You are on page 1of 19

Exercise in Cardiovascular Disease

Cardiovascular Effects of Exercise Training


Molecular Mechanisms
Stephan Gielen, MD; Gerhard Schuler, MD; Volker Adams, PhD

I n the natural habitat of our ancestors, physical activity


was not a preventive intervention but a matter of
survival. In this hostile environment with scarce food and
Cardiac Effects of Exercise
Left Ventricular Myocardium and
Ventricular Arrhythmias
ubiquitous dangers, human genes were selected to opti-
The maintenance of left ventricular (LV) mass and function
mize aerobic metabolic pathways and conserve energy for
depends on regular exercise. Prolonged periods of physical
potential future famines.1 Cardiac and vascular functions
inactivity, as studied in bed rest trials, lead to significant
were continuously challenged by intermittent bouts of
reductions in LV mass and impaired cardiac compliance,
high-intensity physical activity and adapted to meet the
resulting in reduced upright stroke volume and orthostatic
metabolic demands of the working skeletal muscle under
intolerance.3 In contrast, a group of bed rest subjects random-
these conditions.
ized to regular supine lower-body negative pressure treadmill
When speaking about molecular cardiovascular effects of
exercise showed an increase in LV mass and a preserved LV
exercise, we should keep in mind that most of the changes
stoke volume.4 In previously sedentary healthy subjects, a
from baseline are probably a return to normal values. The
12-week moderate exercise program induced a mild cardiac
statistical average of physical activity in Western societies is
hypertrophic response as measured by cardiac magnetic
so much below the levels normal for our genetic background
resonance imaging.5 These findings highlight the plasticity of
that sedentary lifestyle in combination with excess food
LV mass and function in relation to the current level of
intake has surpassed smoking as the No. 1 preventable cause
physical activity.
of death in the United States.2
Physical activity has been shown to have beneficial Normal LV Contractile Function
effects on glucose metabolism, skeletal muscle function, In individuals with normal LV function, exercise-related
ventilator muscle strength, bone stability, locomotor coor- cardiac effects may be subdivided into 3 entities: (1) preven-
dination, psychological well-being, and other organ func- tion of cardiac pathologies associated with aging; (2) cardiac
tions. However, in the context of this review, we will focus adaptation to strenuous regular exercise training resulting in
entirely on important molecular effects on the cardiovas- physiological cardiac hypertrophy commonly known as ath-
cular system. The aim of this review is to provide a lete’s heart; and (3) prevention of impaired systolic and/or
bird’s-eye view on what is known and unknown about the diastolic cardiac function associated with short-term chal-
physiological and biochemical mechanisms involved in lenges (eg, ischemia/reperfusion [I/R], doxorubicin).
mediating exercise-induced cardiovascular effects. The
Prevention of Age-Related Cardiac Pathologies
resulting map is surprisingly detailed in some areas (ie,
endothelial function), whereas other areas, such as direct Oxidative Stress. Theoretically, tissues with fewer cell divi-
cardiac training effects in heart failure, are still incom- sions are more susceptible to cumulative damage by reactive
pletely understood. oxygen species (ROS) than tissues with high replication rates.
For practical purposes, we have decided to use primarily an In animal models of aging, increased malondialdehyde levels
anatomic approach to present key data on exercise effects on were observed in sedentary old rats. Although the expression
of antioxidative enzymes such as superoxide dismutase
cardiac and vascular function. For the cardiac effects, the left
(SOD) was unchanged,6,7 its enzymatic activity was reduced,8
ventricle and the cardiac valves will be described separately;
resulting in a net decline of antioxidative protection. Regular
for the vascular effects, we will follow the arterial vascular physical exercise seems to delay the accumulation of ROS-
tree, addressing changes in the aorta, the large conduit mediated cell damage by improving the antioxidative protec-
arteries, the resistance vessels, and the microcirculation tion in the myocardium. In trained old rats, Rinaldi et al7
before turning our attention toward the venous and the found increased myocardial protein expression of MnSOD
pulmonary circulation (Figure 1). and Cu/Zn SOD.

From the Department of Internal Medicine/Cardiology, University of Leipzig, Heart Center, Leipzig, Germany.
Correspondence to Volker Adams, PhD, Department of Internal Medicine/Cardiology, University of Leipzig, Heart Center, Strümpellstraße 39, 04289
Leipzig, Germany. E-mail adav@medizin.uni-leipzig.de
(Circulation. 2010;122:1221-1238.)
© 2010 American Heart Association, Inc.
Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.110.939959

1221
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
1222 Circulation September 21, 2010

Figure 1. Summary of exercise-mediated effects on different parts of the cardiovascular system. Max. indicates maximum; HFNEF,
heart failure with normal ejection fraction; ANP, atrial natriuretic peptide; and BNP, brain natriuretic peptide.

Heat Shock Proteins. Heat shock proteins (HSP) are highly that so-called healthy aging is in fact primarily sedentary
conserved chaperones that are involved in the folding of aging and that the lack of physical activity may be more
newly synthesized or damaged proteins. The overexpression relevant to the observed pathologies than aging by itself.
of HSP70 in cardiomyocytes protects against ischemic dam-
age and is associated with increased cell survival.9 Stress- Cardiac Adaptation to Strenuous Exercise and
induced synthesis of HSP70 is reduced in the aged cardio- Cardiac Fatigue
vascular system, consistent with a diminished stress Athlete’s Heart. In normal individuals, regular high-intensity
resistance.10 HSP70 and HSP27 were also elevated in trained physical activity for ⬎5 to 6 hours per week may result in
old rats versus sedentary old rats, indicating improved resis- cardiac adaptations known as athlete’s heart, resulting in
tance to ischemic insults.7 compensatory myocardial hypertrophy, which is more eccen-
Cardiomyocyte Apoptosis and Regeneration. Despite the tric in endurance training and more concentric in resistance
increase in LV mass generally observed with aging, the training.17 Endurance-trained athletes show mean LV end-di-
number of cardiomyocytes declines by ⬇30%.11 The expo- astolic diameters of 53.7 mm compared with 49.6 mm in
nential increase of cardiomyocyte death by apoptosis or normal subjects17; however, 14% of 1309 athletes examined
necrosis and the loss of cardiac regeneration by stem cells by Pelliccia et al18 showed LV end-diastolic diameters be-
have been discussed as key factors for this decline in cell tween 66 and 70 mm. The diagnostic strategies to distinguish
number.12,13 The increase in apoptosis may be a consequence normal LV hypertrophy from hypertrophic cardiomyopathy
of decreased mitochondrial membrane stability and perme- are a matter of continuing discussions (reviewed by Fagard19
ability transition pore formation, which leads to cytochrome c and Maron20).
release into the cytosol, resulting in activation of caspase-3 LV physiological hypertrophy is caused by a proportional
and -9.12 Exercise training seems to be highly protective increase in myocardial cell length and width without evidence
against cardiomyocyte apoptosis by modulating proapoptotic of myocardial hyperplasia in the majority of cases and is
and antiapoptotic genes.14 mediated via increased cardiac insulin-like growth factor-1
(IGF-1) expression and activation of phophoinositide-3 ki-
Human Functional Data. On the functional level in humans, nase (PI3K) (p110␣).21 Transgenic mice with decreased
cardiac aging is associated with a significant increased wall cardiac PI3K activity (dnPI3K mice)22 and mice lacking the
stress, decreased elasticity, impaired early LV diastolic relax- p85 subunit of PI3K23 show a reduced basal cardiac size and
ation, increased end-systolic LV volume, and reduced con- an attenuation of exercise-induced myocardial hypertrophy.
tractile reserve.15 Arbab-Zadeh et al16 were able to show that Studies by Carè et al24 in exercised rats suggest that
lifelong physical activity prevents the development of dia- downregulation of cardiac-specific microRNAs (miRNA,
stolic LV dysfunction in older age. This finding also indicates miR-133, and miR-1) is regularly involved in exercise-
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
Gielen et al Cardiovascular Effects of Exercise Training 1223

induced cardiac hypertrophy. Reduction of miR-133 levels in approach to I/R protection to exercise is to differentiate
cardiac myocytes caused marked hypertrophy with increased between the mechanisms that are involved in I/R protection
protein synthesis and fetal gene expression. MiR-1 overex- and those that are essential. Mechanisms such as collateral
pression, on the other hand, significantly reduced protein formation,38 elevated HSP,39,40 increased myocardial cyclo-
synthesis and fetal gene expression.24 Its hypertrophic effects oxygenase-2 expression,41 and higher levels of endoplasmic
are probably mediated via RhoA, Cdc42, and Whsc2, whose reticulum stress proteins have all been shown to be not
RNA 3⬘ untranslated regions comprise flanking nucleotides
essential for exercise-mediated I/R protection.42 Therefore,
matching miR-133.
The role of miR-1 in physiological hypertrophy was we will focus on mechanisms found to be a condition sine qua
further elucidated by Elia et al,25 who documented that IGF-1 non for I/R protection. Among these are improved antioxida-
and IGF-1 receptor are targets of miR-1 and that miR-1 and tive protection, changes in mitochondrial metabolism and
IGF-1 are inversely correlated in cardiac hypertrophy.25IGF-1 protein expression, increased expression of sarcolemmal/
activation, on the other hand, inhibits the miR-1 transcription mitochondrial K⫹ATP channels, and attenuation of I/R-
factor Foxo3a via an AKT-dependent pathway, further am- induced calpain activation (Figure 2).
plifying its hypertrophic effects.
Using a transgenic mouse model with overexpression of a Antioxidative Protection. Production of ROS, accumulation
constitutively activated AKT (AKT-E40K), Catalucci et al26 of hydrogen ions, and generation of reactive nitrogen species
confirmed that IGF-1/AKT also improved LV contractility by play a major role in the complex pathophysiology of I/R
phosphorylation of phospholamban at threonine. By translo- injury,43,44 and the quantity of ROS production critically
cating to the sarcoplasmatic reticulum, activated AKT can depends on the duration of anoxia and reoxygenation.44 The
mediate improved calcium handling by disinhibition of the primary source of ROS in I/R situations seem to be the
sarcoplasmic reticulum Ca2⫹ ATPase pump (SERCA2a) mitochondria.45 In animal models, exercise training protects
through phospholamban phosphorylation. cardiac myocytes against I/R-induced oxidative stress and the
Cardiac plasticity in response to exercise training is
mitochondria against I/R-associated structural damage.46,47
surprisingly rapid. In a rat model of high-intensity interval
Several mechanisms for training-induced cardioprotection
treadmill running for 2, 4, 8, and 13 weeks followed by 2 or
4 weeks of detraining, Kemi and colleagues27 identified are under discussion (Figure 2). There is broad consensus that
cardiomyocyte cell length, diastolic relaxation, and calcium increased antioxidative protection by training-induced up-
decay as main factors for the training-induced increase in regulation of key antioxidative enzymes such as MnSOD,
VO2max by multiple regression analysis. These training- glutathione peroxidase, and catalase plays an important
related cardiac adaptations seem to be dependent on training role48 –50; however, myocardial MnSOD is only increased at
intensity.28 high training intensities. Recent studies have confirmed that
MnSOD is essential for full protection against I/R-induced
Cardiac Fatigue. Prolonged strenuous activity, as in the
Ironman Triathlon, can lead to transient reductions of LV myocardial infarction (ie, ischemia ⬎20 minutes) and related
systolic and diastolic function.29 Several mechanisms are ventricular arrhythmias; however, protection against myocar-
involved in cardiac fatigue, including changes in preload dial stunning seems to work even in the absence of MnSOD.51
conditions, myocardial stunning, ␤1-receptor desensitization,
Changes in Mitochondrial Metabolism and Protein Expres-
and altered cardiac autonomic regulation.30
Preload conditions were highlighted as a potential mech- sion. Cardiac mitochondria seem to contribute to exercise-
anism by Dawson et al, 31 who demonstrated the lack of induced protection against I/R injury in several ways, as
changes in LV function when central venous pressure was follows: (1) Exercise training results in reduced generation of
well maintained. However, other researchers with different ROS by cardiac mitochondria52,53; (2) mitochondria isolated
training protocols found depressed LV contractile function from the myocardium of exercised animals are more resistant
unrelated to preload conditions, which confirms the relevance to calcium-induced opening of the mitochondrial permeabil-
of intrinsic myocardial damage.32 In a dog model, Vatner and ity transition pore,52 a major proapoptotic stimulus; and (3)
Hittinger33 demonstrated that prolonged strenuous exercise exercise induces a downregulation of mitochondrial mono-
can indeed lead to ischemic myocardial dysfunction and amine oxidase-A,54 which seems to be a major source of
postischemic myocardial stunning in the absence of epicar- oxidative stress by H2O2 generation.55 Monoamine oxidase-A
dial coronary stenoses.
knockout animals are protected from I/R-induced myocardial
Prolonged strenuous exercise results in a 5-fold increase in
damage, highlighting the importance of this pathway for I/R
circulating catecholamines34 with consecutive desensitization
of ␤1-adrenoreceptors.35 This results in a blunted chrono- injury.55
tropic and inotropic response to dobutamine.32 Role of Sarcolemmal/Mitochondrial K⫹ATP Channels. Recently,
Prevention of Functional Decline After Exposure the sarcolemmal K⫹ATP channel has been described as a
to Ischemia potential mechanism for training-induced I/R protection.
I/R injury is a classic model of an acute cardiac injury with Opening the sarcolemmal K⫹ATP channel accelerates cardio-
great clinical relevance in the setting of myocardial infarction myocyte repolarization by increasing K⫹ outflow and short-
with interventional reperfusion therapy. Human epidemiolog- ens the action potential.56 As a result of the shorter action
ical data indicate that regular endurance exercise reduces the potential, Ca2⫹ overloading is prevented by reducing opening
risk of death during clinical I/R injury.36 rates of the L-type Ca2⫹ channel.56
Key Mechanisms for Protection Against I/R Injury. As Attenuation of I/R-Induced Calpain Activation. Calpain is a
summarized by Powers et al, 37 the problem of a mechanistic calcium-dependent protease with 2 isoforms (calpain I [milli]
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
1224 Circulation September 21, 2010

