Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Review

This Review is in a thematic series on Exercise and the Cardiovascular System, which includes the following articles:

Exercise and the Cardiovascular System: Clinical Science and Cardiovascular Outcomes
[Circ Res 2015;117:207–219]
The Role of Exercise in Cardiac Aging: From Physiology to Molecular Mechanisms

The Role of Exercise in Cardiac Aging


From Physiology to Molecular Mechanisms
Jason Roh, James Rhee, Vinita Chaudhari, Anthony Rosenzweig

Abstract: Aging induces structural and functional changes in the heart that are associated with increased risk of
cardiovascular disease and impaired functional capacity in the elderly. Exercise is a diagnostic and therapeutic tool,
with the potential to provide insights into clinical diagnosis and prognosis, as well as the molecular mechanisms
by which aging influences cardiac physiology and function. In this review, we first provide an overview of how
aging impacts the cardiac response to exercise, and the implications this has for functional capacity in older
adults. We then review the underlying molecular mechanisms by which cardiac aging contributes to exercise
intolerance, and conversely how exercise training can potentially modulate aging phenotypes in the heart. Finally,
we highlight the potential use of these exercise models to complement models of disease in efforts to uncover
new therapeutic targets to prevent or treat heart disease in the aging population.   (Circ Res. 2016;118:279-295.
DOI: 10.1161/CIRCRESAHA.115.305250.)
Downloaded from http://ahajournals.org by on November 15, 2020

Key Words: aging ■ cardiovascular diseases ■ exercise ■ heart ■ phenotype

I mprovements in public health over the past century have led


to dramatic increases in life expectancy. By 2030, adults aged
>65 years will account for ≈20% of the general population in
maladaptive myocardial remodeling and the development of
HF in the elderly. However, there are also factors intrinsic
to cardiac aging, occurring at cellular and molecular levels,
the United States.1 Moreover, the oldest demographic groups, which may impair the overall function of the heart as it ap-
consisting of individuals ≥85 years, now represent the fast- proaches senescence.4–6
ing growing segment in the United States and are estimated to Although it can be difficult to completely separate extrinsic
increase by >230% by 2050.1 Thus, understanding the factors from intrinsic factors in cardiac aging given the interactions
limiting health and quality of life in the elderly will be increas- between aging, disease, and environment, genome-wide tran-
ingly important over the coming years. Among these factors, scriptome analyses of whole hearts and isolated cardiomyo-
cardiovascular disease represents the leading cause of mortality cytes from healthy young and old mice have provided some
in the elderly, accounting for ≈40% of all deaths.1,2 Heart failure insights into the molecular mechanisms of aging in the heart.
(HF), in particular, is reaching epidemic proportions with ≈88% Specific transcriptional alterations in pathways related to stress
of HF deaths and >75% of all HF hospitalizations in the United response, mitochondrial function, fatty acid metabolism, con-
States now occurring in adults aged ≥65 years.3 tractility, hypertrophy, inflammation, and extracellular matrix
Although advanced age is considered a major independent production have been identified as key molecular phenotypes
risk factor for HF, the mechanisms by which aging predis- of cardiac aging, which interestingly parallel many of the tran-
poses older adults to HF are not completely understood. The scriptional changes that occur in the failing heart.7,8 Although
cumulative impact of repeated insults and injuries (ie, myo- the aged heart is generally capable of meeting the basal energy
cardial infarctions and hypertension) to the heart through- requirements of the body, its performance under physiologic or
out its lifetime is undoubtedly an important contributor to pathologic stress can be significantly impaired, which can lead

Original received October 1, 2015; revision received November 19, 2015; accepted December 7, 2015.
From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee),
Massachusetts General Hospital and Harvard Medical School, Boston.
Correspondence to Anthony Rosenzweig, MD, Division of Cardiology, Massachusetts General Hospital, 55 Fruit St, Bigelow 800, Mailstop 817, Boston,
MA 02114. E-mail arosenzweig@partners.org
© 2016 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.115.305250

279
280  Circulation Research  January 22, 2016

nervous systems to increase blood flow and oxygen supply to


Nonstandard Abbreviations and Acronyms
the working skeletal muscle. At rest, muscle receives ≈20% of
β-AR β-adrenergic receptor the total blood flow, but during exercise, this can increase to
AC adenylyl cyclase >80%.13 Thus, impairments in any of these systems can lead to
CaMKII Ca2+/calmodulin kinase 2 significant decreases in peak cardiac output (CO) and overall
CO cardiac output exercise capacity.
GDF11 growth differentiation factor 11 The heart’s contributions to augmenting CO in response to
HF heart failure the increased metabolic demands of exercise have been well
HR heart rate characterized, and essentially depend on dynamic regulation of
IGF-1 insulin like growth factor-1 2 physiological parameters, heart rate (HR) and stroke volume
mtDNA mitochondrial DNA (SV). In healthy young adults, exercise-induced adrenergic
NFE2L2 nuclear factor-erythroid-derived 2-like 2 stimulation rapidly increases both HR and SV, with the latter
PGC-1α peroxisome proliferator-activated receptor gamma coactivator 1α being primarily enhanced by increased myocardial contractil-
PKA protein kinase A ity and decreased peripheral vascular resistance. SV increases
PolG polymerase gamma
proportionally with exercise intensity until ≈40% to 50% of
ROS reactive oxygen species
maximal capacity, after which it tends to plateau, and addition-
al augmentation of CO is driven by a further increase in HR.14
SERCA2a sarcoplasmic reticulum Ca2+–ATPase 2a
Although older adults are still capable of augmenting their
SIRT silent information regulator
CO in response to exercise, the relative increase is typically
SV stroke volume
diminished when compared with their younger counterparts.
TAC transverse aortic constriction
Reduced maximal HR, also known as chronotropic incompe-
VO2max maximum oxygen consumption
tence, is a major contributor to the diminished cardiac response
to exercise in older adults. Normal aging results in a progressive
decline in maximal HR by ≈0.7 bpm/y.15 Although the mecha-
to exercise intolerance and dyspnea, the primary symptoms of
nisms for chronotropic incompetence are not completely un-
HF. Thus, defining the mechanisms by which aging affects the
derstood, degenerative changes in the conduction system along
heart’s ability to respond to stressful stimuli becomes integral
with impaired autonomic regulation likely play central roles.16
to understanding the role of aging in HF pathophysiology.
Exercise, as an inducible form of physiologic stress, repre- Importantly, age-related decrease in peak HR strongly corre-
lates with diminished exercise capacity and is an independent
Downloaded from http://ahajournals.org by on November 15, 2020

sents a powerful tool in cardiac aging research for this reason.


Exercise physiology has provided a wealth of knowledge into predictor of adverse cardiovascular events and mortality.17,18
how age-related changes in cardiac structure and function trans- The impact of aging on SV augmentation with exercise
late to decreased exercise capacity,9,10 a strong determinant of is not as clear with varying degrees of SV reserve reported in
HF prognosis, quality of life, and mortality in the elderly.11,12 different studies.19–22 In general, aged hearts are still capable
Furthermore, exercise-based studies in aged animal models are of increasing SV in response to exercise, albeit at levels insuf-
now beginning to identify the molecular mechanisms of cardiac ficient to offset the reduction in maximum HR. Interestingly,
aging that contribute to exercise intolerance, and intriguingly sug- the mechanism by which the heart augments SV with exercise
gest that exercise, as a therapeutic intervention, can potentially changes with age. Although enhanced myocardial contractil-
mitigate or reverse some aspects of the aging process in the heart. ity is the primary means of increasing SV in young hearts,
Both aging and exercise are complex systemic processes exercise increases SV in aged hearts mainly through increased
that influence nearly every facet of the cardiovascular sys- end-diastolic volumes with minimal changes in contractility.10
tem. Here, we primarily focus on the role of exercise in the Overall, normal aging significantly diminishes both the
aged heart, with an emphasis on cardiomyocyte biology. We chronotropic and inotropic responses of the heart to exercise
first provide an overview of how aging impacts the cardiac (Table 1). Clinically, this phenomenon is referred to as im-
response to acute exercise and the implications this has on paired cardiac reserve, which is the inability of the heart to
declining functional capacity in the elderly. We then review adequately augment CO to meet the increased demands of
the literature on exercise in aged animals, highlighting the physiologic stress, whether induced by exercise or pharmaco-
major molecular mechanisms by which cardiomyocyte aging logically (ie, dobutamine). In conjunction with age-associated
is thought to contribute to exercise intolerance and how ex- alterations in peripheral mechanisms of oxygen extraction
ercise training potentially modulates these properties in the and utilization in skeletal muscle,19,21,23,24 inadequate oxygen
aged heart. Finally, given the parallels in exercise and cardiac delivery from impaired cardiac reserve is a major contribu-
phenotypes observed in advanced age and HF, we discuss the tor to decreased functional capacity in the elderly, especially
potential implications of these findings in the context of the those with HF.19,25,26 Maximum oxygen consumption (VO2max),
growing HF epidemic in the aging population. which is the maximal rate the body can consume oxygen dur-
ing incremental exercise, is an established metric of exercise
Effects of Aging on the Cardiac capacity. With normal aging, VO2max declines by ≈10% per
Response to Exercise decade in healthy ambulatory individuals,22 but this decline
Exercise exerts a physiologic stress on the body, requiring a notably accelerates at ages >70 years and in HF.27 This sug-
coordinated response by the cardiovascular, pulmonary, and gests that mechanisms that lead to impaired cardiac reserve
Roh et al   Exercise and Cardiac Aging   281

