biochem. 1997;245:215–232. 905. doi: 10.1016/j.jacc.2007.05.017.
109. Comi G. Treatment of multiple sclerosis: role of natalizumab. Neurol Sci. 127. Steinacker JM, Opitz-Gress A, Baur S, Lormes W, Bolkart K, Sunder-
2009;30 Suppl 2:S155–S158. doi: 10.1007/s10072-009-0147-2. Plassmann L, Liewald F, Lehmann M, Liu Y. Expression of myosin
110. Schmidt KJ, Büning J, Jankowiak C, Lehnert H, Fellermann K. Crohn’s heavy chain isoforms in skeletal muscle of patients with peripheral arte-
targeted therapy: myth or real goal? Curr Drug Discov Technol. rial occlusive disease. J Vasc Surg. 2000;31:443–449.
2009;6:290–298. 128. England JD, Ferguson MA, Hiatt WR, Regensteiner JG. Progression of
111. Enlinomab Acute Stroke Investigators. Use of anti-ICAM-1 therapy in neuropathy in peripheral arterial disease. Muscle Nerve. 1995;18:380–
ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 387. doi: 10.1002/mus.880180403.
2001;57:1428–1434. 129. Garg PK, Liu K, Ferrucci L, Guralnik JM, Criqui MH, Tian L, Sufit R,
112. Lin KC, Castro AC. Very late antigen 4 (VLA4) antagonists as anti-in- Nishida T, Tao H, Liao Y, McDermott MM. Lower extremity nerve func-
flammatory agents. Curr Opin Chem Biol. 1998;2:453–457. tion, calf skeletal muscle characteristics, and functional performance in
113. Tardif JC, Tanguay JF, Wright SS, Duchatelle V, Petroni T, Grégoire JC, peripheral arterial disease. J Am Geriatr Soc. 2011;59:1855–1863. doi:
Ibrahim R, Heinonen TM, Robb S, Bertrand OF, Cournoyer D, Johnson 10.1111/j.1532-5415.2011.03600.x.
Hiatt et al   Exercise Limitation in Peripheral Artery Disease   1539

130. McDermott MM, Ferrucci L, Guralnik J, Tian L, Liu K, Hoff F, Liao 147. Brass EP, Hoppel CL. Relationship between acid-soluble carnitine and
Y, Criqui MH. Pathophysiological changes in calf muscle predict coenzyme A pools in vivo. Biochem J. 1980;190:495–504.
mobility loss at 2-year follow-up in men and women with periph- 148. Hiatt WR, Nawaz D, Brass EP. Carnitine metabolism during exercise
eral arterial disease. Circulation. 2009;120:1048–1055. doi: 10.1161/ in patients with peripheral vascular disease. J Appl Physiol (1985).
CIRCULATIONAHA.108.842328. 1987;62:2383–2387.
131. McDermott MM, Liu K, Tian L, Guralnik JM, Criqui MH, Liao Y, 149. Hiatt WR, Wolfel EE, Regensteiner JG, Brass EP. Skeletal muscle car-
Ferrucci L. Calf muscle characteristics, strength measures, and mortality nitine metabolism in patients with unilateral peripheral arterial disease.
in peripheral arterial disease: a longitudinal study. J Am Coll Cardiol. J Appl Physiol (1985). 1992;73:346–353.
2012;59:1159–1167. doi: 10.1016/j.jacc.2011.12.019. 150. Barker GA, Green S, Green AA, Walker PJ. Walking performance, oxy-
132. Singh N, Liu K, Tian L, Criqui MH, Guralnik JM, Ferrucci L, Liao Y, gen uptake kinetics and resting muscle pyruvate dehydrogenase complex
McDermott MM. Leg strength predicts mortality in men but not in wom- activity in peripheral arterial disease. Clin Sci (Lond). 2004;106:241–
en with peripheral arterial disease. J Vasc Surg. 2010;52:624–631. doi: 249. doi: 10.1042/CS20030244.
10.1016/j.jvs.2010.03.066. 151. Andreux PA, Houtkooper RH, Auwerx J. Pharmacological approaches to
133. Attie KM, Borgstein NG, Yang Y, Condon CH, Wilson DM, Pearsall AE, restore mitochondrial function. Nat Rev Drug Discov. 2013;12:465–483.
Kumar R, Willins DA, Seehra JS, Sherman ML. A single ascending-dose doi: 10.1038/nrd4023.
study of muscle regulator ACE-031 in healthy volunteers. Muscle Nerve. 152. Hou XY, Green S, Askew CD, Barker G, Green A, Walker PJ. Skeletal
2013;47:416–423. doi: 10.1002/mus.23539. muscle mitochondrial ATP production rate and walking perfor-
134. Campbell C, Escolar D, Mah J. A phase 2, randomized, placebo con- mance in peripheral arterial disease. Clin Physiol Funct Imaging.
trolled, multiple ascending-dose study of ACE-031, a soluble activin 2002;22:226–232.
receptor Type IIB, in boys with Duchenne muscular dystrophy (DMD). 153. Russell AP, Feilchenfeldt J, Schreiber S, Praz M, Crettenand A, Gobelet
Neurology. 2012;78:P04.088. C, Meier CA, Bell DR, Kralli A, Giacobino JP, Dériaz O. Endurance
135. Wagner KR, Fleckenstein JL, Amato AA, et al. A phase I/IItrial of training in humans leads to fiber type-specific increases in levels of per-
MYO-029 in adult subjects with muscular dystrophy. Ann Neurol. oxisome proliferator-activated receptor-gamma coactivator-1 and peroxi-
2008;63:561–571. doi: 10.1002/ana.21338. some proliferator-activated receptor-alpha in skeletal muscle. Diabetes.
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