Figure 2. Exercise training has the potential to prevent myocardial damage related to I/R injury. Exercise training induces an upregulation of
key antioxidative enzymes such as MnSOD, glutathione peroxidase (GPX), and catalase. Opening the sarcolemmal (sarco) K⫹ATP channel
accelerates cardiomyocyte repolarization by increasing K⫹ outflow. As a result of the shorter action potential, Ca2⫹ overloading is prevented
by reducing opening rates of the L-type Ca2⫹ channel. Although not essential for I/R protection, increased levels of HSP72 were observed
after training. HSP72 can prevent apoptotic cell death, likely by interaction with caspase-3. Adapted from Powers et al 37 with kind permis-
sion of Springer Science and Business Media. Copyright © 2008, Springer Science and Business Media

and calpain II [micro]), which are activated by prolonged signed to assess the loss of cardioprotective effects after
exposure to elevated cytosolic calcium levels and contribute training cessation, Lennon et al62 were able to show a
to myocardial I/R injury.57 French and colleagues58 showed persistence of protection against myocardial stunning for up
that calpain inhibition attenuates I/R-induced contractile to 9 days, but cardioprotection was completely lost at 18 days
myocardial dysfunction. Exercise training reduces I/R- after exercise termination.
associated calpain activation, most likely by improved anti-
Systolic Heart Failure
oxidative protection and prevention of I/R-induced SERCA2a
Reduced exercise capacity and early fatigue in chronic heart
and phospholamban degradation.58
failure (CHF) are unrelated to the degree of LV systolic
Thresholds and Time Course of I/R Protection dysfunction but are determined primarily by peripheral alter-
ations with impaired endothelial function, reduced aerobic
Training Intensity. In a rat model of 40 minutes per day of oxidative metabolism in the skeletal muscle, skeletal muscle
treadmill exercise at 55% to 60% of VO2max for a total time atrophy, ventilatory dysfunction, and neurohumoral chang-
of 16 weeks, Starnes et al59 were unable to confirm a es.63 In the context of this review, we will focus on the
significant level of I/R protection. This study therefore cardiac effects of exercise only.
provided evidence for a threshold effect (ie, that a minimum
intensity and duration of exercise have to be exceeded to Training Effects on LV Function and Reverse Remodeling
benefit from I/R protection). Lennon et al,60 on the other In one of the first small prospective studies of aerobic
hand, compared 2 training protocols at 55% and 75% of endurance training in CHF patients (n⫽12), Sullivan et al64
VO2max for 60 minutes per day on 3 consecutive days and confirmed that 4 to 6 months of training did not worsen LV
did not find any difference in training-induced I/R protec- ejection fraction and tended to improve maximal cardiac
tion between both groups, indicating that there may not be output. The extent of the cardiac changes did not, however,
a linear dose-response relation between training intensity explain the large 23% improvement in peak oxygen uptake so
and cardioprotection. that peripheral changes in limb perfusion and oxidative
metabolism must account for the larger part of the beneficial
Training Duration. Surprisingly, the protective effect is symptomatic training effects.
similar after short-term (3 to 5 days) and long-term (weeks to The first larger prospective randomized study to actually
months) exercise training.40,46,61 In a trial specifically de- provide evidence for a training-induced reverse remodeling
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
Gielen et al Cardiovascular Effects of Exercise Training 1225

Figure 3. In systolic heart failure, endurance exercise can induce reverse remodeling with improvement of systolic and diastolic LV
function and reduction of LV end-diastolic diameters. It does so by interfering with the vicious cycle of CHF-associated skeletal muscle
hypoperfusion with increased local ROS and cytokine generation resulting in endothelial function and sympathetic activation, which fur-
ther increases afterload and worsens LV function. The reverse remodeling is a consequence of peripheral training effects (eg, improved
antioxidative protection in the skeletal muscle, increased parasympathetic tone, improved endothelial function as a result of increased
NO production and bioavailability, and direct cardiac effects related to improved Ca handling, increased PI3K [p110␣] activity, reduction
of fibrosis, and cardiomyocyte apoptosis). NE indicates norepinephrine; AT II, angiotensin II; ecSOD, extracellular SOD; GPX, glutathi-
one peroxidase; and ox. Phos, oxidative phosphorylation.

came from Hambrecht et al,65 who demonstrated that endur- tutive activation of PI3K (p110␣, caPI3K) improved survival
ance training led to reverse LV remodeling with modest by ⬇20%.22 In an aortic banding model of pathological
improvements of ejection fraction from 30% to 35% and hypertrophy, caPI3K mice showed a blunted increase in heart
reductions of LV end-diastolic diameter in a mixed popula- size compared with wild-type mice.22 Levels of interstitial
tion of subjects with ischemic and dilated cardiomyopathy. fibrosis were reduced in caPI3K mice, highlighting the
These findings were corroborated by the Exercise in Left potential of PI3K as an antifibrotic signal. Another important
Ventricular Dysfunction and Chronic Heart Failure study.66 A downstream effect of PI3K is the protection against apoptotic
recent study in our institution indicates that the reverse LV cardiomyocyte death via AKT.
remodeling occurs as early as 4 weeks after the initiation of
Anabolic/Catabolic Balance in the Myocardium. Animal
endurance training (unpublished data, 2010).
studies in which an left anterior descending artery ligation
Mechanisms Explaining Reverse Remodeling in CHF model was used demonstrated a significant upregulation of
In the absence of myocardial biopsies for molecular analysis components of the ubiquitin-proteasome system as well as
of myocardial changes induced by training, most authors myostatin.68,69 Both of these factors were significantly re-
interpreted this favorable training effect as secondary to duced by exercise training over a period of 4 weeks.68,69
afterload reduction with reduced resting blood pressure due to
improved endothelial function.65– 67 However, animal models Calcium Handling. Alterations in calcium handling are also
reveal that there are clear direct myocardial effects of training associated with pathological hypertrophy and transition from
that are related to signaling pathways of myocardial hyper- hypertrophy to failure: SERCA2a protein levels were reduced
trophy and fibrosis (Figure 3) in mouse and dog models of heart failure and were normal-
ized by exercise training.70,71 In addition, exercise training
Phosphoinositide-3 Kinase (p110␣). On the molecular basis, activates Ca2⫹/calmodulin-dependent protein kinase II, lead-
recent animal studies suggest that activation of PI3K (p110␣) ing to a hyperphosphorylation of phospholamban,72 which in
is involved in exercise-mediated cardioprotection: In mice its phosphorylated form no longer inhibits SERCA2a. In
with dilated cardiomyopathy, both swim training and consti- conjunction with an increased expression of Na⫹-Ca2⫹ ex-
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
1226 Circulation September 21, 2010

changer,73 this leads to improved calcium cycling and finally as shown by Yu et al.81 Although data on training effects in
to better cardiomyocyte function. For more detailed informa- patients with diabetes mellitus derived from small studies are
tion on exercise-induced improvements on the contractile still controversial,82,83 data on patients with established heart
apparatus and calcium cycling, please see a recent review by failure with normal ejection fraction are convincingly
Kemi and Wisloff.74 positive.84
CHF with systolic dysfunction is associated primarily with
Neurohormonal Adaptations. An aerobic training program in
significant diastolic dysfunction. Prospective randomized
patients with CHF regularly reduces the resting heart rate,
training studies confirmed that as early as after 4 weeks of
which indicates a reduction in sympathoadrenergic drive.
endurance training, diastolic dysfunction is significantly im-
This has also been confirmed for serum catecholamine levels:
proved with reduced E/E⬘ ratio (unpublished data, 2010).
Coats et al75 showed a 16% reduction of radiolabeled norepi-
nephrine secretion after 8 weeks of training. This reduction in Molecular Mechanisms
adrenergic tone was accompanied by an increase in heart rate In diastolic heart failure, calcium homeostasis is affected by
variability. In addition to the reduction in circulating cate- a reduced expression of SERCA2a,85 which is the key player
cholamines, Braith et al76 described a 25% to 30% reduction for calcium reuptake into the sarcoplasmic reticulum and is
of angiotensin II, aldosterone, arginine vasopeptide, and atrial regulated in its activity by phospholamban and sarcolipin and
natriuretic peptide after 4 months of walking training in increased diastolic calcium leakage via the ryanodine recep-
patients with CHF. tors.86,87 Phospholamban affects both the calcium affinity of
In a rat model of ischemic CHF, the beneficial training SERCA and its calcium reuptake rates. Animal studies in
effects on local neurohumoral balance were analyzed in the healthy rats indicate that endurance training can increase both
noninfarcted LV myocardium. Xu and colleagues77 found a SERCA and phospholamban expression,73 a finding that was
significant reduction of myocardial angiotensin-converting confirmed in a mouse model of sympathetic hyperactivity-
enzyme mRNA expression and AT1-receptor expression after induced heart failure (congenic ␣2A/␣2C-adrenoceptor knock-
8 weeks of treadmill training. This finding is of special out mice).88 No data are available with regard to training-
importance given the fact that ⬎90% of angiotensin II is associated effects on calcium leakage. On the structural level,
produced locally in the myocardium and implies that local 12 weeks of treadmill training in old Fischer 344 rats did not
angiotensin II levels are significantly reduced by training.
affect collagen volume fraction but significantly reduced
This reduction also translates into reduced fibrogenesis, as
collagen cross-links by advanced glycation end-products.89
indicated by reduced tissue inhibitor of metalloproteinase-1
expression with unchanged matrix metalloproteinase
(MMP)-1 expression and reduced collagen volume fraction in
Cardiac Valves
the exercised animals.77 Aortic Valve Sclerosis/Stenosis
Currently, no effective therapy to prevent calcified aortic
Diastolic Heart Failure valve disease is established. Calcified aortic valve disease
Heart failure with normal ejection fraction is a comparatively
confers significant morbidity and mortality as the severity of
new disease category that includes patients who display the
disease progresses.90
typical clinical, physiological, and neurohormonal heart fail-
Degenerative calcified aortic valve disease and atheroscle-
ure phenotype in the absence of reduced LV ejection frac-
tion.78 Thus far, no specific pharmaceutical therapy for heart rosis share similar mechanisms (ie, clinical risk factors and
failure with normal ejection fraction has been found. How- histopathological features). Pathological examinations of dis-
ever, a number of clinical and experimental studies indicate a eased aortic valves showed areas of subendothelial thickening
protective and curative role of exercise training for this with the disruption of the basement membrane, accumulation
disease entity. of inflammatory infiltrates, and calcium deposits.91,92 It is
well accepted that regular physical exercise prevents the
Protective Effects progression of atherosclerosis by modulating endothelial
It is well established in sports physiology that endurance function or oxidative stress.93,94 Using low-density lipopro-
training improves LV diastolic filling at rest and during tein receptor knockout mice, we recently demonstrated that
exercise.79 Endurance exercise interventions improve early regular exercise training as primary prevention prevents the
diastolic relaxation at all ages in healthy individuals, but in development of aortic valve sclerosis.95 It has been proposed
the presence of an elevated atrial filling rate in elderly that apoptosis of endothelial cells and myofibroblasts in the
patients, this could be normalized.80 A landmark study for the valve,96 elastin degradation and collagen synthesis by MMPs
preventive effects of lifelong physical activity against age- and/or cathepsin,97 increased oxidative stress,98 and enhanced
related diastolic dysfunction was published by Arbab-Zadeh osteogenesis99 contribute to aortic valve degeneration. In
et al in 2004.16 They were able to show with invasive consequence, procalcific gene expression such as bone mor-
measurement of LV pressure-volume relations that senior phogenetic protein 2 (BMP2), CBFA1/Runx2 (runt-related
athletes did not exhibit any degree of diastolic dysfunction
transcription factor 2), and osteocalcin was amplified.98 The
compared with young healthy sedentary controls.
activation of BMP2 and CBFA1/Runx2, particularly in the
Curative Effects noncalcified valve tissue, reflects an early stage of transdif-
Patients after myocardial infarction and with preexistent ferentiation of myofibroblast to a more osteoblast-like
abnormal relaxation patterns benefit from endurance training, phenotype.100
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
Gielen et al Cardiovascular Effects of Exercise Training 1227