Table 1.  Summary of Age-Associated Changes in by Wisloff and colleagues35 have used a breeding selection
Cardiovascular Performance at Peak Exercise strategy in rats based on exercise capacity (referred to as the
CV Parameter at Peak Exercise Effects of Aging aerobic hypothesis). From 1996 to 2011, selective breeding of
a genetically heterogeneous N:NIH rat stock (28 generations,
Cardiac output ↓/NC
n=11 606 rats) eventually generated 2 distinct lines that dif-
Heart rate ↓ fered in maximal running capacity by ≈7-fold. Comparative
LV stroke volume ↑/↓/NC analyses of hearts and isolated cardiomyocytes from aged rats
LV end-diastolic volume ↑ with low and high intrinsic running capacities subsequently
LV contractility ↓ identified mitochondrial dysfunction,36 abnormal calcium
Early diastolic filling rate ↓
(Ca2+) handling,37 increased hypertrophy,38 and microvascular
dysfunction as key molecular phenotypes in the heart associ-
VO2max ↓
ated with exercise intolerance in aging (Figure).
(AV) O2 difference ↓ We will now explore in more detail how these features of
Effects of aging are derived from comparison of aerobic exercise testing of cardiomyocyte aging impair the aged heart’s response to acute
healthy young adults (20–30 years) and healthy older adults (60–80 years). exercise, and how exercise interventions potentially modulate
CV indicates cardiovascular; LV, left ventricle; and NC, no change. Data
these aging phenotypes. Although adaptive changes in the vas-
summarized.19–22
culature are important in both aging and exercise physiology,
a complete discussion of this topic is beyond the scope of this
in aging may be particularly relevant to the increased HF risk review, and we refer the interested reader to the following ref-
seen with advanced age. erences as an introduction to this area.4,39

Rodent Models of Cardiac Aging and Exercise Exercise and Autonomic Regulation
Intolerance of the Aged Heart
Although human studies have provided valuable insights into
The heart’s response to acute exercise is largely regulated by
how aging influences cardiovascular physiology and functional
the autonomic nervous system. During exercise, increased
capacity, limited access to tissue has been a major obstacle to
sympathetic tone augments both HR and contractility, while
elucidating the molecular mechanisms of aging that impair car-
concomitant parasympathetic withdrawal further enhances the
diac reserve. In this regard, rodent models have been particularly
chronotropic response. As the heart ages, however, its respon-
useful because of their relatively short lifespans, genetic manipu-
siveness to autonomic stimuli significantly diminishes. Evidence
Downloaded from http://ahajournals.org by on November 15, 2020

lability, and similar cardiac aging phenotypes to humans.5 On


in humans and animals suggests that these age-associated chang-
the basis of survival data, mice and rats, ≈24 months of age, are
es in cardiac autonomic regulation play important roles in de-
typically used to model older humans28 although even this pre-
clining cardiac reserve and exercise capacity seen with aging.40
specified age cutoff must be carefully considered given the wide
variation in lifespan across strains.29 In general, rodent hearts at Age-Associated Autonomic Dysregulation and
this age exhibit similar structural and functional phenotypes to Impaired Cardiac Reserve
older human hearts, including impaired contractile reserves, dia- Sympathetic dysregulation in the aged heart is primarily
stolic dysfunction, hypertrophy, fibrosis, and vascular stiffening.30 derived through a process known as β-adrenergic receptor
Importantly, despite having increased basal metabolic re- (β-AR) desensitization. With normal aging, circulating nor-
quirements and higher resting HR, rodents demonstrate com- epinephrine levels increase by 10% to 15% per decade.41 In
parable exercise physiology to humans, which can be reliably the heart, local norepinephrine levels also increase with age
assessed when careful attention is paid to exercise testing con- caused by diminished reuptake and increased tissue spill-
ditions.31 Continuous invasive hemodynamic monitoring in over.42 Greater β-AR occupancy by catecholamines triggers a
adult (3–4 months) mice has shown that they augment CO by compensatory mechanism in aged cardiomyocytes that results
≈2-fold (9.6±0.6 mL/min at rest to 18.9±0.9 mL/min at peak in desensitization of the postsynaptic machinery, and ultimate-
exercise) in response to acute exercise.32 The increased CO is ly blunted intracellular Ca2+ transients and impaired inotropic
primarily derived from a marked increase in HR (489±18 bpm and chronotropic responses to adrenergic stimulation.43,44
at rest to 798±9 bpm at peak exercise) and modest SV augmen- The mechanisms underlying β-AR desensitization in the
tation. Moreover, similar to humans, as rodents age, exercise aged heart are complex, with alterations occurring at multiple
capacity progressively declines. VO2max decreases by ≈28% in levels along the β-AR/G-protein/adenylyl cyclase (AC) path-
healthy 24-month-old C57BL/6J mice when compared with way. Reduced β-AR density has been reported in older human45
12-month-old mice.33 A similar pattern is seen in Fischer 344 × and rat46 hearts, implying that at least part of this process is
Brown Norway F1 (F344/BNF1) rats, which display 10% and modulated at the receptor level. In addition, numerous altera-
33% decreases in VO2max at 24 and 35 months, respectively, tions in downstream G proteins and AC catalytic units have been
when compared with 12-month-old rats.34 identified in the aging myocardium. Evidence from senescent
Even in rodents, defining intrinsic factors of aging that in- rats and guinea pigs has suggested that cardiac Gi protein lev-
fluence cardiac reserve and exercise capacity is difficult. Given els and pertussin toxin–mediated Gi ribosylation increase with
the central role of the autonomic nervous system on cardiac ex- age.47,48 However, other studies in humans and rats have dem-
ercise response, we devote a substantial amount of this review onstrated that Gi levels are unchanged in aged cardiomyocytes,
on autonomic dysregulation in the aged heart. Recent studies and furthermore, their reduced contractile response to adrenergic
282  Circulation Research  January 22, 2016

Figure.   Multiple mechanisms have been proposed for impaired cardiomyocyte function observed in aging, and how exercise
partially reverses their effects. (1) Diminished cardiac performance in the pathological hypertrophy of aging is linked to decreased
insulin-like growth factor-I (IGF-I)–phosphatidylinositol 3-kinase (PI3K)–AKT and β-adrenergic receptor (β-AR)–cAMP–protein kinase A
(PKA) signaling, decreased sarcoplasmic reticulum Ca2+–ATPase2a (SERCA2a) expression and activity and inefficient calcium handling,
and mitochondrial dysfunction secondary to excessive reactive oxygen species (ROS). (2) Exercise confers physiological hypertrophy and
cardioprotection in the form of enhanced β-adrenergic and IGF-1 signaling, SERCA2a activity and calcium handling, and mitochondrial
dynamics, the latter mediated largely through peroxisome proliferator-activated receptor γ coactivator (PGC)-1α. (3) These benefits of
Downloaded from http://ahajournals.org by on November 15, 2020

exercise potentially mitigate the effects of aging (Illustration Credit: Ben Smith).

stimulation cannot be rescued by inhibiting Gi with pertussin Given that exercise primarily mediates its effects on
toxin.45,46 Rather, these studies argue that age-related β-AR de- the heart through dynamic regulation of the autonomic sys-
sensitization is primarily mediated through diminished β-AR tem, it seems likely that these age-associated changes in β-
density, reduced Gs, and impairments in AC activity. adrenergic and muscarinic receptor pathways play important
Although the mechanisms responsible for the age-depen- roles in the impaired cardiac response to exercise in older
dent decline in cardiac β-AR responsiveness are not complete- adults. Notably, downregulation of β-AR density and activ-
ly understood, it is clear that this process results in impaired ity is seen in failing hearts from younger adults, who exhibit
cAMP production and protein kinase A (PKA) activity, which similar declines in cardiac reserve and exercise capacity.51
are necessary for augmenting intracellular Ca2+ transients and Likewise, acute β-AR blockade in healthy young adults reca-
enhancing cardiac contractility during exercise.49,50 Impaired pitulates the aging cardiac response to exercise with blunted
cAMP/PKA signaling may, in part, be because of persistent maximal HR, decreased myocardial contractility, and in-
activation of Ca2+/calmodulin kinase II (CaMKII), another creased end-diastolic volumes.59 Collectively, these data sup-
downstream effector of β-AR signaling. Interestingly, al- port an important functional role for altered sympathetic and
though constitutive β-AR stimulation leads to downregulation parasympathetic signaling in cardiac phenotypes associated
of PKA signaling, CaMKII activity remains high. Persistent with aging.
CaMKII activity can desensitize cardiomyocytes to PKA sig-
naling51,52 and, moreover, has been linked to apoptosis and Effects of Exercise Training on β-AR Desensitization
pathologic hypertrophy in failing cardiomyocytes.53,54 in the Aged Heart
Alterations in parasympathetic control of the aged heart There is modest evidence in older humans and rats indicating
have not been as extensively studied. In rats, the data have that exercise training can reverse, or resensitize, the aged heart
been conflicting with age-associated changes in the density to adrenergic stimuli and improve cardiac reserve.60 Nine
and function of cardiac muscarinic M2 receptors reported to be months of aerobic exercise in previously sedentary, older men
unchanged, decreased, or increased.55 In humans, the density (≈65 years) increased exercise capacity by 28%, in addition to
of cardiac M2 receptors seems to decline with age.56 Moreover, improving contractility and early diastolic filling rates at peak
aged human hearts demonstrate impaired chronotropic re- exercise.61 Importantly, these exercise-induced changes were
sponses to acute parasympathetic withdrawal, suggesting that completely abrogated by acute β1-receptor blockade, suggest-
impaired muscarinic receptor activity may contribute to the ing that the observed effects of training on the aged heart were
blunted HR response to exercise in the elderly.57,58 mediated through direct modulation of β-AR signaling.
Roh et al   Exercise and Cardiac Aging   283