136. Deleted in proof. 2003;52:2874–2881.


137. Lach-Trifilieff E, Minetti GC, Sheppard K, Ibebunjo C, Feige JN,
154. Kramer DM, Ahlsen M, Norrbom J, Jansson E, Hjeltnes N, Gustafsson
Hartmann S, Brachat S, Rivet H, Koelbing C, Morvan F, Hatakeyama S, T, Krook A. Human skeletal muscle fibre type variations corre-
Glass DJ. An antibody blocking activin type II receptors induces strong late with PPARα, PPARd and PGC-1α mRNA. Acta Physiol (Oxf)..
skeletal muscle hypertrophy and protects from atrophy. Mol Cell Biol. 2006;188:207–216.
2014;34:606–618. doi: 10.1128/MCB.01307-13. 155. Peters JM, Foreman JE, Gonzalez FJ. Dissecting the role of peroxisome
138. Grassi B. Regulation of oxygen consumption at exercise onset: is it really proliferator-activated receptor-β/δ (PPARβ/δ) in colon, breast, and lung
controversial? Exerc Sport Sci Rev. 2001;29:134–138. carcinogenesis. Cancer Metastasis Rev. 2011;30:619–640. doi: 10.1007/
139. Bauer TA, Regensteiner JG, Brass EP, Hiatt WR. Oxygen uptake kinetics s10555-011-9320-1.
during exercise are slowed in patients with peripheral arterial disease. J 156. Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang
Appl Physiol (1985). 1999;87:809–816. Y. Sirtuin 3, a new target of PGC-1alpha, plays an important role in
140. Bauer TA, Brass EP, Barstow TJ, Hiatt WR. Skeletal muscle StO2 ki- the suppression of ROS and mitochondrial biogenesis. PLoS One.
netics are slowed during low work rate calf exercise in peripheral ar- 2010;5:e11707. doi: 10.1371/journal.pone.0011707.
terial disease. Eur J Appl Physiol. 2007;100:143–151. doi: 10.1007/ 157. Ranhotra HS. Estrogen-related receptor alpha and mitochondria: tale of
s00421-007-0412-0. the titans. J Recept Signal Transduct Res. 2014;15:1–5.
141. Barker GA, Green S, Green AA, Walker PJ. Walking performance, oxy- 158. Procaccio V, Bris C, Chao de la Barca JM, Oca F, Chevrollier A, Amati-
gen uptake kinetics and resting muscle pyruvate dehydrogenase complex Bonneau P, Bonneau D, Reynier P. Perspectives of drug-based neuropro-
activity in peripheral arterial disease. Clin Sci (Lond). 2004;106:241– tection targeting mitochondria. Rev Neurol (Paris). 2014;170:390–400.
249. doi: 10.1042/CS20030244. doi: 10.1016/j.neurol.2014.03.005.
142. Kemp GJ, Hands LJ, Ramaswami G, Taylor DJ, Nicolaides A, Amato 159. Shao D, Liu Y, Liu X, Zhu L, Cui Y, Cui A, Qiao A, Kong X, Liu
A, Radda GK. Calf muscle mitochondrial and glycogenolytic ATP syn- Y, Chen Q, Gupta N, Fang F, Chang Y. PGC-1 beta-regulated mito-