In the low-density lipoprotein receptor knockout mouse metabolic control is dominant in small resistance arteries, in
model, regular exercise attenuated the deleterious elevation of which the myogenic response to increased perfusion pressure
oxidative stress and reduced expression of BMP2, Runx2, is also most pronounced. Although the effects of exercise on
and osteoblast differentiation marker. Additionally, the dis- the microcirculation and the capillary bed have been studied
ruption of the aortic valve endothelium was prevented.95 extensively, little is known about the training-induced mech-
anism of vascular changes in the venous circulation.
Vascular Effects of Exercise
“It has been said that one is as old as one’s arteries. In view Aorta
of the supreme importance of endothelium in arterial func- Although the aorta is a standard vessel to quantify changes in
tion, I should like to modify this statement by saying that one endothelial function in animal experiments involving small
is as old as one’s endothelium.”101 Perhaps Rudolf Altschul rodents, it is rarely a target vessel in human vascular research.
himself would be surprised to see to what extent his prophetic The composition of the extracellular matrix has changed
words have become scientific consensus half a century later. through the evolution of species and is influenced by pulse
There are several reasons why endothelial research became so pressure amplitude and wall tension. In fact, there seems to be
significant for today’s mechanistic concepts of exercise- a universal elastic modulus that determines the relative
mediated cardiovascular effects: (1) Endothelial dysfunction proportion of elastic lamellae, collagen fibers, and smooth
has been found to be a condition sine qua non for atherogen- muscle cells in the aortic wall.109 Aging, coronary artery
esis (ie, an obligatory initial step in early atherosclerosis).102 disease, and myocardial infarction are often accompanied by
Thus, prevention of endothelial dysfunction will necessarily increased aortic stiffness and reduced compliance.110,111
prevent atherosclerosis development. (2) The presence of Among healthy individuals, high-intensity exercise led to
endothelial dysfunction predicts future cardiovascular vascular remodeling with up to 51% enlarged brachial con-
events.103,104 Hence, endothelial function measurements were duit artery area and reduced distal aortic cross-sectional areas
proposed as a surrogate end point in clinical research.105 (3) (up to ⫺8%), whereas aortic distensibility remained un-
Endothelial function can be assessed by a large variety of changed.112 However, endurance and strength training have
methods in both animal and human studies. Surgical/invasive opposite effects on aortic compliance and vascular stiffness;
methods include organ bath measurements of arterial rings in an elegant study, Otsuki et al113 measured plasma
harvested in animals or during aortocoronary bypass surgery endothelin-1, nitric oxide (NO), and arterial stiffness in
in humans and catheter-based assessment of acetylcholine- young, healthy endurance-trained versus strength-trained
induced vasodilatation with angiography of the target vessel. men. They demonstrated that aortic pulse-wave velocity as an
Noninvasive methods such as brachial/radial ultrasound, established index of vascular stiffness was significantly
magnetic resonance imaging, and venous occlusion plethys- increased in strength athletes and reduced in endurance
mography permit serial studies comparing endothelial func- athletes versus healthy controls.113 Strength athletes dis-
tion before and after an exercise intervention. These classic played elevated endothelin-1 levels that correlated with aortic
methodologies are increasingly supplemented by commercial pulse-wave velocity. Reductions of aortic stiffness after
semiautomatic devices using, for example, digital pulse endurance training were confirmed in patients with hyperten-
amplitude tonometry to measure microvascular function.106 sion114 and coronary artery disease.115
It is beyond the scope of this article to provide in-depth
review of endothelial function in health and disease. We will Large Arterial Conduit Vessels
focus on 2 important aspects of vascular research in exercise Conduit vessels 2 to 4 mm in diameter are the preferred target
physiology: (1) the heterogeneity of molecular mechanisms vessels in clinical research because they may be assessed
involved in vasomotor function along the vascular bed and readily by either noninvasive of invasive methods. After the
(2) the molecular mechanisms for exercise-mediated im- first description of coronary endothelial dysfunction,116 it was
provements of vascular endothelial function with special quickly recognized that coronary endothelial function
emphasis on clinical studies in overt heart disease (coronary could serve as a stress index integrating the cardiovascular
artery disease and heart failure). effects of risk factors and may provide pivotal diagnostic
and prognostic information in patients with coronary heart
Differential Effects on Different Parts of the disease.103
Vascular Bed
The vasomotor responsiveness is not uniformly distributed Exercise and Conduit Vessel Vasomotor Function in
along the arterial tree. Four basic mechanisms have been Coronary Artery Disease
identified to regulate vascular tone in response to local The clinical effects of endurance exercise training on coro-
metabolic demand in the downstream vascular territory: (1) nary endothelial function were first studied in humans by
endothelium-mediated flow-induced vasodilatation, (2) myo- Hambrecht et al117: A 4-week endurance training program
genic control, (3) metabolic vasodilatation, and (4) sympa- was effective in attenuating the paradoxical arterial vasocon-
thetic control.107,108 The relative contribution of these mech- striction in epicardial conduit vessels by ⫺54% and increased
anisms to vasodilatation is different in different average peak flow velocity by ⫹78% in response to intra-
microdomains of the coronary vascular tree. Endothelium- coronary acetylcholine infusion. The improvements in coro-
dependent vasodilatation is most relevant in small conduit nary endothelial function were partially lost during a consec-
and resistance arteries down to 150 ␮m of diameter108; utive 5-month home-based endurance training program at
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
1228 Circulation September 21, 2010

Figure 4. The majority of exercise effects on the vascular endothelium are mediated by intermittent increases of laminar shear stress
associated with increased cardiac output during physical exertion. On the luminal side of the endothelial cells, direct signaling can
occur through deformation of the glycocalyx. This shear stress–induced signal can activate calcium ion channels, phospholipase activ-
ity leading to calcium signaling, prostaglandin I2 (PGI2) release, and cAMP-mediated smooth muscle cell relaxation. VEGF receptor 2
(VEGFR2) located at the luminal surface can associate with vascular endothelial cadherin, [beta]-catenin, and PI3K to phosphorylate Akt
and induce AKT-mediated eNOS phosphorylation, which leads to higher NO production. Mechanical forces are also transmitted by the
cytoskeleton to adhesion sites, where transmembrane integrins bound to the extracellular matrix serve as a focus for deformation. This
integrin-mediated pathway leads to the activation of Ras–mitogen-activated protein kinase activity. In turn, Ras releases nuclear
factor-␬B (NF␬B) from its cytosolic inhibitor and thus enables its translocation to the nucleus, where it binds to the promoters of multi-
ple target genes, including the eNOS gene. Increased NO synthesis in response to shear stress induces extracellular SOD (ecSOD) in a
feed-forward fashion to inhibit the degradation of NO by ROS with consecutive formation of the toxic peroxynitrite. Akt indicates pro-
tein kinase B; PECAM-1, platelet endothelial cell adhesion molecule-1; Ras, small GTPase; ONOO⫺, peroxynitrite; and AA, Arachidonic
acid. Reprinted from Davies et al 121 with permission of the publisher. Copyright ©2008, Cambridge University Press.

lower intensity, and changes were related to daily training frictional forces caused by antegrade blood flow. Whereas
durations.118 increased radial strain (eg, as a result of arterial hypertension)
In a subsequent study examining training effects on the increases the atherogenic risk, laminar shear stress as a result
peripheral conduit artery function, Gokce et al119 measured of pulsatile continuous blood flow is a potent survival signal
brachial and femoral artery endothelial function in patients for endothelial cells.
with coronary artery disease before and after a 10-week leg On the molecular level, laminar shear stress affects multi-
exercise program. They documented significant improvement ple signaling pathways121 (Figure 4), which include the PI3K,
in endothelium-dependent, flow-mediated dilation in conduit extracellular signal-regulated kinase 5 (also known as
arteries of the leg but not the arm and hypothesized that mitogen-activated protein kinase 7), and NO pathways.122
training effects could be limited to the trained limb. This may, Endothelial cells must therefore express specific mechano-
however, also depend on the distribution of endothelial transducers that convert physical stress into biochemical
dysfunction and the intensity of the training program because signals. The cytoskeleton plays an important part in the
Linke et al120 were able to confirm improved radial artery sensing of shear stress, and high strain is observed at the
endothelial function after pure bicycle ergometer training in luminal and basal membrane. In addition, cellular adhesion
patients with CHF. proteins such as platelet endothelial cell adhesion molecule-1,
Mechanosensors in the Vascular Wall vascular endothelial cadherin, and the transmembrane ty-
Arteries are exposed to 2 main forces: radial strain as a result rosine kinase vascular endothelial growth factor receptor-2
of the blood pressure wave that is propagated along the (⫽KDR) were found to mediate biochemical responses to
arterial tree and laminar shear stress as a result of the flow. Vascular endothelial growth factor receptor-2 activates
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
Gielen et al Cardiovascular Effects of Exercise Training 1229

PI3K, which is essential for AKT-mediated phosphorylation ing programs resulted in an increased vascular expression
and activation of endothelial NO synthase (eNOS).123 In of antioxidative enzymes, such as SOD, catalase, and
contrast to laminar shear stress, the atherogenic flow patterns glutathione peroxidase133–135 and a reduced expression of
include turbulent or disturbed flow, low flow, gradients, and ROS-generating enzymes such as nicotinamide adenine
flow reversal. These conditions are associated with high rates dinucleotide phosphate (NAD[P]H) oxidase and xanthine
of endothelial cell proliferation and apoptosis, increased oxidase.94,136 Nicotinamide adenine dinucleotide phos-
production of ROS, and increased expression of inflamma- phate (NAD[P]H) oxidase expression and activity are
tory markers inducing a preatherosclerotic vascular pheno- regulated mainly by the dominant influence of angiotensin
type.122 In the presence of additional atherogenic risk factors, II type 1 receptor activation by inducing a rapid rac1
such as hypertension, dyslipidemia, and diabetes mellitus, translocation to the cell membrane.137 In addition, a
atherogenesis is significantly accelerated. 4-week exercise training program in patients with coronary
artery disease resulted in a significantly lower expression
NO Generation in the Endothelium of angiotensin II type 1 receptor compared with a seden-
A key component of intact endothelial function is a functional
tary control group.94
eNOS. Located in the luminal endothelial cell membrane, it
produces NO from L-arginine. By diffusion, the short-lived Exercise and EPCs.
NO reaches the vascular smooth muscle cells in the media The first description of postnatal vasculogenesis and the
and causes relaxation via cyclic guanosine monophosphate- characterization of EPCs derived from adult bone marrow
dependent pathways. Potential mechanisms of endothelial changed our concept of vascular plasticity in health and
dysfunction are (1) reduced eNOS substrate and cofactor disease.138 In addition to EPCs, mesenchymal stem cells also
availability (eg, reduction of L-arginine levels, increase of the possess the potential for vascular regeneration.139 Both mes-
competitive eNOS inhibitor asymmetrical dimethylarginine enchymal stem cells and EPCs are mobilized from the bone
[ADMA])124; (2) a reduced quantity and activity of eNOS and marrow in response to ischemia, exercise, and neurohormonal
inactivation of NO by ROS125; and (3) vascular regeneration factors (eg, vascular endothelial growth factor [VEGF],
by endothelial progenitor cells (EPCs).126 placental growth factor) and critically contribute to maintain-
ing the integrity of the endothelial cell layer. EPC number and
Exercise and eNOS Substrate/Cofactor Availability. Another
function correlate with the number of cardiovascular risk
enzyme also responsible for the generation of ROS in the
factors and disease severity and predict cardiovascular events
vascular is NO synthase. Under a number of pathological
and death from cardiovascular causes.140
conditions, the enzymatic reduction of molecular oxygen by
It is a matter of continuing debate whether training-induced
eNOS is no longer coupled to L-arginine oxidation, resulting
flow changes or ischemia induction is required to stimulate
in production of superoxide rather than NO.127 A number of
EPC release. In 2 clinical studies, we demonstrated that an
mechanisms have been reported to contribute to eNOS
increase in circulating EPCs was only achieved in response to
uncoupling, including tetrahydrobiopterin (BH4) deficiency,
exercise-induced ischemia, whereas matrigel assays indicated
shortage of L-arginine or HSP90, eNOS dephosphorylation on
improved EPC function in nonischemic training, as
threonine residue 495, or elevated ADMA levels.127 With
well.141,142 Other authors showed exercise-related EPC in-
respect to the BH4 concentration, cell culture studies provided
crease also in the absence of ischemia.143,144 Prolonged
the first evidence that increased shear stress dramatically
strenuous exercise (half-marathon, marathon, or spartathlon),
increases BH4 levels.128 This result was confirmed in an
however, showed no change or a significant increase in
aortocaval fistula model in the rat, wherein aortic blood flow
circulating progenitor cells.145–148
is enhanced proximal but decreased distal to the fistula,129 as
The principal mechanism of EPC mobilization from the
well as in a rat exercise training model.130 In addition, for
bone marrow seems to depend on the activation of eNOS in
circulating levels of ADMA, a beneficial effect of exercise
the presence of several mobilizing factors such as VEGF149 or
training was documented.131
placental growth factor.150 Gene-targeting studies using either
Exercise, eNOS Activity, and Oxidative Stress. To assess MMP-2151 or MMP-9152 knockout mice demonstrated that the
molecular changes in NO production in human arterial presence of MMPs is crucial in ischemia-induced mobiliza-
conduit vessels, we randomized patients with coronary artery tion of EPCs and hence neovascularization. At the molecular/
disease scheduled for elective coronary artery bypass graft cellular level, the following scenario for the mobilization is
surgery to 4 weeks of endurance training or control and proposed (Figure 5): Under steady state conditions, progeni-
harvested left internal mammary artery tissue not needed for tor cells reside in a niche of the bone marrow, bound to
bypass grafting.132 Patients in the training group had 2-fold stroma cells via adhesion molecules such as vascular cell
higher eNOS protein expression and 4-fold higher eNOS adhesion molecule/very late antigen-4.153 Signal-induced up-
Ser1177 phosphorylation levels in left internal mammary regulation of MMP-9 results in a release of sKitL, conferring
artery endothelium. A linear correlation was confirmed be- signals that enhance mobility of progenitor cells into a
tween Akt phosphorylation and phospho-eNOS levels and vascular-enriched niche favoring liberalization of the cells
between phospho-eNOS and Delta mean peak blood flow into the circulation.154 How can we explain the exercise-
velocity. induced mobilization of EPCs? In several studies, it was
NO bioavailability is also influenced by ROS-mediated demonstrated that exercise increases the concentration of
breakdown. In animal and human studies, exercise train- NO,132 which in turn can activate MMP-9 in bone marrow,155
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
1230 Circulation September 21, 2010