Similar findings have been demonstrated in aged rats. proteins involved in excitation–contraction coupling. Decreased
Although 12 weeks of moderate intensity treadmill running in levels of sarcoplasmic reticulum Ca2+–ATPase2a (SERCA2a)
28-month-old Sprague–Dawley rats did not change β-AR density, are thought to be a primary mechanism for the prolonged Ca2+
it significantly decreased downstream Gi activity and enhanced transients in the aged myocardium.69–72 Cardiac SERCA2a
isoprenaline-stimulated AC activity.47 A follow-up study, in gene transfer in senescent rats restores diastolic function back
which 24-month-old Wistar–Kyoto rats were run at 70% to 80% to youthful levels.73 In addition, aged-associated alterations in
VO2max for 12 weeks, demonstrated that higher intensity training SERCA2a regulatory proteins, including phospholamban,74
increased β-AR density and AC activity in aged hearts, resulting PKA,49 and CaMKII,75 have also been documented in the aged
in enhanced responsiveness to adrenergic stimulation and resto- heart, with the direction of these changes expected to decrease
ration of inotropic, lusitropic, and chronotropic properties.62 SERCA2a activity and prolong Ca2+ transients. Evidence of age-
Although numerous differences in experimental conditions related changes in other proteins involved in cardiomyocyte Ca2+
are present between these 2 studies (Table 2), it is intriguing regulation, including the Na+/Ca2+ exchanger, ryanodine recep-
to hypothesize that exercise dose or subject age may have in- tors, and calsequestrin, has not been as consistent or would not
fluenced the varying effects of training on β-AR density in necessarily be expected to significantly alter Ca2+ transients.76
the aged hearts. Indeed, data from humans and rodents have
suggested that a threshold dose of exercise may be necessary Effects of Exercise Training on Ca2+ Handling in the
to generate significant changes in the heart.63–65 In adult rats, Aged Heart
direct comparison of moderate (65%–70% VO2max) and high- Whether exercise training can improve intracellular Ca2+ cy-
intensity treadmill running (85%–90% VO2max) demonstrated cling and performance of the aged heart is not entirely clear.
that higher intensity training not only improved exercise capac- In healthy young rodents, aerobic exercise training leads to
ity to a greater extent but also correlated with a dose-dependent faster rise and decay rates of Ca2+ transients in cardiomyo-
increase in cardiomyocyte hypertrophy, contractility/relax- cytes and subsequent improvements in systolic and diastolic
ation, and Ca2+ handling.65 Furthermore, age also seems to play functions.65,77 The mechanisms for these exercise-induced al-
a role in exercise-induced modulation of β-AR signaling. In terations in Ca2+ cycling in young hearts are potentially medi-
young animals, aerobic training decreases cardiac Gi activity, ated through more effective coupling of L-type Ca2+ channels
but generally has little to no effect on β-adrenergic/muscarinic and ryanodine receptors, increased SERCA2a and Na+/Ca2+
receptor densities or downstream AC activity.47 In fact, direct exchanger expression, enhanced SERCA2a function via tran-
comparison of high-intensity treadmill running (75% VO2max) sient CaMKII activation or phospholamban inhibition, and
in young (3 months) versus old (23 months) F344 rats showed improved myofilament Ca2+ sensitivity.77–79
Aerobic training studies in aged rodents suggest that these
Downloaded from http://ahajournals.org by on November 15, 2020

that adrenergic-stimulated AC activity was actually decreased


in young rats, while upregulated in older rats.66 benefits are not limited to young animals and seem to be primar-
ily driven by enhanced SERCA2a expression. Eight to 10 weeks
of treadmill running increase SERCA2a levels in the hearts of
Exercise and Ca2+ Regulation in the Aged Heart
24-month-old F344 rats.80 Furthermore, isolated cardiomyocytes
Calcium handling is regulated by β-adrenergic signaling in
from these rats display improved Ca2+ cycling and more rapid
cardiomyocytes and plays a central role in modulating cellu-
contractility and relaxation times.81 Twelve weeks of high-inten-
lar contraction and relaxation through excitation–contraction
sity treadmill running (70%–85% VO2max) in young (6 months)
coupling. Numerous age-related changes in key components
and old (24 months) F344/BNF1 rats also largely reverse impair-
of cardiomyocyte Ca2+ handling, however, impair both the
ments in early diastolic filling rates in the older cohort. This ef-
systolic and the diastolic properties of the aged heart.
fect is not seen in younger animals, suggesting that exercise has
Age-Associated Impairments in Ca2+ Handling specific modulatory effects on age-related impairments in active
To augment myocardial contractility, relaxation, and overall car- myocardial relaxation, potentially through improved Ca2+ cy-
diac performance during acute exercise, excitation–contraction cling.82 Swimming old (21 months) Wistar rats also induces sim-
coupling must be quickly modified within individual cardio- ilar increases in cardiac SERCA2a expression.83 However, other
myocytes to increase the rate of rise and decay of intracellular studies have demonstrated that SERCA2a and other related Ca2+
Ca2+ transients. In young cardiomyocytes, peak contractions channels (ie, ryanodine receptor, Na+/Ca2+ exchanger) are not
and Ca2+ transients increase and decay more rapidly at higher increased in aged rodents by aerobic training.84,85 Notably, these
stimulation frequencies.67,68 Although aged cardiomyocytes dis- latter studies were done at significantly lower exercise intensities
play similar peak contractions at slow stimulation rates, they (Table 2), emphasizing the importance of evaluating exercise
produce much smaller increases in peak Ca2+ transients and cell protocols in interpreting results of training.
shortening at more rapid pacing rates.68 In addition, rates of Ca2+
decay are significantly prolonged in aged cardiomyocytes when Exercise and Age-Related Cardiac
compared with younger cells. At an organ level, these findings Hypertrophy
translate to preserved systolic function under resting conditions, Cardiac hypertrophy, a composite of cardiomyocyte growth
but prolonged myocardial relaxation (a hallmark of age-related and increased extracellular matrix deposition, is a hallmark
diastolic dysfunction) and impairments in the ability to aug- feature of cardiac aging86,87 and is associated with diastolic
ment contractility at the faster HR elicited by exercise. dysfunction, HF, and mortality in the elderly.88,89 Although
Deficits in intracellular Ca2+ handling in aged cardiomyo- age-related vascular remodeling undoubtedly influences car-
cytes are largely derived from age-associated changes in the diomyocyte growth in the aged heart, both human and animal
284  Circulation Research  January 22, 2016

Table 2.  Summary of Studies in Aging Rodent Models Evaluating the Effects of Aerobic Exercise Training on Cardiac Aging
Phenotypes
Aging Animal Model Exercise Training Effects of Exercise Training
Cardiac (Compared With Sedentary
Parameter Species Strain Age, mo Type Protocol Duration, wk Control) References
Autonomic Rat Sprague– 28 Treadmill running, RSP 30 min/d, 20 m/min 12 β-AR density (NC), M-R density Böhm et al47
regulation Dawley (NC), Gi (↓), AC activity (↑)
Rat Wistar–Kyoto 24 Treadmill running, CP 45 min/d, 17 m/min, 15% 12 β-AR density (↑), AC activity (↑) Leosco et al62
(70%–85% VO2max)
Rat F344 23 Treadmill running, RDP 75% VO2max 9 Gs activity (NC), AC activity (↑) Scarpace et al66
Ca handling
2+
Rat F344 23–24 Treadmill running, RDP 60 min/d, 16 m/min, 5% 8–10 SERCA2a (↑), contractility (↑) Tate et al80,81
Rat F344/BNF1 24 Treadmill running, CP 45 min/d 17 m/min, 15% 12 Early diastolic filling (↑) Brenner et al82
(70%–85% VO2max)
Rat F344BN 29 Treadmill running, RSP 60 min/d, 5→10m/min, 10% 20–28 SERCA2a (NC), RyR (NC), Ca2+ Thomas et al84
cycling (NC)
Rat Wistar 21 Swimming, CP 90 min/d, 35–37°C 8 SERCA2a (↑), contractility (↑) Iemitsu et al83
Mouse C57BL/6 24 Treadmill running, CP 15 min/d, 15 m/min 10 (3 d/wk) SERCA2a (NC), NCX (NC) Walton et al85
Mouse C57BL/6 12 Treadmill running, CP 15 min/d, 15 m/min 52 (3 d/wk) SERCA2a (↓), NCX (NC) Walton et al85
Hypertrophy Rat F344/BNF1 24 Treadmill running, CP 45 min/d, 17 m/min, 15% 12 HW/TL (NC) Brenner et al82
(70%–85% VO2max)
Rat F344 25 Treadmill running, 20→60 min/d, 4→15 m/min 12 LV (NC), BW (↓), LV/BW (NC), Choi et al127
RSP, RDP (70%–75% VO2max) BP (NC)
Rat F344/BNF1 29 Treadmill running, RSP 60 min/d (in 6×10 min reps) 20–28 HW (↓), BW (↓), HW/BW (↑) Wright et al126
5→10 m/min, 10%
Rat F344BN 29 Treadmill running, RSP 60 min/d, 5→10 m/min, 10% 20–28 HW (↓) Thomas et al84
Rat F344 24 Treadmill running, CP 60 min/d, 15 m/min, 15% 12 CM size (↓), apoptosis (↓) Kwak et al130
(75% VO2max)
Rat Wistar–Kyoto 21 Treadmill running, CP 60 min/d (50%–60% VO2max) 13 CM size (NC), LV/TL (NC), BP Rossoni et al131
Downloaded from http://ahajournals.org by on November 15, 2020

(NC)
Rat Wistar 18 Treadmill running, CP 60 min/d, 30 m/min 6 HW(↑), BW (↓), HW/BW (↑), Wang et al128
Rat Sprague– 28 Treadmill running, RSP 30 min/d, 20 m/min 12 HW/BW (NC) Böhm et al47
Dawley
Rat Wistar 21 Swimming, CP 90 min/d, 35–37°C 8 LV (NC), BW (↓), LV/BW (↑) Iemitsu et al83
Rat Sprague– 18 Swimming, RDP 20→60 min/d, 25±2°C 12 HW (↓), HW/TL (↓) Liao et al99
Dawley
Rat Sprague– 18 Swimming, RDP 20→60 min/d, 25±2°C 12 LV (↓), LV/TL (↓), apoptosis (↓) Lai et al134
Dawley
Rat Wistar 21 Swimming, CP 90 min/d, 35–37°C 8 CM size (NC), LV (NC), BP (NC) Iemitsu et al132
Mouse C57BL/6 24 Treadmill running, CP 15 min/d, 15 m/min 10 (3 d/wk) CM size (↑), HW/BW (↑), HW/ Walton et al85
TL (NC)
Mouse C57BL/6 12 Treadmill running, CP 15 min/d, 15 m/min 52 (3 d/wk) CM size (↑), HW/BW (NC), HW/ Walton et al85
TL (NC)
Mouse PolG mutator 3 Treadmill running, CP 45 min/d, 15 m/min 20 (3 d/wk) HW (↓), Wall thickness (↓) Safdar et al172
Mouse C57BL/6 6–18 Swimming, CP 90-min session ×2/d, 4 LV (↑), LV/BW (↑) Derumeaux et al129
30–32°C
Fibrosis Rat F344/BNF1 29 Treadmill running, RSP 60 min/d (in 6×10 min reps), 20–28 Fibrosis (↓), collagen cross Wright et al126
5→10 m/min, 10% linking (↓)
Rat F344 25 Treadmill running, 20→60 min/d, 4 →15 m/ 12 Fibrosis (NC), collagen cross- Choi et al127
RSP, RDP min (70%–75% VO2max) linking (↓), passive stiffness (↓)
Rat Sprague– 18 Swimming, RDP 20→60 min/d, 25±2°C, 12 Fibrosis (↓) Liao et al99
Dawley
Mouse C57BL/6 24 Treadmill running, CP 15 min/d, 15 m/min 10 (3 d/wk) Fibrosis (NC) Walton et al85
Mouse C57BL/6 12 Treadmill running, CP 15 min/d, 15 m/min 52 (3 d/wk) Fibrosis (↓) Walton et al85
Mouse C57BL/6 6–18 Swimming, CP 90-min session ×2/d, 4 Fibrosis (↓), contractility (↑), Derumeaux et al129
30–32°C diastolic function (NC)