thesis in patients with claudication due to peripheral vascular disease chondrial biogenesis and function in myotubes is mediated by NRF-
analysed using 31P magnetic resonance spectroscopy. Clin Sci (Lond). 1 and ERR alpha. Mitochondrion. 2010;10:516–527. doi: 10.1016/j.
1995;89:581–590. mito.2010.05.012.
143. Rouslin W. Mitochondrial complexes I, II, III, IV, and V in myocardial 160. Pokrywka A, Cholbinski P, Kaliszewski P, Kowalczyk K, Konczak D,
ischemia and autolysis. Am J Physiol. 1983;244:H743–H748. Zembron-Lacny A. Metabolic modulators of the exercise response:
144. Brass EP, Hiatt WR, Gardner AW, Hoppel CL. Decreased NADH doping control analysis of an agonist of the peroxisome proliferator-
dehydrogenase and ubiquinol-cytochrome c oxidoreductase in activated receptor δ (GW501516) and 5-aminoimidazole-4-carboxamide
peripheral arterial disease. Am J Physiol Heart Circ Physiol. ribonucleotide (AICAR). J Physiol Pharmacol. 2014;65:469–476.
2001;280:H603–H609. 161. Ariazi EA, Clark GM, Mertz JE. Estrogen-related receptor alpha and
145. Patel RP, T Cornwell, Darley-Usmar VM. The biochemistry of nitric ox- estrogen-related receptor gamma associate with unfavorable and fa-
ide and peroxynitrite: implications for mitochondrial function. In: Packer vorable biomarkers, respectively, in human breast cancer. Cancer Res.
L, Cadenas E, eds. Understanding the Process of Aging: The Roles of 2002;62:6510–6518.
Mitochondria, Free Radicals, and Antioxidants. New York, NY: Marcel 162. Suzuki T, Miki Y, Moriya T, Shimada N, Ishida T, Hirakawa H, Ohuchi
Dekker. 1999;39–56. N, Sasano H. Estrogen-related receptor alpha in human breast carcino-
146. Bieber LL. Carnitine. Annu Rev Biochem. 1988;57:261–283. doi:
ma as a potent prognostic factor. Cancer Res. 2004;64:4670–4676. doi:
10.1146/annurev.bi.57.070188.001401. 10.1158/0008-5472.CAN-04-0250.
Pathogenesis of the Limb Manifestations and Exercise Limitations in Peripheral Artery
Disease
William R. Hiatt, Ehrin J. Armstrong, Christopher J. Larson and Eric P. Brass
Downloaded from http://circres.ahajournals.org/ by guest on May 11, 2018

Circ Res. 2015;116:1527-1539


doi: 10.1161/CIRCRESAHA.116.303566
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2015 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/116/9/1527

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation Research is online at:


http://circres.ahajournals.org//subscriptions/

Anda mungkin juga menyukai