Figure 5. A model for mobilization of circulating progenitor cells from the bone marrow by increased shear stress induced by exercise.
Increased laminar shear stress results in an activation of eNOS, resulting in an increase of NO. Subsequently, MMP-9 and MMP-2 are
activated, resulting in the release of sKitL. SKitL confers signals enhancing mobility of circulating progenitor cells. Along with stromal-
derived factor-1␣ (SDF-1␣ gradient, the circulating progenitor cells are mobilized into the peripheral circulation. CPC indicates circulat-
ing progenitor cells; VCAM-1, vascular cell adhesion molecule-1; and VLA4, very late antigen-4.

leading to enhanced mobilization of progenitor cells, as accurate technique, which also permits insight into changes of
depicted above. As soon as the EPCs are circulating, the most plaque composition.
important factors for tissue engraftment of the mobilized cells
Arterial Resistance Vessels
are the local concentration of stromal-derived factor-1␣ and
Larger and medium-sized resistance vessels (⬎150 ␮m) are
its cell receptor CXCR4.156 This notion is further supported exposed to the predominant influence of endothelium-
by the observation that mice lacking CXCR4 die in utero mediated vasodilatation, whereas smaller arterioles are sub-
because of defects in vascular development.157 Although the ject to metabolic and myogenic control. The vasomotor state
animal data make shear stress–induced NO generation a of the distal microvasculature is influenced by the dominant
likely mechanism for NO-mediated EPC mobilization, animal effects of local myocardial metabolic demands.
experiments with ischemic exercise are still lacking. There-
fore, the aforementioned controversy still awaits experimen- Metabolic Control
tal resolution. In principle, all concepts for metabolic control of resistance
vessel diameter are based on the presence of an oxygen/met-
Exercise and Plaque Regression in CAD abolic sensor coupled to vascular smooth muscle cell tension
Although it is high on the agenda in lipid research, plaque to control vascular tone.162 Four models are currently under
regression ceased to be a research focus of exercise research discussion, as follows:
in recent years. Two decades ago, a series of angiographic
long-term follow-up studies documented that regular endur- 1. An oxygen sensor in the vessel wall leads to production
ance exercise training can retard the progression of coronary of the vasodilatory prostaglandin G2 in response to
atherosclerosis158,159 or even reduce stenosis diameter if decreases in PO2.163 There is evidence for prostaglan-
din-mediated vasodilation via KATP channels.164
combined with lipid-management techniques of smoking
2. Adenosine serves as a myocardial oxygen sensor be-
cessation and other lifestyle interventions.160,161 Surprisingly, cause it is generated at an accelerated rate when oxygen
not a single study has addressed exercise-mediated changes in supply-to-demand relationship falls.165
plaque volume by intravascular ultrasound thus far to control 3. Carbon dioxide production, which in turn leads to tissue
the findings of the early regression studies with a more acidosis, is known to increase coronary flow.166 Acido-
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
Gielen et al Cardiovascular Effects of Exercise Training 1231

Figure 6. In the microcirculation, several pathways for metabolic vasodilation have been described. PO2-dependent regulation repre-
sents hypoxic coronary flow control; metabolic rate of oxygen (MRO2)– dependent regulation represents normoxic coronary flow con-
trol. Under hypoxemic conditions, Ca2⫹i is increased in endothelial cells, leading to phospholipase activation with consecutive genera-
tion of prostaglandins and cAMP-mediated metabolic vasodilation. Alternatively, ATP release either from erythrocytes or from the
working skeletal muscle activates the adenosine purinergic A2A receptor, resulting in activation of the KATP channel with potassium loss
and hyperpolarization of the vascular smooth muscle cell. As a consequence, voltage-gated Ca2⫹ channels (VGCC) are opened, and
Ca2⫹i is decreased with vascular relexation and vasodilation. Acidosis can directly influence the opening probability of the KATP chan-
nel. Exercise increases the adenosine sensitivity, resulting in larger metabolic vasorelaxation. Hb indicates hemoglobin; AC, adenylate
cyclase; and PGI2, prostaglandin I2. Adapted from Deussen et al 162 with kind permission of Springer Science and Business Media.
Copyright ©2006, Springer Science and Business Media

sis in turn increases the sensitivity of adenosine A2A Myogenic Control


receptors. It is well established in porcine animal models of treadmill
4. Erythrocyte ATP release may cause vasodilatation, as training that myogenic vasoconstriction to increases in per-
documented in studies confirming the necessity of fusion pressure (particularly ⬎40 mm Hg) is augmented after
erythrocytes to mediate vasodilation under hypoxic exercise.170 Evidence is accumulating that L-type voltage-
conditions.167 gated calcium channels and protein kinase C determine
myogenic tone under physiological conditions.171 Korzick et
All vasodilatory stimuli converge on the activation of the al172 demonstrated that increased myogenic tone in trained
KATP channels, the opening of which leads to vascular pigs involves changes in protein kinase C-␣ expression and
smooth muscle cell hyperpolarization and reduced activation phosphorylation, which augment the depolarization-related
of voltage-gated calcium channels (Figure 6). opening of voltage-gated calcium channels in smooth muscle
Exercise training increases resistance vessel sensitivity and cells and increase of intracellular calcium.
maximal responsiveness to adenosine. This has been con-
Angiogenesis and Microcirculation
firmed in dogs and miniature swine in vivo.168,169 In humans, In the cardiac muscle, White et al173 conclusively showed in
adenosine-induced microvascular function improved by 29% a porcine model of exercise training that training increases
after exercise training, as assessed by coronary flow reserve the total vascular bed cross-sectional area by up to 37% after
ratio in response to intracoronary adenosine infusion. With 16 weeks. In humans, changes in cardiac vascularization are
regard to the different mechanisms described above, no data difficult to assess.
are currently available to describe exercise-mediated specific More data, however, are available for the skeletal muscle.
changes. In addition to an effect on metabolic alterations and the
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
1232 Circulation September 21, 2010

Figure 7. A model depicting exercise-


induced angiogenesis in the peripheral
skeletal muscle. A central player for
enhanced capillary density, VEGF can be
activated either by PGC-1␣ or by hypox-
ia-inducible transcription factor 1␣ (HIF-
1␣) because of local hypoxia or a reduc-
tion in energy equivalents. TF indicates
transcription factor.

catabolic/anabolic balance within the skeletal muscle, exer- protein kinase.181 However, if this pathway is only responsi-
cise also affects the vascularization of the skeletal muscle. It ble for basal VEGF expression and capillarization and not for
has been known for a long time that endurance exercise exercise-induced angiogenesis is still an ongoing discus-
training induces a significant increase in muscle capillaries in sion.182 In addition to activation of AMPK, the activation of
either animals or humans.174,175 This process is largely the p38MAPK (Mitogen-activated protein kinase) pathway
coordinated by soluble factors emanating from tissues in need by exercise training also leads to the activation of PGC-1␣.183
of vascularization. One of the best factors to study in this Last but not least, the increased production of ROS by
scenario is VEGF. It is a powerful activator of endothelial exercise training can also contribute to the induction of
proliferation and is crucial for nearly all forms of neovascu- PGC-1␣.184 Therefore, one may summarize that endurance
larization.176 With respect to exercise training, several studies exercise by activating PGC-1␣ via AMPK or p38MAPK or
showed that VEGF transcription in skeletal muscle is in- ROS leads to an enhanced transcription of VEGF, finally
creased by short-term177 as well as endurance training.142 It is resulting in angiogenesis (Figure 7).
thought that local skeletal muscle hypoxia stabilizes the
Venous Circulation
transcription factor hypoxia inducible factor-1␣, leading to Compared with the arterial bed, the venous circulation has
the induction of VEGF. However, this concept must be received less attention in vascular research, particularly with
questioned because the deletion of hypoxia inducible regard to training effects. The reasons are diverse, ranging
factor-1␣ in the skeletal muscle increases rather than de- from methodological problems to underestimation of the
creases capillary density.178 physiological relevance of venous function.185
In recent years, the transcriptional coactivator peroxisome Key parameters of venous function such as venous capac-
proliferator-activated receptor-␥ coactivator (PGC)-1␣ has itance, which reflects the blood volume stored in the venous
been regarded as an important exercise-mediated regulator of system, and venous compliance, which represents the change
VEGF transcription. Mice lacking PGC-1␣ in their skeletal in volume in response to a change in pressure, change with
muscle failed to increase capillary density in response to aging, posture, and physical activity. Aging alters the com-
exercise training.179 The central role of PGC-1␣ is further- position of the venous wall, resulting in lower venous
more supported by the observation that transgenic expression compliance in sedentary senior individuals.186 In both young
of PGC-1␣ in skeletal muscle dramatically increases capillary and elderly endurance-trained men, however, venous compli-
density.180 The mechanism by which PGC-1␣ induces VEGF ance was 70% to 120% higher compared with their sedentary
transcription appears to involve the coactivation of the orphan age-matched counterparts. It is currently unclear whether
nuclear receptor ERR␣.180 Which signals are upstream of these changes are primarily the result of factors such as
PGC-1␣ mediating the exercise-induced angiogenesis? One increased venous NO availability and decreased venous
pathway is the exercise-dependent modulation of high-energy smooth muscle tone, sympathetic ␣-adrenergic vasomotor
phosphates and the concomitant activation of AMP-activated control, or structural effects on elastin-to-collagen ratio.
Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015
Gielen et al Cardiovascular Effects of Exercise Training 1233

Immobilization, as tested in bed rest studies, demonstrated nation for the discrepant results can be derived from 2 studies
a significant decline in venous capacitance after 52 days, by Johnson et al.200,201 Whereas long-term training enhanced
which was not affected by resistive vibration exercise.187 endothelium-dependent vasorelaxation in pulmonary arteries
Venous compliance remained unaffected. by mechanisms of increased reliance on NO and reduced
In patients with heart failure, exercise forearm venous production of a prostanoid constrictor in miniature swine with
capacitance and venous outflow, as measured by strain-gauge surgically induced chronic coronary artery occlusion, it did
plethysmography, are significantly reduced and seem to be not alter pulmonary vasorelaxation in normal pigs.200 How-
related to maximal walking distance.188 Although the meth- ever, short-term 1-week training was effective in improving
odology of strain-gauge plethysmography was used in a bradykinin-induced vasorelaxation and in increasing pulmo-
number of studies to examine training effects on vascular nary artery eNOS protein expression. SOD remained un-
function,189,190 changes of venous capacitance induced by changed.201 In essence, training-induced changes in pulmo-
training have not been reported thus far. From animal nary vasomotion are modified by preexistent endothelial
experiments, we know that the capillary domain area values dysfunction and by training duration.
for venular capillaries were significantly decreased after 4 In a rat model, Handoko et al202 induced compensated and
weeks of training.191 progressive pulmonary hypertension using different doses of
monocrotaline. Only the compensated pulmonary hyperten-
Pulmonary Circulation sion animals benefited from exercise training with increased
Pulmonary hypertension is a frequent accompanying problem
exercise tolerance and right ventricular capillarization,
in heart failure or valvular heart disease. Similar to heart
whereas animals with progressive right ventricular failure had
failure, exercise has long been regarded as detrimental and
reduced cardiac output and died prematurely from right
was discouraged in pulmonary hypertension because of the ventricular failure.
fear of fatal cardiovascular compromise.