(Continued)
Roh et al   Exercise and Cardiac Aging   285

Table 2.  Continued


Aging Animal Model Exercise Training Effects of Exercise Training
Cardiac (Compared With Sedentary
Parameter Species Strain Age, mo Type Protocol Duration, wk Control) References

Mitochondrial Rat Wistar 18 Treadmill running, CP 60 min/d, 30 m/min, 6 Cardiac mito respiration (↑), Wang et al128
function ROS (↓)
Rat Sprague– 5 wk Treadmill running, RSP 30 min/d, 4.2 m/min→12 m/ 36 Cardiac PGC1α (↑), SIRT1 (↑), Bayod et al181
Dawley min at 1 m/min/30 s mito biogenesis (NC),
Mouse PolG mutator 3 Treadmill running, CP 45 min/d, 15 m/min 20 (3 d/wk) Cardiac mtDNA, mito Safdar et al172
respiration (↑),
cardiac mtDNA (↑)
Grade is 0% unless specified. Training frequency is 5 days/wk unless specified. (↑) indicates increase; (↓), decrease; β-AR, β-adrenergic receptor; AC, adenylyl
cyclase; BP, blood pressure; BW, body weight; CM, cardiomyocyte; CP, constant protocol; HW, heart weight; LV, left ventricular weight; M-R, muscarinic receptor; NC,
No change; NCX, Na+/Ca2+ exchanger; mito, mitochondrial; PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1α; RDP, ramped duration protocol;
ROS, reactive oxygen species; RSP, ramped speed protocol; RyR, ryanodine receptor; SERCA2a, sarcoplasmic reticulum Ca2+–ATPase; SIRT1, silent information regulator;
TL, tibial length; and VO2max, maximum oxygen consumption.

studies indicate that mechanisms independent of changing he- growth in aging hearts. Similarly, cardiac-specific suppression
modynamics also contribute.86,90 Cardiomyocyte hypertrophy of PI3K,103 as well as systemic mammalian target of rapamy-
in the aged heart may, in part, be a compensatory reaction to a cin inhibition with rapamycin,104 reverses hypertrophy and
cumulative loss of myocytes with normal aging.91,92 Declining lipofuscin accumulation in aged murine hearts. The role of
regenerative potential in the aged heart93 seems insufficient to histone deacetylases, particularly NAD-dependent sirtuins,
counterbalance this loss. Although age-related hypertrophy has recently emerged as important regulators of age-related
minimizes myocardial wall stress and can help maintain over- cardiac hypertrophy and longevity, and will be discussed in
all cardiac function, at a cellular level, it can also be viewed detail later in this review.
as a marker of increased stress and altered homeostasis and Most recently, heterochronic parabiosis studies in mice
is generally felt to be a pathologic process associated with have suggested that there may, in fact, be age-specific mecha-
increased apoptosis, impaired Ca2+ regulation, and defective nisms of cardiomyocyte hypertrophy. Using a novel aptamer-
macroautophagy.94–96 based proteomics screen in this mouse model, Loffredo et al105
Downloaded from http://ahajournals.org by on November 15, 2020

found that systemic levels of growth differentiation factor-11


Mechanisms of Age-Related Cardiac Hypertrophy
(GDF11), a secreted member of the transforming growth
Many of the molecular mechanisms underlying cardiomyocyte
factor-β superfamily, decline with normal aging. Interestingly,
hypertrophy in the aged heart seem similar to the intracellular
signaling pathways that drive pathologic growth in hyperten- restoration of GDF11 levels in 24-month-old C57BL/6 mice
sion and HF.6,97 Chronically activated neurohormonal systems, reversed age-related cardiomyocyte hypertrophy and im-
including the adrenergic, endothelin, and renin–angiotensin– proved SERCA2a expression in the heart. Recent work by
aldosterone systems, along with increased workload and bio- Smith et al,106 however, found that GDF11 therapy did not
mechanical strain on the remaining cardiomyocytes, stimulate alter cardiomyocyte hypertrophy in old C57BL/6 mice and,
numerous growth pathways, including the mitogen-activated moreover, did not affect cardiac function. The reason for these
protein kinases, histone deacetylases, calcineurin/nuclear fac- differing results is currently unclear as similar GDF11 inter-
tor of activated T cells, and insulin-like growth factor-I (IGF- ventions and aged murine strains were used in both studies.
I)–phosphatidylinositol 3-kinase (PI3K)–AKT–mammalian Interestingly, GDF11 shares many structural and func-
target of rapamycin pathways. For a detailed discussion of tional properties with myostatin, another transforming growth
these growth pathways in the heart, we refer the interested factor -β superfamily member (also known as GDF8). Aging
reader to an excellent review by Heineke and Molkentin.98 studies in germline myostatin knockout mice have suggested
Although many of these signaling pathways, including that although systemic myostatin inhibition induces mod-
p38 MAPK, c-jun N-terminal kinase, extracellular signal- est cardiac hypertrophy in senescent mice, it also decreases
regulated kinase-1/2, calcineurin/nuclear factor of activated myocardial fibrosis and improves systolic function.107 Indeed,
T cells, and PI3K/Akt/ mammalian target of rapamycin, are both hearts and isolated cardiomyocytes from aged germline
upregulated in aging rodent hearts,6,99,100 whether they are di- knockouts demonstrate improved β-adrenergic responsive-
rectly regulated by aging and what the relative contribution of ness, Ca2+ handling, and enhanced contractility to sympa-
each of these pathways in age-related cardiac hypertrophy is thetic stimulation.107–109 Taken together, these data suggest that
not clearly defined. Cardiac specific gain- and loss-of-func- myostatin inhibition may induce physiologic, as opposed to
tion studies have provided supportive evidence for a direct pathologic, hypertophy in the aged heart. As further evidence,
role of neurohormonal pathways in age-related cardiomyo- chronic pressure overload through transverse aortic constric-
cyte hypertrophy. Genetic ablation of cardiomyocyte endo- tion (TAC) does not alter the hypertrophic response in cardi-
thelin-A receptors attenuates age-associated cardiomyocyte ac-specific myostatin knockout mice,110 and likewise, GDF11
hypertrophy,101 whereas cardiomyocyte-specific β-AR1102 or therapy has no effect on TAC-induced pathologic hypertro-
angiotensin II97 overexpression induces significant myocyte phy.105 Recent work by Egerman et al111 has highlighted the
286  Circulation Research  January 22, 2016

difficulty in distinguishing between myostatin and GDF11 in proving to be a particularly potent regulator of cardiomyocyte
some of the currently available assays, emphasizing the need growth and proliferation in vitro. Subsequent in vivo studies
for further study to understand the potentially overlapping showed that miRNA-222 was required for exercise-induced
roles of these closely related peptides in cardiac aging. hypertrophy, and its forced expression protected against ad-
verse remodeling after ischemic injury. These results not only
Mechanisms of Exercise-Induced Cardiac demonstrate that integrating different exercise regimens can
Hypertrophy be a particularly robust approach to identifying critical bio-
Similar to aging and HF, exercise can induce a dramatic in- logical networks but also underscore the differential responses
crease in cardiac mass that is predominantly mediated by car- elicited by distinct protocols and thus the challenges in com-
diomyocyte growth.112,113 However, unlike age-related cardiac paring the data from 1 regimen in isolation.
hypertrophy, exercise elicits a more physiologic growth that
is felt to be cardioprotective. Not only are the outcomes of Effects of Exercise Training on Age-Related Cardiac
these 2 kinds of cardiac growth different, but the underlying Hypertrophy
molecular mechanisms are also quite distinct.114 The concept of distinct forms of cardiac hypertrophy is par-
Exercise-induced hypertrophy is mediated largely through ticularly relevant in the aging heart. As opposed to the case
increased IGF-1 signaling in the heart.115 Cardiac-specific in young animals, in which aerobic training generally induc-
IGF-1 receptor knockout mice do not develop cardiac hyper- es some degree of hypertrophy,124 training studies in senes-
trophy in response to exercise, suggesting that initial IGF-1 cent animals have shown extensive variability in the cardiac
signaling is necessary for exercise-induced cardiac growth.116 growth response to exercise,126–133 with a substantial number
Stimulated IGF-1 receptors subsequently activate PI3K, a of studies indicating that it can paradoxically reverse aged-
family of heterodimeric kinases that regulate membrane lipid related hypertrophy (Table 2).
phosphoinositides. Cardiac-specific expression of a dominant- A small subset of these studies has evaluated the effects of
negative PI3K 110α isoform also inhibits exercise-induced exercise training on cardiomyocyte growth in the aged heart.
cardiac growth.117,118 Similarly, germline deletion of the PI3K- Kwak et al130 trained young (3 months) and old (24 months)
effector, Akt1, abolishes exercise-induced cardiac hypertro- F344 rats on a high-intensity running protocol (75% VO2max)
phy.119 Conversely, forced overexpression of Akt in the heart for 12 weeks. Although training induced cardiomyocyte hy-
protects cardiomyocytes from hypoxic injury and apopto- pertrophy in the young rats, it resulted in regression of car-
sis,120,121 supporting the notion that Akt could contribute to ex- diomyocyte size (69% decrease in cross-sectional area) in
ercise-induced cardioprotection. Taken together, these studies the aged cohort. Alternatively, low-moderate intensity tread-
Downloaded from http://ahajournals.org by on November 15, 2020