Human Data Conclusion


The first reports on training effects on pulmonary pressure To summarize, cardiac effects of exercise include an im-
came from aerobic training studies in patients with stable proved protection against I/R injury primarily as a result of
CHF. Lee et al192 were the first to demonstrate the lack of any higher antioxidative protection and improved LV diastolic
exercise-induced rise in resting pulmonary artery pressure in and systolic function in chronic heart failure due to favorable
patients with CHF, a finding confirmed by Dubach et al193 18 changes in neurohormonal state, activation of PI3K (p110␣)
years later. In a large prospective randomized study involving attenuating pathological hypertrophy, normalization of car-
73 patients with CHF, Hambrecht et al65 were finally able to diomyocyte calcium handling, and inhibition of the catabolic
demonstrate that pulmonary vascular resistance at rest and at ubiquitin-proteasome system.
peak exercise was decreased after 6 months of endurance Vascular effects are based on improved endothelium-
training. mediated flow-induced vasodilatation in conduit arteries and
Just recently, the first clinical study in patients with stable larger resistance arteries, higher myogenic control, and in-
precapillary pulmonary hypertension confirmed that 15 creased metabolic vasodilatation in small resistance arteries.
weeks of endurance exercise are effective in significantly Vascular regeneration by mobilization of endothelial progen-
improving 6-minute walking distance on top of optimal itor cells is augmented by exercise. Recently, improved
medical therapy.194 endothelial vasodilatation has also been confirmed in the
pulmonary artery.
Animal Experiments Extending our concept of molecular mechanisms of exer-
The first rat model of pulmonary hypertension to be used with cise is essential to further optimize training interventions with
exercise training was hypobaric hypoxia. Rats were trained regard to their clinical efficacy and to identify novel targets
for 4 and 6 weeks under hypobaric hypoxia (equivalent to for pharmaceutical intervention. However, certain areas (ie,
altitudes of 2500 and 5500 m). Kashimura et al196 found that microcirculation and the venous system) are still incom-
the increase in resting mean pulmonary arterial pressure with pletely understood and merit further molecular studies. Re-
increasing equivalent altitude was lower in the 2 trained sidual incomplete molecular understanding should not pre-
groups than in the control group and greater after 6 than after vent the clinical use of training interventions with established
4 weeks of exercise.195 In a follow-up study, they showed that clinical benefit.
6 weeks of training can attenuate the hypoxia-induced and
angiotensin II–induced PH. These findings were extended by Sources of Funding
Favret et al,197 who demonstrated that exercise training Dr Gielen is supported by the Deutsche Forschungsgemeinschaft,
improved lung gas exchange and attenuated acute hypoxic grant GI535/1–1.
pulmonary hypertension but did not prevent pulmonary hy-
pertension of prolonged hypoxia. Disclosures
In rabbits, it was soon documented that treadmill exercise None.
improved acetylcholine-induced endothelium-dependent va-
sodilatation not only in aortic rings but also in precontracted
References
1. Booth F, Laye M, Lees S, Rector R, Thyfault J. Reduced physical
pulmonary artery rings,198 a finding that could not be repro- activity and risk of chronic disease: the biology behind the conse-
duced in Sprague-Dawley rats, however.199 A possible expla- quences. Eur J Appl Physiol. 2008;102:381–390.

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015


1234 Circulation September 21, 2010

2. Mokdad AH, Giles WH, Bowman BA, Mensah GA, Ford ES, Smith 25. Elia L, Contu R, Quintavalle M, Varrone F, Chimenti C, Russo MA,
SM, Marks JS. Changes in health behaviors among older Americans, Cimino V, De Marinis L, Frustaci A, Catalucci D, Condorelli G.
1990 to 2000. Public Health Rep. 2004;119:356 –361. Reciprocal regulation of microRNA-1 and insulin-like growth factor-1
3. Perhonen MA, Franco F, Lane LD, Buckey JC, Blomqvist CG, Zerwekh signal transduction cascade in cardiac and skeletal muscle in physiolog-
JE, Peshock RM, Weatherall PT, Levine BD. Cardiac atrophy after bed ical and pathological conditions. Circulation. 2009;120:2377–2385.
rest and spaceflight. J Appl Physiol. 2001;91:645– 653. 26. Catalucci D, Latronico MVG, Ceci M, Rusconi F, Young HS, Gallo P,
4. Dorfman TA, Levine BD, Tillery T, Peshock RM, Hastings JL, Santonastasi M, Bellacosa A, Brown JH, Condorelli G. Akt increases
Schneider SM, Macias BR, Biolo G, Hargens AR. Cardiac atrophy in sarcoplasmic reticulum Ca2⫹ cycling by direct phosphorylation of
women following bed rest. J Appl Physiol. 2007;103:8 –16. phospholamban at Thr17. J Biol Chem. 2009;284:28180 –28187.
5. Sipola P, Heikkinen J, Laaksonen DE, Kettunen R. Influence of 12 27. Kemi OJ, Haram PM, Wisloff U, Ellingsen O. Aerobic fitness is asso-
weeks of jogging on magnetic resonance-determined left ventricular ciated with cardiomyocyte contractile capacity and endothelial function
characteristics in previously sedentary subjects free of cardiovascular in exercise training and detraining. Circulation. 2004;109:2897–2904.
disease. Am J Cardiol. 2009;103:567–571. 28. Kemi OJ, Haram PM, Loennechen JP, Osnes JB, Skomedal T, Wisloff
6. Navarro-Arevalo A, Canavate C, Sanchez-del-Pino M. Myocardial and U, Ellingsen O. Moderate vs. high exercise intensity: differential effects
skeletal muscle aging and changes in oxidative stress in relationship to on aerobic fitness, cardiomyocyte contractility, and endothelial function.
rigorous exercise training. Mech Ageing Dev. 1999;108:207–217. Cardiovasc Res. 2005;67:161–172.
7. Rinaldi B, Corbi G, Boccuti S, Filippelli W, Rengo G, Leosco D, Rossi 29. Whyte GP, George K, Sharma S, Lumley S, Gates P, Prasad K,
F, Filippelli A, Ferrara N. Exercise training affects age-induced changes McKenna WJ. Cardiac fatigue following prolonged endurance exercise
in SOD and heat shock protein expression in rat heart. Exp Gerontol. of differing distances. Med Sci Sports Exerc. 2000;32:1067–1072.
2006;41:764 –770. 30. Scott J, Warburton DER. Mechanisms underpinning exercise-induced
8. Devi SA, Prathima S, Subramanyam MVV. Dietary vitamin E and changes in left ventricular function. Med Sci Sports Exerc. 2008;40:
physical exercise, II: antioxidant status and lipofuscin-like substances in 1400 –1407.
aging rat heart. Exp Gerontol. 2003;38:291–297. 31. Dawson EA, Shave R, Whyte G, Ball D, Selmer C, Jans O, Secher NH,
9. Martin JL, Mestril R, Hilal-Dandan R, Brunton LL, Dillmann WH. George KP. Preload maintenance and the left ventricular response to
Small heat shock proteins and protection against ischemic injury in prolonged exercise in men. Exp Physiol. 2007;92:383–390.
cardiac myocytes. Circulation. 1997;96:4343– 4348. 32. Welsh RC, Warburton DER, Humen DP, Taylor DA, McGavock J,
10. Starnes JW, Choilawala AM, Taylor RP, Nelson MJ, Delp MD. Myo- Haykowsky MJ. Prolonged strenuous exercise alters the cardiovascular
cardial heat shock protein 70 expression in young and old rats after response to dobutamine stimulation in male athletes. J Physiol. 2005;
identical exercise programs. J Gerontol A Biol Sci Med Sci. 2005;60: 569:325–330.
963–969. 33. Vatner SF, Hittinger L. Coronary vascular mechanisms involved in
11. Olivetti G, Melissari M, Capasso JM, Anversa P. Cardiomyopathy of the decompensation from hypertrophy to heart failure. J Am Coll Cardiol.
aging human heart: myocyte loss and reactive cellular hypertrophy. Circ
1993;22:34A– 40A.
Res. 1991;68:1560 –1568.
34. Marrero MB, Horwath SM, Wilkerson JE. Acute blood biochemical
12. Phaneuf S, Leeuwenburgh C. Cytochrome c release from mitochondria
alterations in response to marathon running. Eur J Appl Physiol Occup
in the aging heart: a possible mechanism for apoptosis with age. Am J
Physiol. 1975;34:173–181.
Physiol. 2002;282:R423–R430.
35. Friedman DB, Ordway GA, Williams RS. Exercise-induced functional
13. Sussman MA, Anversa P. Myocardial aging and senescence: where have
desensitization of canine cardiac beta-adrenergic receptors. J Appl
the stem cells gone? Annu Rev Physiol. 2004;66:29 – 48.
Physiol. 1987;62:1721–1723.
14. Kwak HB, Song W, Lawler JM. Exercise training attenuates age-
36. Reeve JL, Duffy AM, O⬘Brien T, Samali A. Don’t lose heart: thera-
induced elevation in Bax/Bcl-2 ratio, apoptosis, and remodeling in the
peutic value of apoptosis prevention in the treatment of cardiovascular
rat heart. FASEB J. 2006;20:791–793.
disease. J Cell Mol Med. 2005;9:609 – 622.
15. Pugh KG, Wei JY. Clinical implications of physiological changes in the
37. Powers SK, Quindry JC, Kavazis AN. Exercise-induced cardiopro-
aging heart. Drugs Aging. 2001;18:263–276.
tection against myocardial ischemia-reperfusion injury. Free Radic Biol
16. Arbab-Zadeh A, Dijk E, Prasad A, Fu Q, Torres P, Zhang R, Thomas JD,
Palmer D, Levine BD. Effect of aging and physical activity on left Med. 2008;44:193–201.
ventricular compliance. Circulation. 2004;110:1799 –1805. 38. Laughlin MH, Oltman CL, Bowles DK. Exercise training-induced adap-
17. Pluim BM, Zwinderman AH, van der Laarse A, van der Wall EE. The tations in the coronary circulation. Med Sci Sports Exerc. 1998;30:
athlete’s heart: a meta-analysis of cardiac structure and function. 352–360.
Circulation. 2000;101:336 –344. 39. Quindry JC, Hamilton KL, French JP, Lee Y, Murlasits Z, Tumer N,
18. Pelliccia A, Culasso F, Di Paolo FM, Maron BJ. Physiologic left ven- Powers SK. Exercise-induced HSP-72 elevation and cardioprotection
tricular cavity dilatation in elite athletes. Ann Intern Med. 1999;130: against infarct and apoptosis. J Appl Physiol. 2007;103:1056 –1062.
23–31. 40. Hamilton KL, Powers SK, Sugiura T, Kim S, Lennon S, Tumer N,
19. Fagard R. Athlete’s heart. Heart. 2003;89:1455–1461. Mehta JL. Short-term exercise training can improve myocardial tol-
20. Maron BJ. Distinguishing hypertrophic cardiomyopathy from athlete’s erance to I/R without elevation in heat shock proteins. Am J Physiol.
heart physiological remodelling: clinical significance, diagnostic 2001;281:H1346 –H1352.
strategies and implications for preparticipation screening. Br J Sports 41. Quindry J, French J, Hamilton K, Lee Y, Selsby J, Powers S. Exercise
Med. 2009;43:649 – 656. does not increase cyclooxygenase-2 myocardial levels in young or
21. Neri Serneri GG, Boddi M, Modesti PA, Cecioni I, Coppo M, Padeletti senescent hearts. J Physiol Sci. 2010;60:181–186.
L, Michelucci A, Colella A, Galanti G. Increased cardiac sympathetic 42. Murlatsis Z, Lee Y, Powers SK. Short-term exercise does not increase
activity and insulin-like growth factor-I formation are associated with ER stress protein expression in cardiac muscle. Med Sci Sports Exerc.
physiological hypertrophy in athletes. Circ Res. 2001;89:977–982. 2007;39:1522–1528.
22. McMullen JR, Amirahmadi F, Woodcock EA, Schinke-Braun M, 43. Zweier JL, Talukder MAH. The role of oxidants and free radicals in
Bouwman RD, Hewitt KA, Mollica JP, Zhang L, Zhang Y, Shioi T, reperfusion injury. Cardiovasc Res. 2006;70:181–190.
Buerger A, Izumo S, Jay PY, Jennings GL. Protective effects of exercise 44. Li C, Jackson RM. Reactive species mechanisms of cellular hypoxia-
and phosphoinositide 3-kinase(p110⫹) signaling in dilated and hyper- reoxygenation injury. Am J Physiol. 2002;282:C227–C241.
trophic cardiomyopathy. Proc Natl Acad Sci U S A. 2007;104:612– 617. 45. Adlam VJ, Harrison JC, Porteous CM, James AM, Smith RAJ, Murphy
23. Luo J, McMullen JR, Sobkiw CL, Zhang L, Dorfman AL, Sherwood MP, Sammut IA. Targeting an antioxidant to mitochondria decreases
MC, Logsdon MN, Horner JW, DePinho RA, Izumo S, Cantley LC. cardiac ischemia-reperfusion injury. FASEB J. 2005;19:1088 –1095.
Class IA phosphoinositide 3-kinase regulates heart size and physiolog- 46. Powers SK, Demirel HA, Vincent HK, Coombes JS, Naito H, Hamilton
ical cardiac hypertrophy. Mol Cell Biol. 2005;25:9491–9502. KL, Shanely RA, Jessup J. Exercise training improves myocardial tol-
24. Carè A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang erance to in vivo ischemia-reperfusion in the rat. Am J Physiol. 1998;
ML, Segnalini P, Gu Y, Dalton ND, Elia L, Latronico MVG, Hoydal M, 275:R1468 –R1477.
Autore C, Russo MA, Dorn GW, Ellingsen O, Ruiz-Lozano P, Peterson 47. Ascensao A, Ferreira R, Magalhnes J. Exercise-induced cardiopro-
KL, Croce CM, Peschle C, Condorelli G. MicroRNA-133 controls tection: biochemical, morphological and functional evidence in whole
cardiac hypertrophy. Nat Med. 2007;13:613– 618. tissue and isolated mitochondria. Int J Cardiol. 2007;117:16 –30.