collectively establish the IGF-1/PI3K/Akt signaling pathway mill running or swimming did not affect cardiomyocyte size
as a central mediator of the cardiac exercise response. in 21-month-old Wistar–Kyoto or spontaneously hyperten-
Genome-wide transcriptome analyses comparing exer- sive rats, despite reductions in blood pressure in the latter
cised hearts to hearts subjected to TAC also demonstrated group.131,132 Moreover, 10 weeks of low-intensity treadmill
distinct sets of transcriptional regulators regulated in physio- running were sufficient to induce cardiomyocyte hypertrophy
logical and pathological hypertrophy.122 Moreover, this screen in aged (24–26 months) C57BL/6 mice.85
identified a transcriptional pathway downstream of CCAAT/ Differences in training protocols and animal models make
enhancer-binding protein beta (C/EBPβ), a member of the it inherently difficult to directly compare studies (Table 2).
basic helix-loop-helix family of DNA-binding transcription In addition, only a few studies adequately address the blood
factors, as downregulated with exercise. Reduction of C/ pressuring–lowering effects of exercise, which are particu-
EBPβ in vitro and in vivo was sufficient to recapitulate many larly relevant in assessing cardiac growth in the context of
of exercise-related phenotypes including a similar gene ex- aging. However, collectively these data again seem to indi-
pression profile, cardiomyocyte hypertrophy, and protection cate that training intensity and age are critical determinants
against HF. Notably, this pathway intersects with Akt signal- in exercise-mediated modulation of cardiac aging phenotypes,
ing. Forced overexpression of C/EBPβ in cardiomyocytes specifically with repression of age-related cardiac hypertro-
blocks Akt1-induced expression of genes characteristic of phy generally occurring in older animals subjected to higher
physiologic hypertrophy, and conversely, Akt1 overexpression intensity protocols.
downregulates C/EBPβ expression. Given the discrepancies among studies, it is not surprising
In addition, work from our laboratory and others have that the molecular basis for the potentially disparate effects
shown that micro-RNAs (miRNAs) and exercise protocols of exercise-mediated growth in young versus old hearts is not
play important roles in the cardiac growth response to exer- entirely clear. It is postulated that exercise’s cytoprotective ef-
cise.123 Exercise protocols vary widely, and the growth re- fects may improve survival in senescent cardiomyocytes, thus
sponses of the heart to different experimental designs are not decreasing the stimulus for reactive pathologic hypertrophy.
identical (Table 2).124 In comparing the differential expression Indeed, hearts from exercise-trained aged rats demonstrate re-
of miRNAs in the hearts of mice that were exercised with ductions in numerous apoptotic indices that are elevated in the
forced swimming versus voluntary wheel running, hearts of aging myocardium.130,133,134 However, whether these exercise-
swum mice had 55 differentially expressed miRNAs when induced changes translate to less cell death and diminish the
compared with sedentary controls, whereas hearts from wheel trigger for pathologic growth in the aged heart is not proven.
run mice had 124 such miRNAs.125 Sixteen miRNAs were con- Moreover, recent work has shown that proapoptotic caspase
cordantly regulated in both exercise models, with miRNA-222 pathways can directly induce pathologic growth in adult
Roh et al   Exercise and Cardiac Aging   287

cardiomyocytes,135 suggesting an alternative mechanism by the various signaling pathways involved in age-related car-
which exercise-induced inhibition of apoptotic pathways may diomyocyte hypertrophy is still largely unknown and remains
suppress pathologic growth in the aged heart. a fertile area for future research.
The underlying signaling mechanisms by which exercise
potentially improves survival of aged cardiomyocytes may Exercise and Mitochondrial Dysfunction in the
be related to the cardioprotective effects of the IGF-1/PI3K/ Aged Heart
Akt pathway. Cardiac-specific overexpression of IGF-1136, The heart requires an enormous amount of energy, primar-
PI3K,137 and Akt1121 have all been shown to improve cardio- ily derived from fatty acid oxidation and subsequent ATP
myocyte survival in adult mouse hearts exposed to either TAC production within mitochondria. The ability to fulfill this en-
or ischemic injury. Importantly, multiple studies have also ergy requirement, especially under stress, is impaired in the
demonstrated that aerobic exercise increases Akt phosphory- aged heart largely due to defective mitochondria. The mito-
lation in senescent rodent hearts,99,132,138 to a similar albeit a chondrial theory of aging is a decades-old idea,149 with the
lesser extent.132,134 It is plausible that partially restored levels underlying premise that oxidative stress increases with age
of Akt activity in exercised aged hearts are sufficient to en- and causes a gradual accumulation of mitochondrial damage
hance cell survival and suppress pathologic growth pathways, and electron transport chain dysfunction.150–153 Increased lev-
but insufficient to promote physiologic growth. els of free radicals in the aged heart lead to impaired mito-
Ultimately, the variability in cardiac growth responses
chondria, which in turn produce more reactive oxygen species
to exercise between young and old animals likely stems
(ROS) resulting in a downward spiral in cardiac performance.
from differences in the substrate of a young versus senes-
Early studies in Drosophila overexpressing ROS scavenging
cent heart, with apoptotic and pathologic hypertrophy path-
enzymes and in mice with enhanced resistance to oxidative
ways constitutively activated in the latter. Indeed, when
stress demonstrate increased lifespan, as do models of caloric
young (3 months) and old (18 months) rats are subjected to
restriction.154–156 However, more recent studies suggest that the
similar 12-week swimming protocols, apoptotic markers,
relationship between ROS and mitochondrial DNA (mtDNA)
mitogen-activated protein kinase, and calcineurin/nuclear
damage leading to aged phenotypes is not so straightforward,
factor of activated T-cell expression decrease in old hearts,
but remain unchanged or increased in young hearts, despite and that widespread ROS elevation in somatic tissues may not
increased Akt activity in both groups.99,134 Interestingly, be the root cause of aging. In fact, certain levels of ROS may
germline Akt1 knockout mice show an exaggerated growth be instrumental for maintaining tissue homeostasis and regen-
response to TAC, suggesting that Akt signaling may be ca- erative potential. In the context of this ongoing debate, we will
discuss how the interplay between ROS and mitochondrial
Downloaded from http://ahajournals.org by on November 15, 2020

pable of directly suppressing pathologic growth pathways


in the aged heart.119 Although the mechanisms by which function impacts cardiac performance during aging, and the
this occurs in cardiomyocytes are unknown, in other cell mechanisms by which exercise may play a beneficial role in
types, Akt has been shown to inhibit numerous mitogen- restoring cardiac energetics.
activated protein kinase pathways (p38 and extracellular
Mitochondrial Dysfunction in Aging
signal-regulated kinase) implicated in pathologic cardiac
Senescence heralds an indisputable decline in mitochondrial
hypertrophy.139–141
function. mtDNA lacks protective histones and is in close
In addition to Akt signaling, it is important to note that
proximity to high levels of ROS, and thus is particularly sus-
exercise also modulates other growth pathways that may be
ceptible to oxidation.157 DNA mutation rate is 10- to 20-fold
particularly relevant to the aging heart. Acute treadmill run-
higher in mitochondria than in nuclei. The role of mtDNA
ning stimulates neuregulin production in skeletal muscle,142
damage in aging is dramatically revealed in Mutator mice.
which has demonstrated antiapoptotic effects on cardiomyo-
cytes through the ErbB family of tyrosine kinases and po- These animals harbor defective proofreading by the mtDNA
tentially downstream PI3K/AKT.143 Exercise also decreases polymerase gamma (PolG) and thus carry significant mtDNA
both skeletal muscle and cardiac myostatin levels in humans deletions and 5-fold more point mutations.158,159 Although this
and rodents with pathologic hypertrophy.144,145 Although the degree of mtDNA damage exceeds that observed in normal
precise role of myostatin and its close homologue GDF11 in aging, no genetic model can represent all the progressive
age-related cardiac hypertrophy awaits clarification, it may changes that mitochondria undergo. Nevertheless, these mice
be that exercise-induced inhibition of this pathway induces display global attributes of premature aging, and cardiac se-
a similar pattern of Akt activity that could inhibit pathologic nescence in the form of hypertrophy, increased fibrosis, and
growth pathways in the aged heart. Indeed, in vitro studies impaired systolic and diastolic functions by 8 months of age.
have demonstrated that myostatin inhibition drives cardio- Tissues of PolG Mutator mice show decreased levels of mi-
myocyte growth through Akt activation.146 Furthermore, al- tochondrial biogenesis, diminished respiratory capacity, and
though physiologic versus pathologic growth pathways are increased apoptosis.159,160 Interestingly, although Mutator mice
largely distinct, there is some overlap. For example, while do not show increased levels of ROS,159,161 the expression of
calcineurin/nuclear factor of activated T-cell signaling is pri- a mitochondrial-specific catalase partially reverses their car-
marily a regulator of pathologic growth of the heart, there diac findings.162 This supports the idea that ROS reduction
is evidence indicating that it also mediates cardioprotective ameliorates the accumulation of mtDNA mutations, and that
effects and may be necessary in certain physiological growth oxidative stress specifically in mitochondria is a major factor
settings.147,148 Ultimately, how exercise dynamically regulates leading to the progerian phenotype.
288  Circulation Research  January 22, 2016

Given that Mutator mice do not reveal dramatically altered mitochondrial gene transcription and replication. Long- and
levels of oxygen free radicals or oxidative damage, attention short-term endurance exercise increases PGC-1α expression
has turned toward possible mechanisms of premature aging in cardiac and skeletal muscles.181–184 Exercise, at least in part
that rely less on global increases in ROS, but on subtle altera- through β-adrenergic signaling, augments PGC-1α activity
tions in subpopulations of cells. Cellular dysfunction or de- and nuclear translocation, resulting in greater mitochondrial
mise may result when a certain threshold of mutational burden biogenesis.183,185 In contrast, PGC-1 shows reduced muscle
is crossed, or if DNA damage of critical subunits of mitochon- expression in aging, coincident with decreased mitochondrial
drial metabolism results in ineffective respiration, resulting function.186,187 The lower mtDNA content, impaired complex
in a heterogeneous response within the myocardium.153,163 IV activity, and decreased ejection fraction of PolG hearts are
Mitochondrial decline creating a cellular mosaic in aged hu- largely corrected by the forced expression of PGC-1α.188 The
man hearts was first exemplified by the arbitrary distribution cardioprotective effects of PGC-1 are likely mediated in part
of cardiomyocytes with undetectable cytochrome c oxidase through its ROS-lowering effects, as PGC-1α induces GPx1
activity,164 and similarly observed in mice lacking mitochon- and superoxide dismutase 2 in models of neurodegeneration.189
drial transcription factor A in heart and skeletal muscles.165 It is unclear whether the benefit comes solely from increased
The latter develop dilated cardiomyopathy and lethal conduc- levels of PGC-1α in the heart or from a systemic contribution
tion blocks. A mosaic of mitochondrial dysfunction in hearts is from concurrent PGC-1α overexpression in skeletal muscle.
also observed in mice with a dominant-negative, cardiac-spe- Aged mice carrying this muscle creatine kinase (MCK)-PGC-
cific mitochondrial helicase, which accelerates the accumula- 1α transgene have improved whole body metabolism in the
tion of mtDNA deletions.166 Aging mice carrying this mutant form of greater insulin sensitivity and reduced sarcopenia and
gene develop diffuse respiratory deficiency that ultimately chronic inflammation.190
manifests as arrhythmias, possibly secondary to aberrant Ca2+ Exercise enhances antioxidant defenses and restores redox
handling. A heterogeneous response to mtDNA mutation that homeostasis in the aging myocardium via NFE2L2 as well.
ultimately contributes to the progeroid phenotype may also NFE2L2 trans-activates genes of the antioxidant response191
derive from stem cell reservoirs that are particularly vulner- and is coactivated by PGC-1α during oxidative stress.192,193
able to ROS elevation. Tissue-specific depots of somatic stem The loss of redox capacity seen in aging is similarly observed
cells are crucial for repair and regeneration.167 PolG Mutator in hearts lacking NFE2L2.194,195 Although aging hearts ex-
mice show impaired neural and hematopoietic progenitor cell hibit reduced NFE2L2-dependent antioxidant mechanisms,
self-renewal as early as embryogenesis, which can be rescued both acute exercise and several weeks of moderate exercise
by administering the antioxidant N-acetylcysteine to pregnant training in aged mice increase NFE2L2 activity and induc-
Downloaded from http://ahajournals.org by on November 15, 2020

females.168 tion of its target pathways to near-normal levels seen in young


counterparts.194,196
Benefits of Exercise Training on Mitochondrial SIRTs are NAD+-dependent deacetylases that regulate
Preservation cellular health and longevity. As sensors of nutrient flux
Substantial evidence supports a role for exercise in mitochon- and redox states, they help to maintain metabolic homeo-
drial preservation.169,170 Four weeks of voluntary treadmill stasis. Two members in particular, SIRT1 and SIRT3, play
running in 7- to 9-week-old mice increase the mitochondri- important roles in both aged myocardium and antioxidant
al number and volume in their left ventricles.171 Exercising pathways. SIRT1 activates hypertrophic pathways via ac-
Mutator mice on a treadmill for 5 months attenuates their car- tivation of Akt, and its forced high expression produces
diac hypertrophy and fibrosis, in addition to protecting against cardiac dysfunction.197 In contrast, more moderate levels
apoptosis and the decrease in complexes of the mitochondrial of SIRT1 transgenic expression reduce age-related hyper-
respiratory chain in the heart.172 As with catalase overexpres- trophy, fibrosis, and dysfunction, as well as damage from
sion, the cardioprotective benefits of exercise in PolG mice oxidative stress from paraquat.198,199 Mice lacking SIRT3
likely involve ROS detoxifying mechanisms. Indeed, exercise have the hallmarks of premature aging and show greater hy-
in a variety of tissues, including the heart, has been shown to pertrophy and fibrosis in response to the pressure overload
increase antioxidant capacity by augmenting ROS scavenging of transverse aortic banding, whereas SIRT3 overexpression
enzymes such as catalase, superoxide dismutase, and glutathi- confers resistance to hypertrophy driven by angiotensin-
one peroxidase.173–178 II.200,201 Like SIRT1, SIRT3 protects against oxidative stress,
An integral relationship exists between exercise and tran- in large part through FOXO3a-dependent mechanisms that
scriptional regulators that limit ROS levels. These factors induce superoxide dismutase and catalase.202 Notably, these
include the family of peroxisome proliferator-activated re- 2 sirtuins are upregulated during exercise in heart and skel-
ceptor γ coactivators, nuclear factor-erythroid-derived 2-like etal muscles and are positive modulators of PGC-1α activ-
2 (NFE2L2), and the sirtuin family (silent information regu- ity.134,203–208 Caloric restriction, likely in concert with SIRTs,
lators [SIRTs]). Peroxisome proliferator-activated receptor helps preserve energy handling in the aging heart and re-
γ coactivator (PGC)-1α and β, regulators of mitochondrial duces cardiomyocyte apoptosis.8 Like exercise, it induces
biogenesis and respiratory capacity, coactivate nuclear re- PGC-1α in the heart and leads to preserved mitochondrial
spiratory factor 1 and 2 and estrogen-related receptors in function during aging.209
the induction of genes important for oxidative phosphoryla- Even in the setting of a heterogeneous response to mtD-
tion and other mitochondrial processes.179,180 Chief among NA damage within aged myocardium, it is likely that exer-
these is mitochondrial transcription factor A, which controls cise nevertheless mitigates damage to discrete subsets of cells
Roh et al   Exercise and Cardiac Aging   289

that are more susceptible to the effects of ROS. In skeletal studies argue that exercise-mediated improvements in func-
muscle at least, moderate intensity endurance exercise in rats tional capacity in older adults are primarily derived from
protects against the age-associated loss of satellite cells.210 peripheral mechanisms of oxygen extraction in the skeletal
Interestingly, despite the large body of evidence supporting a muscle.
causal relationship between ROS and mitochondrial dysfunc- The reasons for these discrepancies are not entirely clear,
tion in cardiac senescence, nonspecific reduction of ROS has but as in the case of animal studies potentially stem from dif-
led to surprising results. In clinical trials, antioxidant dietary ferences in exercise protocols, techniques for measuring car-
supplements are not associated with reduced mortality, but diac structure/function, and the varying ages of participants.
rather, in the case of β carotene, vitamin A, and vitamin E, Cardiac senescence, as with other aging processes, is a pro-
increased mortality.211 Some studies paint a more complex gressive phenomenon. Thus, the often-used inclusion criteria
picture, suggesting that an exercise-induced increase in ROS for older adults as simply >65 years can yield variable results
signals to and activates endogenous mechanisms of antioxi- because a 65-year-old heart is often different from an 85-
dant defense. In both human and rat skeletal muscles, oral year old’s. Furthermore, with emerging data from aged ani-
administration of the antioxidant vitamin C reduces mitochon- mals (Table 2) indicating that a sufficient dose of exercise is
drial biogenesis induced by exercise and lowers the expres- likely necessary to alter established aging phenotypes in the
sion of PGC-1α, nuclear respiratory factor 1, mitochondrial heart,63,64 the requisite or optimal dose needed for older hu-
transcription factor A, and cytochrome c.212 The combination mans remains to be determined. Moreover, the intensity of ex-
of vitamins C and E likewise blunts exercise-mediated in- ercise used in animal studies may not be realistically achieved
creases in PGC-1α, PGC-1β, and ROS scavengers in skeletal by frail older adults with cardiovascular disease. Ultimately,
muscle in healthy human subjects.213 These studies underscore well-controlled, dose–response studies are needed to answer
the delicate balance between the harmful effects of excessive these questions. However, the growing body of exercise lit-
ROS that accelerates senescence and the requirement for some erature in aged animals provides unique insights into how ex-
basal level of ROS that maintains critical signaling pathways ercise can modulate the aging process in the heart, and thus,
and cellular homeostasis. In the aging heart, this concept of a framework for potentially identifying novel targets for treat-
mitohormesis surely plays a crucial role in conveying exer- ing age-related heart diseases.
cise’s benefits.
Conclusions
Can Exercise Reverse Cardiac Aging in The rapidly changing distribution of age in our population
Humans? demands a deeper understanding of cardiac senescence.
Downloaded from http://ahajournals.org by on November 15, 2020

As highlighted throughout this review, exercise training in Exercise testing has already provided valuable insights into
aged animal models has raised the exciting possibility that ex- how cardiac physiology changes with age, and with further
ercise can reverse cardiac aging phenotypes associated with refinements will inevitably be a powerful tool for generating
HF. Whether similar effects can be derived from exercise in and translating discoveries from preclinical animal models.
older humans, however, has yet to be defined. Although the impact of exercise training on human cardiac
Cross-sectional studies comparing sedentary and athletic aging phenotypes remains incompletely defined, emerging
older adults have suggested that lifelong physical activity is data from rodent models have suggested the exciting pos-
associated with less age-related changes in the heart.63,214–216 sibility that exercise can effectively modulate the aging
However, inherent limitations in cross-sectional analyses in- process in the heart. These studies hold great promise for
clude potential selection bias of fitter individuals and those identifying novel targets for age-specific, tailored therapies
adhering to healthier lifestyles leading to unrecognized con- in the older patient.
founding or even reverse causality in which individuals with
better cardiac function are more likely to be lifelong exercisers. Sources of Funding
Thus, it is impossible to conclude from such studies whether This work was supported by grants from the National Institutes of
lifelong exercise is the source of these changes. Furthermore, Health (A. Rosenzweig [R01HL110733 and R01HL122987], Dr Roh
whether exercise can actually reverse established age-related [T32HL073734], and J. Rhee [T32GM007592]). Roh was also sup-
myocardial changes, and if so, whether these changes are di- ported by a LaDue Fellowship Award from Harvard Medical School.
rectly causal in improving exercise capacity or cardiovascular A. Rosenzweig is a principal faculty member of the Harvard Stem
Cell Institute.
outcomes in the elderly, remain unknown.
Many small prospective studies have attempted to ad-
dress such questions by looking at the effects of exercise
Disclosures
None.
training on cardiac structure and function in previously
sedentary older adults, with or without HF. Although some
studies have suggested that training improves resting car-
References
1. Yazdanyar A, Newman AB. The burden of cardiovascular disease in the
diac parameters associated with HF in the elderly, includ- elderly: morbidity, mortality, and costs. Clin Geriatr Med. 2009;25:563–
ing diastolic dysfunction,217 systolic reserve capacity,61 and 577, vii. doi: 10.1016/j.cger.2009.07.007.
chronotropic incompetence,19 there are an equal number of 2. Go AS, Mozaffarian D, Roger VL, et al; American Heart Association
Statistics Committee and Stroke Statistics Subcommittee. Heart dis-
studies that have shown that training, while similarly im-
ease and stroke statistics–2013 update: a report from the American
proving exercise capacity, does not significantly alter any of Heart Association. Circulation. 2013;127:e6–e245. doi: 10.1161/
these cardiac aging phenotypes.24,218–220 Rather, these latter CIR.0b013e31828124ad.
290  Circulation Research  January 22, 2016