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015


Gielen et al Cardiovascular Effects of Exercise Training 1235

48. Hamilton KL, Quindry JC, French JP, Staib J, Hughes J, Mehta JL, net balance of cardiac Ca2⫹ handling protein expression in heart failure.
Powers SK. MnSOD antisense treatment and exercise-induced pro- Physiol Genomics. 2007;29:246 –252.
tection against arrhythmias. Free Radic Biol Med. 2004;37:1360 –1368. 71. Lu L, Mei DF, Gu AG, Wang S, Lentzner B, Gutstein DE, Zwas D,
49. Powers SK, Criswell D, Lawler J, Martin D, Lieu FK, Ji LL, Herb RA. Homma S, Yi GH, Wang J. Exercise training normalizes altered
Rigorous exercise training increases superoxide dismutase activity in calcium-handling proteins during development of heart failure. J Appl
ventricular myocardium. Am J Physiol. 1993;34:H2094 –H2098. Physiol. 2002;92:1524 –1530.
50. Kanter MM, Hamlin RL, Unverferth DV, Davis HW, Merola AJ. Effect 72. Kemi OJ, Ellingsen O, Ceci M, Grimaldi S, Smith GL, Condorelli G,
of exercise training on antioxidant enzymes and cardiotoxicity of doxo- Wisloff U. Aerobic interval training enhances cardiomyocyte contrac-
rubicin. J Appl Physiol. 1985;59:1298 –1303. tility and Ca2⫹ cycling by phosphorylation of CaMKII and Thr-17 of
51. Lennon SL, Quindry JC, Hamilton KL, French JP, Hughes J, Mehta JL, phospholamban. J Mol Cell Cardiol. 2007;43:354 –361.
Powers SK. Elevated MnSOD is not required for exercise-induced 73. Wisloff U, Loennechen JP, Falck G, Beisvag V, Currie S, Smith G,
cardioprotection against myocardial stunning. Am J Physiol. 2004;287: Ellingsen O. Increased contractility and calcium sensitivity in cardiac
H975–H980. myocytes isolated from endurance trained rats. Cardiovasc Res. 2001;
52. Marcil M, Bourduas K, Ascah A, Burelle Y. Exercise training induces 50:495–508.
respiratory substrate-specific decrease in Ca2⫹-induced permeability 74. Kemi OJ, Wisloff U. Mechanisms of exercise-induced improvements in
transition pore opening in heart mitochondria. Am J Physiol. 2006;290: the contractile apparatus of the mammalian myocardium. Acta Physiol.
H1549 –H1557. 2010;199:425– 439.
53. Starnes JW, Barnes BD, Olsen ME. Exercise training decreases rat heart 75. Coats AJS, Adamopoulos S, Radaelli A, McCance A, Meyer TE,
mitochondria free radical generation but does not prevent Ca2⫹-induced Bernardi L, Solda PL, Davey P, Ormerod O, Forfar C, Conway J,
dysfunction. J Appl Physiol. 2007;102:1793–1798. Sleight P. Controlled trial of physical training in chronic heart
54. Kavazis AN. Exercise preconditioning of the myocardium. Sports Med. failure: exercise performance, hemodynamics, ventilation, and
2009;39:923–935. autonomic function. Circulation. 1992;85:2119 –2131.
55. Pchejetski D, Kunduzova O, Dayon A, Calise D, Seguelas MH, Leducq 76. Braith R, Welsch M, Feigenbaum M, Kluess HA, Pepine C. Neuroen-
N, Seif I, Parini A, Cuvillier O. Oxidative stress– dependent sphingosine docrine activation in heart failure is modified by endurance training.
kinase-1 inhibition mediates monoamine oxidase A–associated cardiac J Am Coll Cardiol. 1999;34:1170 –1175.
cell apoptosis. Circ Res. 2007;100:41– 49. 77. Xu X, Wan W, Powers AS, Li J, Ji LL, Lao S, Wilson B, Erikson JM,
56. Gross GJ, Peart JN. KATP channels and myocardial preconditioning: an Zhang JQ. Effects of exercise training on cardiac function and myo-
update. Am J Physiol. 2003;285:H921–H930. cardial remodeling in post myocardial infarction rats. J Mol Cell
57. Goll DE, Thompson VF, Li H, Wei W, Cong J. The calpain system. Cardiol. 2008;44:114 –122.
Physiol Rev. 2003;83:731– 801. 78. Paulus WJ, Tschope C, Sanderson JE, Rusconi C, Flachskampf FA,
58. French JP, Quindry JC, Falk DJ, Staib JL, Lee Y, Wang KKW, Powers Rademakers FE, Marino P, Smiseth OA, De Keulenaer G, Leite-Moreira
SK. Ischemia-reperfusion-induced calpain activation and SERCA2a
AF, Borbely A, Edes I, Handoko ML, Heymans S, Pezzali N, Pieske B,
degradation are attenuated by exercise training and calpain inhibition.
Dickstein K, Fraser AG, Brutsaert DL. How to diagnose diastolic heart
Am J Physiol. 2006;290:H128 –H136.
failure: a consensus statement on the diagnosis of heart failure with
59. Starnes JW, Taylor RP, Ciccolo JT. Habitual low-intensity exercise does
normal left ventricular ejection fraction by the Heart Failure and Echo-
not protect against myocardial dysfunction after ischemia in rats. Eur
cardiography Associations of the European Society of Cardiology. Eur
J Cardiovasc Prev Rehabil. 2005;12:169 –174.
Heart J. 2007;28:2539 –2550.
60. Lennon SL, Quindry JC, French JP, Kim S, Mehta JL, Powers SK.
79. Brandao MU, Wajngarten M, Rondon E, Giorgi MC, Hironaka F,
Exercise and myocardial tolerance to ischaemia-reperfusion. Acta
Negrao CE. Left ventricular function during dynamic exercise in
Physiol Scand. 2004;182:161–169.
untrained and moderately trained subjects. J Appl Physiol. 1993;75:
61. Demirel HA, Powers SK, Zergeroglu MA, Shanely RA, Hamilton K,
1989 –1995.
Coombes J, Naito H. Short-term exercise improves myocardial tolerance
80. Levy WC, Cerqueira MD, Abrass IB, Schwartz RS, Stratton JR.
to in vivo ischemia-reperfusion in the rat. J Appl Physiol. 2001;91:
Endurance exercise training augments diastolic filling at rest and during
2205–2212.
62. Lennon SL, Quindry J, Hamilton KL, French J, Staib J, Mehta JL, exercise in healthy young and older men. Circulation. 1993;88:
Powers SK. Loss of exercise-induced cardioprotection after cessation of 116 –126.
exercise. J Appl Physiol. 2004;96:1299 –1305. 81. Yu CM, Li LS-W, Lam MF, Siu DC-W, Miu RK-M, Lau CP. Effect of
63. Volterrani M, Clark AL, Ludman PF, Swan JW, Adamopoulos S, a cardiac rehabilitation program on left ventricular diastolic function and
Piepoli M, Coats AJS. Predictors of exercise capacity in chronic heart its relationship to exercise capacity in patients with coronary heart
failure. Eur Heart J. 1994;15:801– 809. disease: experience from a randomized, controlled study. Am Heart J.
64. Sullivan MJ, Higginbotham MB, Cobb FR. Exercise training in patients 2004;147:874.
with severe left ventricular dysfunction: hemodynamic and metabolic 82. Smart N, Haluska B, Jeffriess L, Marwick TH. Exercise training in
effects. Circulation. 1988;78:506 –515. systolic and diastolic dysfunction: effects on cardiac function, functional
65. Hambrecht R, Gielen S, Linke A, Fiehn E, Yu J, Walther C, Schoene N, capacity, and quality of life. Am Heart J. 2007;153:530 –536.
Schuler G. Effects of exercise training on left ventricular function and 83. Brassard P, Legault S, Garneau C, Bogaty P, Dumesnil JG, Poirier P.
peripheral resistance in patients with chronic heart failure: a randomised Normalization of diastolic dysfunction in type 2 diabetics after exercise
trial. JAMA. 2000;283:3095–3101. training. Med Sci Sports Exerc. 2007;39:1896 –1901.
66. Giannuzzi P, Temporelli PL, Corra U, Tavazzi L. Antiremodeling effect 84. Gary RA, Sueta CA, Dougherty M, Rosenberg B, Cheek D, Preisser J,
of long-term exercise training in patients with stable chronic heart Neelon V, McMurray R. Home-based exercise improves functional
failure: results of the Exercise in Left Ventricular Dysfunction and performance and quality of life in women with diastolic heart failure.
Chronic Heart Failure (ELVD-CHF) Trial. Circulation. 2003;108: Heart Lung. 2004;33:210 –218.
554 –559. 85. Pieske B, Maier LS, Bers DM, Hasenfuss G. Ca2⫹ handling and
67. Hambrecht R, Fiehn E, Weigl C, Gielen S, Hamann C, Kaiser R, Yu J, sarcoplasmic reticulum Ca2⫹ content in isolated failing and nonfailing
Adams V, Niebauer J, Schuler G. Regular physical exercise corrects human myocardium. Circ Res. 1999;85:38 – 46.
endothelial dysfunction and improves exercise capacity in patients with 86. Marks AR. Ryanodine receptors/calcium release channels in heart
chronic heart failure. Circulation. 1998;98:2709 –2715. failure and sudden cardiac death. J Mol Cell Cardiol. 2001;33:615– 624.
68. Adams V, Link A, Gielen S, Erbs S, Hambrecht R, Schuler G. Modu- 87. Marx SO, Reiken S, Hisamatsu Y, Jayaraman T, Burkhoff D, Rosemblit
lation of Murf-1 and MAFbx expression in the myocardium by physical N, Marks AR. PKA phosphorylation dissociates FKBP12.6 from the
exercise training. Eur J Cardiovasc Prev Rehabil. 2008;15:293–299. calcium release channel (ryanodine receptor): defective regulation in
69. Lenk K, Schur R, Linke A, Erbs S, Matsumoto Y, Adams V, Schuler G. failing hearts. Cell. 2000;101:365–376.
Impact of exercise training on myostatin expression in the myocardium 88. Medeiros A, Rolim NPL, Oliveira RSF, Rosa KT, Mattos KC, Casarini
and skeletal muscle in a chronic heart failure model. Eur J Heart Fail. DE, Irigoyen MC, Krieger EM, Krieger JE, Negrao CE, Brum PC.
2009;11:342–348. Exercise training delays cardiac dysfunction and prevents calcium
70. Rolim NPL, Medeiros A, Rosa KT, Mattos KC, Irigoyen MC, Krieger handling abnormalities in sympathetic hyperactivity-induced heart
EM, Krieger JE, Negrao CE, Brum PC. Exercise training improves the failure mice. J Appl Physiol. 2008;104:103–109.