3. Fang J, Mensah GA, Croft JB, Keenan NL. Heart failure-related hospital- with preserved ejection fraction. J Am Coll Cardiol. 2010;56:845–854.
ization in the U.S., 1979 to 2004. J Am Coll Cardiol. 2008;52:428–434. doi: 10.1016/j.jacc.2010.03.077.
doi: 10.1016/j.jacc.2008.03.061. 26. Proctor DN, Joyner MJ. Skeletal muscle mass and the reduction of VO2max
4. Lakatta EG. Arterial and cardiac aging: major shareholders in cardiovas- in trained older subjects. J Appl Physiol (1985). 1997;82:1411–1415.
cular disease enterprises: part III: cellular and molecular clues to heart and 27. Fleg JL, Morrell CH, Bos AG, Brant LJ, Talbot LA, Wright JG,

arterial aging. Circulation. 2003;107:490–497. Lakatta EG. Accelerated longitudinal decline of aerobic capacity in
5. Loffredo FS, Nikolova AP, Pancoast JR, Lee RT. Heart failure with pre- healthy older adults. Circulation. 2005;112:674–682. doi: 10.1161/
served ejection fraction: molecular pathways of the aging myocardium. CIRCULATIONAHA.105.545459.
Circ Res. 2014;115:97–107. doi: 10.1161/CIRCRESAHA.115.302929. 28. Turturro A, Witt WW, Lewis S, Hass BS, Lipman RD, Hart RW.
6. Dai DF, Santana LF, Vermulst M, et al. Expression of catalase tar- Growth curves and survival characteristics of the animals used in
geted to mitochondria attenuates murine cardiac aging. Circulation. the Biomarkers of Aging Program. J Gerontol A Biol Sci Med Sci.
2009;119:2789–2797. 1999;54:B492–B501.
7. Bodyak N, Kang PM, Hiromura M, Sulijoadikusumo I, Horikoshi N, 29. Yuan R, Tsaih SW, Petkova SB, Marin de Evsikova C, Xing S, Marion MA,
Khrapko K, Usheva A. Gene expression profiling of the aging mouse car- Bogue MA, Mills KD, Peters LL, Bult CJ, Rosen CJ, Sundberg JP, Harrison
diac myocytes. Nucleic Acids Res. 2002;30:3788–3794. DE, Churchill GA, Paigen B. Aging in inbred strains of mice: study design
8. Lee CK, Allison DB, Brand J, Weindruch R, Prolla TA. Transcriptional and interim report on median lifespans and circulating IGF1 levels. Aging
profiles associated with aging and middle age-onset caloric restriction Cell. 2009;8:277–287. doi: 10.1111/j.1474-9726.2009.00478.x.
in mouse hearts. Proc Natl Acad Sci U S A. 2002;99:14988–14993. doi: 30. Lakatta EG, Sollott SJ. Perspectives on mammalian cardiovascular ag-
10.1073/pnas.232308999. ing: humans to molecules. Comp Biochem Physiol A Mol Integr Physiol.
9. Kitzman DW, Higginbotham MB, Cobb FR, Sheikh KH, Sullivan MJ. 2002;132:699–721.
Exercise intolerance in patients with heart failure and preserved left ven- 31. Platt C, Houstis N, Rosenzweig A. Using exercise to measure and

tricular systolic function: failure of the Frank-Starling mechanism. J Am modify cardiac function. Cell Metab. 2015;21:227–236. doi: 10.1016/j.
Coll Cardiol. 1991;17:1065–1072. cmet.2015.01.014.
10. Kappagoda T, Amsterdam EA. Exercise and heart failure in the elderly. 32. Lujan HL, DiCarlo SE. Cardiac output, at rest and during exercise, before
Heart Fail Rev. 2012;17:635–662. doi: 10.1007/s10741-011-9297-4. and during myocardial ischemia, reperfusion, and infarction in conscious
11. Myers J, Prakash M, Froelicher V, Do D, Partington S, Atwood JE.
mice. Am J Physiol Regul Integr Comp Physiol. 2013;304:R286–R295.
Exercise capacity and mortality among men referred for exercise testing. doi: 10.1152/ajpregu.00517.2012.
N Engl J Med. 2002;346:793–801. doi: 10.1056/NEJMoa011858. 33. Schefer V, Talan MI. Oxygen consumption in adult and AGED C57BL/6J
12. Blair SN, Kampert JB, Kohl HW III, Barlow CE, Macera CA, Paffenbarger mice during acute treadmill exercise of different intensity. Exp Gerontol.
RS Jr, Gibbons LW. Influences of cardiorespiratory fitness and other pre- 1996;31:387–392.
cursors on cardiovascular disease and all-cause mortality in men and 34. Olfert IM, Balouch J, Mathieu-Costello O. Oxygen consumption dur-
women. JAMA. 1996;276:205–210.
ing maximal exercise in Fischer 344 x Brown Norway F1 hybrid rats. J
13. Laughlin MH. Cardiovascular response to exercise. Am J Physiol.

Gerontol A Biol Sci Med Sci. 2004;59:801–808.
1999;277:S244–S259.
35. Koch LG, Britton SL, Wisløff U. A rat model system to study complex
14. Vella CA, Robergs RA. A review of the stroke volume response to up-
disease risks, fitness, aging, and longevity. Trends Cardiovasc Med.
right exercise in healthy subjects. Br J Sports Med. 2005;39:190–195. doi:
2012;22:29–34. doi: 10.1016/j.tcm.2012.06.007.
10.1136/bjsm.2004.013037.
Downloaded from http://ahajournals.org by on November 15, 2020

36. Bye A, Langaas M, Høydal MA, Kemi OJ, Heinrich G, Koch LG, Britton
15. Gulati M, Shaw LJ, Thisted RA, Black HR, Bairey Merz CN, Arnsdorf
SL, Najjar SM, Ellingsen Ø, Wisløff U. Aerobic capacity-dependent dif-
MF. Heart rate response to exercise stress testing in asymptomatic women:
ferences in cardiac gene expression. Physiol Genomics. 2008;33:100–109.
the st. James women take heart project. Circulation. 2010;122:130–137.
doi: 10.1152/physiolgenomics.00269.2007.
doi: 10.1161/CIRCULATIONAHA.110.939249.
37. Koch LG, Kemi OJ, Qi N, et al. Intrinsic aerobic capacity sets a divide
16. Brubaker PH, Kitzman DW. Chronotropic incompetence: causes, con-
for aging and longevity. Circ Res. 2011;109:1162–1172. doi: 10.1161/
sequences, and management. Circulation. 2011;123:1010–1020. doi:
CIRCRESAHA.111.253807.
10.1161/CIRCULATIONAHA.110.940577.
38. Ritchie RH, Leo CH, Qin C, Stephenson EJ, Bowden MA, Buxton KD,
17. Higginbotham MB, Morris KG, Williams RS, Coleman RE, Cobb FR.
Physiologic basis for the age-related decline in aerobic work capacity. Am Lessard SJ, Rivas DA, Koch LG, Britton SL, Hawley JA, Woodman OL.
J Cardiol. 1986;57:1374–1379. Low intrinsic exercise capacity in rats predisposes to age-dependent cardi-
18. Lauer MS, Francis GS, Okin PM, Pashkow FJ, Snader CE, Marwick TH. ac remodeling independent of macrovascular function. Am J Physiol Heart
Impaired chronotropic response to exercise stress testing as a predictor of Circ Physiol. 2013;304:H729–H739. doi: 10.1152/ajpheart.00638.2012.
mortality. JAMA. 1999;281:524–529. 39. Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A. Mechanisms
19. Ogawa T, Spina RJ, Martin WH III, Kohrt WM, Schechtman KB, Holloszy of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci.
JO, Ehsani AA. Effects of aging, sex, and physical training on cardiovas- 2010;65:1028–1041. doi: 10.1093/gerona/glq113.
cular responses to exercise. Circulation. 1992;86:494–503. 40. Kaye DM, Esler MD. Autonomic control of the aging heart. Neuromolecular
20. Rodeheffer RJ, Gerstenblith G, Becker LC, Fleg JL, Weisfeldt ML, Med. 2008;10:179–186. doi: 10.1007/s12017-008-8034-1.
Lakatta EG. Exercise cardiac output is maintained with advanc- 41. Esler M, Skews H, Leonard P, Jackman G, Bobik A, Korner P. Age-
ing age in healthy human subjects: cardiac dilatation and increased dependence of noradrenaline kinetics in normal subjects. Clin Sci (Lond).
stroke volume compensate for a diminished heart rate. Circulation. 1981;60:217–219.
1984;69:203–213. 42. Esler MD, Turner AG, Kaye DM, Thompson JM, Kingwell BA, Morris M,
21. Stratton JR, Levy WC, Cerqueira MD, Schwartz RS, Abrass IB.
Lambert GW, Jennings GL, Cox HS, Seals DR. Aging effects on human
Cardiovascular responses to exercise. Effects of aging and exercise train- sympathetic neuronal function. Am J Physiol. 1995;268:R278–R285.
ing in healthy men. Circulation. 1994;89:1648–1655. 43. Davies CH, Ferrara N, Harding SE. Beta-adrenoceptor function chang-
22. Fleg JL, O’Connor F, Gerstenblith G, Becker LC, Clulow J, Schulman es with age of subject in myocytes from non-failing human ventricle.
SP, Lakatta EG. Impact of age on the cardiovascular response to dynam- Cardiovasc Res. 1996;31:152–156.
ic upright exercise in healthy men and women. J Appl Physiol (1985). 44. Xiao RP, Spurgeon HA, O’Connor F, Lakatta EG. Age-associated changes
1995;78:890–900. in beta-adrenergic modulation on rat cardiac excitation-contraction cou-
23. Fleg JL, Lakatta EG. Role of muscle loss in the age-associated reduction pling. J Clin Invest. 1994;94:2051–2059. doi: 10.1172/JCI117559.
in VO2 max. J Appl Physiol (1985). 1988;65:1147–1151. 45. White M, Roden R, Minobe W, Khan MF, Larrabee P, Wollmering M,
24. Haykowsky MJ, Brubaker PH, Stewart KP, Morgan TM, Eggebeen J, Port JD, Anderson F, Campbell D, Feldman AM. Age-related changes in
Kitzman DW. Effect of endurance training on the determinants of peak beta-adrenergic neuroeffector systems in the human heart. Circulation.
exercise oxygen consumption in elderly patients with stable compen- 1994;90:1225–1238.
sated heart failure and preserved ejection fraction. J Am Coll Cardiol. 46. Xiao RP, Tomhave ED, Wang DJ, Ji X, Boluyt MO, Cheng H, Lakatta EG,
2012;60:120–128. doi: 10.1016/j.jacc.2012.02.055. Koch WJ. Age-associated reductions in cardiac beta1- and beta2-adrener-
25. Borlaug BA, Olson TP, Lam CS, Flood KS, Lerman A, Johnson BD, gic responses without changes in inhibitory G proteins or receptor kinases.
Redfield MM. Global cardiovascular reserve dysfunction in heart failure J Clin Invest. 1998;101:1273–1282. doi: 10.1172/JCI1335.
Roh et al   Exercise and Cardiac Aging   291