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015


1236 Circulation September 21, 2010

89. Choi SY, Chang HJ, Choi SI, Kim KI, Cho YS, Youn TJ, Chung WY, 112. Petersen SE, Wiesmann F, Hudsmith LE, Robson MD, Francis JM,
Chae IH, Choi DJ, Kim HS, Kim CH, Oh BH, Kim MH. Long-term Selvanayagam JB, Neubauer S, Channon KM. Functional and structural
exercise training attenuates age-related diastolic dysfunction: asso- vascular remodeling in elite rowers assessed by cardiovascular magnetic
ciation of myocardial collagen cross-linking. J Korean Med Sci. 2009; resonance. J Am Coll Cardiol. 2006;48:790 –797.
24:32–39. 113. Otsuki T, Maeda S, Iemitsu M, Saito Y, Tanimura Y, Ajisaka R,
90. Goldbarg SH, Elmariah S, Miller MA, Fuster V. Insight into degen- Miyauchi T. Vascular endothelium-derived factors and arterial stiffness
erative aortic valve disease. J Am Coll Cardiol. 2007;50:1205–1213. in strength- and endurance-trained men. Am J Physiol. 2007;292:
91. Agmon Y, Khandheria BK, Meissner I, Sicks JD, O’Fallon WM, H786 –H791.
Wiebers DO, Whisnant JP, Seward JB, Tajik AJ. Aortic valve sclerosis 114. Collier SR, Kanaley JA, Carhart R Jr, Frechette V, Tobin MM, Hall AK,
and aortic atherosclerosis: different manifestations of the same disease? Luckenbaugh AN, Fernhall B. Effect of 4 weeks of aerobic or resistance
Insights from a population-based study. J Am Coll Cardiol. 2001;38: exercise training on arterial stiffness, blood flow and blood pressure in
827– 834. pre- and stage-1 hypertensives. J Hum Hypertens. 2008;22:678 – 686.
92. Aikawa E, Aikawa M, Libby P, Figueiredo JL, Rusanescu G, Iwamoto 115. Edwards DG, Schofield RS, Magyari PM, Nichols WW, Braith RW.
Y, Fukuda D, Kohler RH, Shi GP, Jaffer FA, Weissleder R. Arterial and Effect of exercise training on central aortic pressure wave reflection in
aortic valve calcification abolished by elastolytic cathepsin S deficiency coronary artery disease. Am J Hypertens. 2004;17:540 –553.
in chronic renal disease. Circulation. 2009;119:1785–1794. 116. Ludmer PL, Selwyn AP, Shook TL, Wayne RR, Mudge GH, Alexander
93. Laufs U, Wassmann S, Czech T, Münzel T, Eisenhauer M, Böhm M, RW, Ganz P. Paradoxical vasoconstriction induced by acetylcholine in
Nickenig G. Physical inactivity increases oxidative stress, endothelial atherosclerotic coronary arteries. N Engl J Med. 1986;315:1046 –1051.
dysfunction, and atherosclerosis. Arterioscler Thromb Vasc Biol. 2005; 117. Hambrecht R, Wolff A, Gielen S, Linke A, Hofer J, Erbs S, Schoene N,
25:809 – 814. Schuler G. Effect of exercise on coronary endothelial function in
94. Adams V, Linke A, Kränkel N, Erbs S, Gielen S, Möbius-Winkler S, patients with coronary artery disease. N Engl J Med. 2000;342:
Gummert JF, Mohr FW, Schuler G, Hambrecht R. Impact of regular 454 – 460.
physical activity on the NAD(P)H oxidase and angiotensin receptor 118. Gielen S, Erbs S, Linke A, Möbius-Winkler S, Schuler G, Hambrecht R.
system in patients with coronary artery disease. Circulation. 2005;111: Home-based versus hospital-based exercise programs in patients with
555–562. coronary artery disease: effects on coronary vasomotion. Am Heart J.
95. Matsumoto Y, Adams V, Jacob S, Mangner N, Schuler G, Linke A. 2003;145:E3.
Regular exercise training prevents aortic valve disease in LDLR 119. Gokce N, Vita JA, Bader DS, Sherman DL, Hunter LM, Holbrook M,
deficient mice. Circulation. 2010;121:759 –767. O’Malley JF, Balady GJ. Effect of exercise on upper and lower
96. Mohler ER III. Mechanisms of aortic valve calcification. Am J Cardiol. extremity endothelial function in patients with coronary artery disease.
2004;94:1396 –1402. Am J Cardiol. 2002;90:124 –127.
97. Helske S, Syvaranta S, Lindstedt KA, Lappalainen J, Oorni K, May- 120. Linke A, Schoene N, Gielen S, Hofer J, Erbs S, Schuler G, Hambrecht
ranpaa MI, Lommi J, Turto H, Werkkala K, Kupari M, Kovanen PT. R. Endothelial dysfunction in patients with chronic heart failure:
Increased expression of elastolytic cathepsins S, K, and V and their systemic effects of lower-limb exercise training. J Am Coll Cardiol.
inhibitor cystatin C in stenotic aortic valves. Arterioscler Thromb Vasc 2001;37:392–397.
Biol. 2006;26:1791–1798. 121. Davis PF, Helmke BP. Endothelial mechanotransduction. In: Mofrad
98. Miller JD, Chu Y, Brooks RM, Richenbacher WE, Peta-Silva R, Heistad MRK, Kamm RD, eds. Cellular Mechanotransduction: Diverse Per-
DD. Dysregulation of antioxidant mechanisms contributes to increased spectives From Molecules to Tissue. New York, NY: Cambridge Uni-
oxidative stress in calcific aortic valvular stenosis in humans. J Am Coll versity Press; 2010:20 – 60.
Cardiol. 2008;52:843– 850. 122. Li YS, Haga JM, Chien S. Molecular basis of the effects of shear stress
99. Weiss RM, Ohashi M, Miller JD, Young SG, Heistad DD. Calcific aortic on vascular endothelial cells. J Biomech. 2005;38:1949 –1971.
valve stenosis in old hypercholesterolemic mice. Circulation. 2006;114: 123. Tzima E, Irani-Tehrani M, Kiosses WB, Dejana E, Schultz DA,
2065–2069. Engelhardt B, Cao G, DeLisser H, Schwartz MA. A mechanosensory
100. Hruska KA, Mathew S, Saab G. Bone morphogenetic proteins in complex that mediates the endothelial cell response to fluid shear stress.
vascular calcification. Circ Res. 2005;97:105–114. Nature. 2005;437:426 – 431.
101. Altschul R. Endothelium, its Development, Morphology, Function, and 124. Böger RH, Bode-Böger SM, Szuba A, Tsao PS, Chan JR, Tangphao O,
Pathology. New York, NY: Macmillan; 1954. Blaschke TF, Cooke JP. Asymmetric dimethylarginine (ADMA): a
102. Ross J. The pathogenesis of atherosclerosis: a perspective for the 1990s. novel risk factor for endothelial dysfunction: its role in hypercholester-
Nature. 1993;362:801– 809. olemia. Circulation. 1998;98:1842–1847.
103. Schächinger V, Britten MB, Zeiher A. Prognostic impact of coronary 125. Wassmann S, Wassmann K, Nickenig G. Modulation of oxidant and anti-
vasodilator dysfunction on adverse long-term outcome of coronary heart oxidant enzyme expression and function in vascular cells. Hypertension.
disease. Circulation. 2000;101:1899 –906. 2004;44:381–386.
104. Perticone F, Ceravolo R, Pujia A, Ventura G, Iacopino S, Scozzafava A, 126. Crosby JR, Kaminski WE, Schatteman G, Martin PJ, Raines EW, Seifert
Ferraro A, Chello M, Mastroroberto P, Verdecchia P, Schillaci G. RA, Bowen-Pope DF. Endothelial cells of hematopoietic origin make a
Prognostic significance of endothelial dysfunction in hypertensive significant contribution to adult blood vessel formation. Circ Res. 2000;
patients. Circulation. 2001;104:191–196. 87:728 –730.
105. Landmesser U, Drexler H. The clinical significance of endothelial dys- 127. Förstermann U, Münzel T. Endothelial nitric oxide synthase in vascular
function. Curr Opin Cardiol. 2005;20:547–551. disease. Circulation. 2006;113:1708 –1714.
106. Ghiadoni L, Versari D, Giannarelli C, Faita F, Taddei S. Non-invasive 128. Widder JD, Chen W, Li L, Dikalov S, Thony B, Hatakeyama K,
diagnostic tools for investigating endothelial dysfunction. Curr Pharm Harrison DG. Regulation of tetrahydrobiopterin biosynthesis by shear
Des. 2008;14:3715–3722. stress. Circ Res. 2007;101:830 – 838.
107. Jones CJ, Kuo L, Davis MJ, Chilian WM. Regulation of coronary blood 129. Lam CF, Peterson TE, Richardson DM, Croatt AJ, d’Uscio LV, Nath
flow: coordination of heterogeneous control mechanisms in vascular KA, Katusic ZS. Increased blood flow causes coordinated upregulation
microdomains. Cardiovasc Res. 1995;29:585–596. of arterial eNOS and biosynthesis of tetrahydrobiopterin. Am J Physiol.
108. Kuo L, Davis MJ, Chilian WM. Longitudinal gradients for endotheli- 2006;290:H786 –H793.
um-dependent and -independent vascular responses in the coronary 130. Sindler AL, Delp MD, Reyes R, Wu G, Muller-Delp JM. Effects of
microcirculation. Circulation. 1995;92:518 –525. ageing and exercise training on eNOS uncoupling in skeletal muscle
109. Wagenseil JE, Mecham RP. Vascular extracellular matrix and arterial resistance arterioles. J Physiol. 2009;587:3885–3897.
mechanics. Physiol Rev. 2009;89:957–989. 131. Richter B, Niessner A, Penka M, Grdic M, Steiner S, Strasser B, Ziegler
110. Dart AM, Lacombe F, Yeoh JK, Cameron JD, Jennings GL, Laufer E, S, Zorn G, Maurer G, Simeon-Rudolf V, Wojta J, Huber K. Endurance
Esmore DS. Aortic distensibility in patients with isolated hypercholes- training reduces circulating asymetric dimethylarginine and myeloper-
terolaemia, coronary artery disease, or cardiac transplant. Lancet. 1991; oxidase levels in persons at risk of coronary events. Thromb Haemost.
338:270 –273. 2005;94:1306 –1311.
111. Avolio A, Jones D, Tafazzoli-Shadpour M. Quantification of alterations 132. Hambrecht R, Adams V, Erbs S, Linke A, Kränkel N, Shu Y, Baither
in structure and function of elastin in the arterial media. Hypertension. Y, Gielen S, Gummert JF, Mohr FW, Schuler G. Regular physical
1998;32:170 –175. activity improves endothelial function in patients with coronary

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015


Gielen et al Cardiovascular Effects of Exercise Training 1237

artery disease by increasing phosphorylation of endothelial nitric 151. Cheng XW, Kuzuya M, Nakamura K, Maeda K, Tsuzuki M, Kim W,
oxide synthase. Circulation. 2003;107:3152–3158. Sasaki T, Liu Z, Inoue N, Kondo T, Jin H, Numaguchi Y, Okumura K,
133. Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, Yokota M, Iguchi A, Murohara T. Mechanisms underlying the
Donato AJ, Lesniewski LA. Voluntary wheel running restores endothe- impairment of ischemia-induced neovascularization in matrix metallo-
lial function in conduit arteries of old mice: direct evidence for reduced proteinase 2-deficient mice. Circ Res. 2007;100:904 –913.
oxidative stress, increased superoxide dismutase activity and down- 152. Huang PH, Chen YH, Wang CH, Chen JS, Tsai HY, Lin FY, Lo WY,
regulation of NADPH oxidase. J Physiol. 2009;587:3271–3285. Wu TC, Sata M, Chen JW, Lin SJ. Matrix metalloproteinase-9 is
134. Fukai T, Siegfried MR, Ushio-Fukai M, Cheng Y, Kojda G, Harrison essential for ischemia-induced neovascularization by modulating bone
DG. Regulation of the vascular extracellular superoxide dismutase by marrow– derived endothelial progenitor cells. Arterioscler Thromb Vasc
nitric oxide and exercise training. J Clin Invest. 2000;105:1631–1639. Biol. 2009;29:1179 –1184.
135. Ennezat PV, Malendowicz LS, Testa M, Colombo PC, Cohen-Solal A, 153. Lapidot T, Petit I. Current understanding of stem cell mobilization: the
Evans T, LeJemetel TH. Physical training in patients with chronic heart roles of chemokines, proteolytic enzymes, adhesion molecules, cyto-
failure enhances the expression of genes antioxidative enzymes. J Am kines, and stromal cells. Exp Hematol. 2002;30:973–981.
Coll Cardiol. 2001;38:194 –198. 154. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal
136. Zanchi NE, Grassmann-Bechera LR, Tanaka LY, Debbas V, Bar- RG, Besmer P, Lyden D, Moore MA, Werb Z, Rafii S. Recruitment of
tholomeu T, Ramirs PR. Moderate exercise training decreases aortic stem and progenitor cells from the bone marrow nighe requires MMP-9
superoxide production in myocardial infarcted rats. Eur J Appl Physiol. mediated release of kit-ligand. Cell. 2002;109:625– 637.
2008;104:1045–1052. 155. Iwakura A, Shastry S, Luedemann C, Hamada H, Kawamoto A, Kishore
137. Seshiah PN, Weber DS, Rocic P, Valppu L, Taniyama Y, Griendling R, Zhu Y, Qin G, Silver M, Thorne T, Eaton L, Masuda H, Asahara T,
KK. Angiotensin II stimulation of NAD(P)H oxidase activity: upstream Losordo DW. Estradiol enhances recovery after myocardial infarction
mediators. Circ Res. 2002;91:406 – 413. by augmenting incorporation of bone marrow– derived endothelial pro-
138. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, genitor cells into sites of ischemia-induced neovascularization via en-
Witzenbichler B, Schatteman G, Isner JM. Isolation of putative pro- dothelial nitric oxide synthase–mediated activation of matrix
genitor endothelial cells for angioneogenesis. Science. 1997;275: metalloproteinase-9. Circulation. 2006;113:1605–1614.
964 –967. 156. Askari AT, Unzek S, Popovic ZB, Goldman CK, Forudi F, Kiedrowski
139. Huang NF, Li S. Mesenchymal stem cells for vascular regeneration. M, Rovner A, Ellis SG, Thomas JD, DiCorleto PE, Topol EJ, Penn MS.
Regen Med. 2008;3:877– 892. Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue
140. Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, Bohm M, regeneration in ischaemic cardiomyopathy. Lancet. 2003;362:697–703.
Nickenig G. Circulating endothelial progenitor cells and cardiovascular 157. Tachibana K, Hirota S, Iizasa H, Yoshida H, Kawabata K, Kataoka Y,
outcomes. N Engl J Med. 2005;353:999 –1007. Kitamura Y, Matsushima K, Yoshida N, Nishikawa Si, Kishimoto T,
141. Adams V, Lenk K, Linke A, Lenz D, Erbs S, Sandri M, Tarnok A, Nagasawa T. The chemokine receptor CXCR4 is essential for vascular-
ization of the gastrointestinal tract. Nature. 1998;393:591–594.
Gielen S, Emmrich F, Schuler G, Hambrecht R. Increase of circulating
158. Schuler G, Hambrecht R, Schlierf G, Niebauer J, Hauer K, Neumann J,
endothelial progenitor cells in patients with coronary artery disease after
Hoberg E, Drinkmann A, Bacher F, Grunze M, Kübler W. Regular
exercise-induced ischemia. Arterioscler Thromb Vasc Biol. 2004;24:
physical exercise and low-fat diet: effects on progression of coronary
684 – 690.
artery disease. Circulation. 1992;86:1–11.
142. Sandri M, Adams V, Gielen S, Linke A, Lenk K, Krankel N, Lenz D,
159. Niebauer J, Hambrecht R, Velich T, Hauer K, Marburger C, Kälberer
Erbs S, Scheinert D, Mohr FW, Schuler G, Hambrecht R. Effects of
B, Weiss C, von Hodenberg E, Schlierf G, Schuler G, Zimmermann
exercise and ischemia on mobilization and functional activation of
R, Kübler W. Attenuated progression of coronary artery disease after
blood-derived progenitor cells in patients with ischemic syndromes:
6 years of multifactorial risk intervention: role of physical exercise.
results of 3 randomized studies. Circulation. 2005;111:3391–3399.
Circulation. 1997;96:2534 –2541.
143. Shaffer RG, Greene S, Arshi A, Supple G, Bantly A, Moores JS,
160. Ornish D, Brown SE, Scherwitz LW, Billings JH, Armstrong WT, Ports
Parmacek MS, Mohler ER III. Effect of acute exercise on endothelial
TA, McLanahan SM, Kirkeeide RL, Brand RJ, Gould KL. Can lifestyle
progenitor cells in patients with peripheral arterial disease. Vasc Med.
changes reverse coronary heart disease? The Lifestyle Heart Trial.
2006;11:219 –226. Lancet. 1990;336:129 –133.
144. Van Craenenbroeck EMF, Vrints CJ, Haine SE, Vermeulen K, Goo- 161. Haskell WL, Alderman EL, Fair JM, Maron DJ, Mackey SF, Superko R,
vaerts I, Van Tendeloo VFI, Hoymans VY, Conraads VMA. A maximal Williams PT, Johnstone IM, Champagne MA, Krauss RM, Farquhar JW.
exercise bout increases the number of circulating CD34⫹/KDR⫹ en- Effects of intensive multiple risk factor reduction on coronary athero-
dothelial progenitor cells in healthy subjects: relation with lipid profile. sclerosis and clinical cardiac events in men and women with coronary
J Appl Physiol. 2008;104:1006 –1013. artery disease: the Stanford Coronary Risk Intervention Project (SCRIP).
145. Bonsignore MR, Morici G, Santoro A, Pagano M, Cascio L, Bonanno A, Circulation. 1994;89:975–990.
Abate P, Mirabella F, Profita M, Insalaco G, Giola M, Vignola AM, 162. Deussen A, Brand M, Pexa A, Weichsel J. Metabolic coronary flow
Majolino I, Testa U, Hogg JC. Circulating hematopoietic progenitor regulation: current concepts. Basic Res Cardiol. 2006;101:453– 464.
cells in runners. J Appl Physiol. 2002;93:1691–1697. 163. Busse R, Förstermann U, Matsuda H, Pohl U. The role of prostaglandins
146. Adams V, Linke A, Breuckmann F, Leineweber K, Erbs S, Kränkel N, in the endothelium-mediated vasodilatory response to hypoxia. Pfugers
Bröcker-Preuss M, Woitek F, Erbel R, Heusch G, Hambrecht R, Schuler Arch. 1984;401:77– 83.
G, Möhlenkamp S. Circulating progenitor cells decrease immediately 164. Bouchard JF, Dumont E, Lamontagne D. Evidence that prostaglandins
after marathon race in advanced-age marathon runners. Eur J Car- I2, E2, and D2 may activate ATP sensitive potassium channels in the
diovasc Prev Rehabil. 2008;15:602– 607. isolated rat heart. Cardiovasc Res. 1994;28:901–905.
147. Möbius-Winkler S, Hilberg T, Menzel K, Golla E, Burman A, Schuler 165. Kroll K, Kinzie DJ, Gustafson LA. Open-system kinetics of myocardial
G, Adams V. Time-dependent mobilization of circulating progenitor phosphoenergetics during coronary underperfusion. Am J Physiol. 1997;
cells during strenuous exercise in healthy individuals. J Appl Physiol. 272:H2563–H2576.
2009;107:1943–1950. 166. Feigl EO. Coronary physiology. Physiol Rev. 1983;63:1–205.
148. Goussetis E, Spiropoulos A, Tsironi M, Skenderi K, Margeli A, Gra- 167. Dietrich HH, Ellsworth ML, Sprague RS, Dacey RG Jr. Red blood cell
phakos S, Baltopoulos P, Papassotiriou I. Spartathlon, a 246 kilometer regulation of microvascular tone through adenosine triphosphate. Am J
foot race: effects of acute inflammation induced by prolonged exercise Physiol. 2000;278:H1294 –H1298.
on circulating progenitor reparative cells. Blood Cells Mol Dis. 2009; 168. DiCarlo SE, Blair RW, Bishop VS, Stone HL. Daily exercise enhances
42:294 –299. coronary resistance vessel sensitivity to pharmacological activation.
149. Asahara T, Takahashi T, Masuda H. VEGF contributes to postnatal J Appl Physiol. 1989;66:421– 428.
neovascularization by mobilizing bone-marrow endothelial progenitor 169. Laughlin MH, Overholser KA, Bhatte MJ. Exercise training increases
cells. EMBO J. 1999;18:3964 –3972. coronary transport reserve in miniature swine. J Appl Physiol. 1989;67:
150. Li B, Sharpe EE, Maupin AB, Teleron AA, Pyle AL, Carmeliet P, 1140 –1149.
Young PP. VEGF and PlGF promote adult vasculogenesis by enhancing 170. Muller JM, Myers PR, Laughlin MH. Exercise training alters myogenic
EPC recruitment and vessel formation at the site of tumor neovascular- responses in porcine coronary resistance arteries. J Appl Physiol. 1993;
ization. FASEB J. 2006;20:1495–1497. 75:2677–2682.

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015


1238 Circulation September 21, 2010

171. Karibe A, Watanabe J, Horiguchi S, Takeuchi M, Suzuki S, Funakoshi 188. Welsch M, Alomari M, Parish TR, Wood RH, Kalb D. Influence of
M, Katoh H, Keitoku M, Satoh S, Shirato K. Role of cytosolic Ca2⫹ and venous function on exercise tolerance in chronic heart failure. J Car-
protein kinase C in developing myogenic contraction in isolated rat diopulm Rehabil. 2002;22:321–326.
small arteries. Am J Physiol. 1997;272:H1165–H1172. 189. Maiorana A, O’Driscoll G, Dembo L, Cheetham C, Goodman C, Taylor
172. Korzick DH, Laughlin MH, Bowles DK. Alterations in PKC signaling R, Green D. Effect of aerobic and resistance exercise training on
underlie enhanced myogenic tone in exercise-trained porcine coronary vascular function in heart failure. Am J Physiol. 2000;279:
resistance arteries. J Appl Physiol. 2004;96:1425–1432. H1999 –H2005.
173. White FC, Bloor CM, McKirnan MD, Carroll SM. Exercise training in 190. Roveda F, Middlekauff HR, Rondon MU, Reis SF, Souza M, Nastari L,
swine promotes growth of arteriolar bed and capillary angiogenesis in Barretto AC, Krieger EM, Negrão CE. The effects of exercise training
heart. J Appl Physiol. 1998;85:1160 –1168. on sympathetic neural activation in advanced heart failure: a randomized
174. Carrow RE, Brown RE, Van Huss WD. Fiber sizes and capillary to fiber controlled trial. J Am Coll Cardiol. 2003;42:854 – 860.
ratios in skeletal muscle of exercised rats. Anat Rec. 1967;159:33–39. 191. Suzuki J, Kobayashi T, Uruma T, Koyama T. Time-course changes in
175. Ingjer F. Effects of endurance training on muscle fiber ATP-ase activity, arteriolar and venular portions of capillary in young treadmill-trained
capillary supply and mitochondrial content in man. J Physiol. 1979;294: rats. Acta Physiol Scand. 2001;171:77– 86.
419 – 432. 192. Lee AP, Ice R, Blessey R, Sanmarco ME. Long-term effect of physical training
176. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its on coronary patients with impaired ventricular function. Circulation. 1979;
receptors. Nat Med. 2003;9:669 – 676.
60:1519–1526.
177. Tang K, Xia FC, Wagner PD, Breen EC. Exercise-induced VEGF
193. Dubach P, Myers J, Dzienkan G, Goebbels U, Reinhart W, Muller P,
transcriptional activation in brain, lung and skeletal muscle. Respir
Buser P, Stulz P, Vogt P, Ratti R. Effect of high intensity exercise
Physiol Neurobiol. 2010;170:16 –22.
training on central hemodynamic response to exercise in men with
178. Mason SD, Rundqvist H, Papandreou I, Duh R, McNulty WJ, Howlett
reduced left ventricular function. J Am Coll Cardiol. 1997;29:
RA, Olfert IM, Sundberg CJ, Denko NC, Poellinger L, Johnson RS.
1591–1598.
HIF-1{alpha} in endurance training: suppression of oxidative metabo-
lism. Am J Physiol. 2007;293:R2059 –R2069. 194. Mereles D, Ehlken N, Kreuscher S, Ghofrani S, Hoeper MM, Halank M,
179. Chinsomboon J, Ruas J, Gupta RK, Thom R, Shoag J, Rowe GC, Meyer FJ, Karger G, Buss J, Juenger J, Holzapfel N, Opitz C, Winkler
Sawada N, Raghuram S, Arany Z. The transcriptional coactivator J, Herth FFJ, Wilkens H, Katus HA, Olschewski H, Grunig E. Exercise
PGC-1␣ mediates exercise-induced angiogenesis in skeletal muscle. and respiratory training improve exercise capacity and quality of life in
Proc Natl Acad Sci U S A. 2009;106:21401–21406. patients with severe chronic pulmonary hypertension. Circulation. 2006;
180. Arany Z, Foo SY, Ma Y, Ruas JL, Bommi-Reddy A, Girnun G, Cooper 114:1482–1489.
M, Laznik D, Chinsomboon J, Rangwala SM, Baek KH, Rosenzweig A, 195. Kashimura O, Sakai A. Effects of physical training on pulmonary
Spiegelman BM. HIF-independent regulation of VEGF and angio- arterial pressure during exercise under hypobaric hypoxia in rats. Int
genesis by the transcriptional coactivator PGC-1␣. Nature. 2008;451: J Biometerol. 1991;35:214 –221.
1008 –1012. 196. Kashimura O, Sakai A, Yanagidaira Y. Effects of exercise-training on
181. Richter EA, Ruderman NB. AMPK and the biochemistry of exercise: hypoxia and angiotensin II-induced pulmonary vasoconstrictions. Acta
implications for human health and disease. Biochem J. 2009;418: Physiol Scand. 1995;155:291–295.
261–275. 197. Favret F, Henderson KK, Allen J, Richalet JP, Gonzalez NC. Exercise
182. Zwetsloot KA, Westerkamp LM, Holmes BF, Gavin TP. AMPK reg- training improves lung gas exchange and attenuates acute hypoxic pul-
ulates basal skeletal muscle capillarization and VEGF expression, but is monary hypertension but does not prevent pulmonary hypertension of
not necessary for the angiogenic response to exercise. J Physiol. 2008; prolonged hypoxia. J Appl Physiol. 2006;100:20 –25.
586:6021– 6035. 198. Chen HI, Li HT. Physical conditioning can modulate endothelium-
183. Akimoto T, Pohnert SC, Li P, Zhang M, Gumbs C, Rosenberg PB, dependent vasorelaxation in rabbits. Arterioscler Thromb Vasc Biol.
Williams RS, Yan Z. Exercise stimulates PGC-1alpha transcription in 1993;13:852– 856.
skeletal muscle through activation of the p39MAPK pathway. J Biol 199. Mitani Y, Maruyama J, Maruyama K, Sakurai M. Exercise training does
Chem. 2005;280:19587–19593. not alter acetylcholine-induced responses in isolated pulmonary artery
184. Kang C, O’Moore KM, Dickman JR, Ji LL. Exercise activation of from rat. Eur Respir J. 1999;13:622– 625.
muscle peroxisome proliferator-activated receptor-[gamma] coactivator- 200. Johnson LR, Parker JL, Laughlin MH. Chronic exercise training
1[alpha] signaling is redox sensitive. Free Radic Biol Med. 2009;47: improves ACh-induced vasorelaxation in pulmonary arteries of pigs.
1394 –1400. J Appl Physiol. 2000;88:443– 451.
185. Gielen S, Hambrecht R. Venous function in chronic heart failure: the 201. Johnson LR, Rush JWE, Turk JR, Price EM, Laughlin MH. Short-term
neglected part of circulatory regulation. J Cardiopulm Rehabil. 2002; exercise training increases ACh-induced relaxation and eNOS protein in
22:327–328. porcine pulmonary arteries. J Appl Physiol 2001;90:1102–1110.
186. Monahan KD, Dinenno FA, Seals DR, Halliwill JR. Smaller age- 202. Handoko ML, de Man FS, Happe CM, Schalij I, Musters RJP,
associated reductions in leg venous compliance in endurance exercise- Westerhof N, Postmus PE, Paulus WJ, van der Laarse WJ, Vonk-
trained men. Am J Physiol. 2001;281:H1267–H1273.
Noordegraaf A. Opposite effects of training in rats with stable and
187. van Duijnhoven N, Bleeker M, de Groot P, Thijssen D, Felsenberg D,
progressive pulmonary hypertension. Circulation. 2009;120:42– 49.
Rittweger J, Hopman M. The effect of bed rest and an exercise coun-
termeasure on leg venous function. Eur J Appl Physiol. 2008;104:
991–998. KEY WORDS: coronary disease 䡲 endothelium 䡲 exercise 䡲 myocardium

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015


Cardiovascular Effects of Exercise Training: Molecular Mechanisms
Stephan Gielen, Gerhard Schuler and Volker Adams

Circulation. 2010;122:1221-1238
doi: 10.1161/CIRCULATIONAHA.110.939959
Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2010 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/122/12/1221

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation 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 is online at:


http://circ.ahajournals.org//subscriptions/

Downloaded from http://circ.ahajournals.org/ at UNIV OF PRINCE EDWARD ISLAND on March 3, 2015

You might also like