47. Böhm M, Dorner H, Htun P, Lensche H, Platt D, Erdmann E. Effects of 67. Lim CC, Apstein CS, Colucci WS, Liao R. Impaired cell shortening and
exercise on myocardial adenylate cyclase and Gi alpha expression in se- relengthening with increased pacing frequency are intrinsic to the senes-
nescence. Am J Physiol. 1993;264:H805–H814. cent mouse cardiomyocyte. J Mol Cell Cardiol. 2000;32:2075–2082. doi:
48. Ferrara N, Böhm M, Zolk O, O’Gara P, Harding SE. The role of Gi- 10.1006/jmcc.2000.1239.
proteins and beta-adrenoceptors in the age-related decline of contraction 68. Isenberg G, Borschke B, Rueckschloss U. Ca2+ transients of cardiomy-
in guinea-pig ventricular myocytes. J Mol Cell Cardiol. 1997;29:439–448. ocytes from senescent mice peak late and decay slowly. Cell Calcium.
49. Jiang MT, Moffat MP, Narayanan N. Age-related alterations in the phos- 2003;34:271–280.
phorylation of sarcoplasmic reticulum and myofibrillar proteins and di- 69. Janczewski AM, Lakatta EG. Modulation of sarcoplasmic reticu-

minished contractile response to isoproterenol in intact rat ventricle. Circ lum Ca(2+) cycling in systolic and diastolic heart failure associ-
Res. 1993;72:102–111. ated with aging. Heart Fail Rev. 2010;15:431–445. doi: 10.1007/
50. Ferrara N, O’Gara P, Wynne DG, Brown LA, del Monte F, Poole-Wilson s10741-010-9167-5.
PA, Harding SE. Decreased contractile responses to isoproterenol in 70. Cain BS, Meldrum DR, Joo KS, Wang JF, Meng X, Cleveland JC

isolated cardiac myocytes from aging guinea-pigs. J Mol Cell Cardiol. Jr, Banerjee A, Harken AH. Human SERCA2a levels correlate in-
1995;27:1141–1150. versely with age in senescent human myocardium. J Am Coll Cardiol.
51. Fowler MB, Laser JA, Hopkins GL, Minobe W, Bristow MR. Assessment 1998;32:458–467.
of the beta-adrenergic receptor pathway in the intact failing human heart: 71. Taffet GE, Tate CA. CaATPase content is lower in cardiac sarcoplasmic
progressive receptor down-regulation and subsensitivity to agonist re- reticulum isolated from old rats. Am J Physiol. 1993;264:H1609–H1614.
sponse. Circulation. 1986;74:1290–1302. 72. Lompré AM, Lambert F, Lakatta EG, Schwartz K. Expression of sarco-
52. Wang W, Zhu W, Wang S, Yang D, Crow MT, Xiao RP, Cheng H.
plasmic reticulum Ca(2+)-ATPase and calsequestrin genes in rat heart dur-
Sustained beta1-adrenergic stimulation modulates cardiac contractility by ing ontogenic development and aging. Circ Res. 1991;69:1380–1388.
Ca2+/calmodulin kinase signaling pathway. Circ Res. 2004;95:798–806. 73. Schmidt U, del Monte F, Miyamoto MI, Matsui T, Gwathmey JK,

doi: 10.1161/01.RES.0000145361.50017.aa. Rosenzweig A, Hajjar RJ. Restoration of diastolic function in senescent rat
53. Zhu WZ, Wang SQ, Chakir K, Yang D, Zhang T, Brown JH, Devic E, hearts through adenoviral gene transfer of sarcoplasmic reticulum Ca(2+)-
Kobilka BK, Cheng H, Xiao RP. Linkage of beta1-adrenergic stimulation ATPase. Circulation. 2000;101:790–796.
to apoptotic heart cell death through protein kinase A-independent activa- 74. Lim CC, Liao R, Varma N, Apstein CS. Impaired lusitropy-frequency in
tion of Ca2+/calmodulin kinase II. J Clin Invest. 2003;111:617–625. doi: the aging mouse: role of Ca(2+)-handling proteins and effects of isopro-
10.1172/JCI16326. terenol. Am J Physiol. 1999;277:H2083–H2090.
54. Backs J, Worst BC, Lehmann LH, Patrick DM, Jebessa Z, Kreusser MM, 75. Xu A, Narayanan N. Effects of aging on sarcoplasmic reticulum Ca2+-
Sun Q, Chen L, Heft C, Katus HA, Olson EN. Selective repression of cycling proteins and their phosphorylation in rat myocardium. Am J
MEF2 activity by PKA-dependent proteolysis of HDAC4. J Cell Biol. Physiol. 1998;275:H2087–H2094.
2011;195:403–415. doi: 10.1083/jcb.201105063. 76. Fares E, Howlett SE. Effect of age on cardiac excitation-contrac-

55. Brodde OE, Michel MC. Adrenergic and muscarinic receptors in the hu- tion coupling. Clin Exp Pharmacol Physiol. 2010;37:1–7. doi:
man heart. Pharmacol Rev. 1999;51:651–690. 10.1111/j.1440-1681.2009.05276.x.
56. Brodde OE, Konschak U, Becker K, Rüter F, Poller U, Jakubetz J, Radke 77. Kemi OJ, Ellingsen O, Smith GL, Wisloff U. Exercise-induced changes
J, Zerkowski HR. Cardiac muscarinic receptors decrease with age. In vi- in calcium handling in left ventricular cardiomyocytes. Front Biosci.
tro and in vivo studies. J Clin Invest. 1998;101:471–478. doi: 10.1172/ 2008;13:356–368.
JCI1113. 78. Kemi OJ, Ceci M, Condorelli G, Smith GL, Wisloff U. Myocardial sarco-
Downloaded from http://ahajournals.org by on November 15, 2020

57. Poller U, Nedelka G, Radke J, Pönicke K, Brodde OE. Age-dependent plasmic reticulum Ca2+ ATPase function is increased by aerobic interval
changes in cardiac muscarinic receptor function in healthy volunteers. J training. Eur J Cardiovasc Prev Rehabil. 2008;15:145–148. doi: 10.1097/
Am Coll Cardiol. 1997;29:187–193. HJR.0b013e3282efd4e0.
58. Stratton JR, Levy WC, Caldwell JH, Jacobson A, May J, Matsuoka D, 79. Kemi OJ, Ellingsen O, Ceci M, Grimaldi S, Smith GL, Condorelli G,
Madden K. Effects of aging on cardiovascular responses to parasympa- Wisløff U. Aerobic interval training enhances cardiomyocyte contrac-
thetic withdrawal. J Am Coll Cardiol. 2003;41:2077–2083. tility and Ca2+ cycling by phosphorylation of CaMKII and Thr-17 of
59. Fleg JL, Schulman S, O’Connor F, Becker LC, Gerstenblith G, Clulow phospholamban. J Mol Cell Cardiol. 2007;43:354–361. doi: 10.1016/j.
JF, Renlund DG, Lakatta EG. Effects of acute beta-adrenergic receptor yjmcc.2007.06.013.
blockade on age-associated changes in cardiovascular performance during 80. Tate CA, Helgason T, Hyek MF, McBride RP, Chen M, Richardson MA,
dynamic exercise. Circulation. 1994;90:2333–2341. Taffet GE. SERCA2a and mitochondrial cytochrome oxidase expres-
60. Leosco D, Parisi V, Femminella GD, Formisano R, Petraglia L, Allocca sion are increased in hearts of exercise-trained old rats. Am J Physiol.
E, Bonaduce D. Effects of exercise training on cardiovascular adrenergic 1996;271:H68–H72.
system. Front Physiol. 2013;4:348. doi: 10.3389/fphys.2013.00348. 81. Tate CA, Taffet GE, Hudson EK, Blaylock SL, McBride RP, Michael LH.
61. Spina RJ, Turner MJ, Ehsani AA. Beta-adrenergic-mediated improvement Enhanced calcium uptake of cardiac sarcoplasmic reticulum in exercise-
in left ventricular function by exercise training in older men. Am J Physiol. trained old rats. Am J Physiol. 1990;258:H431–H435.
1998;274:H397–H404. 82. Brenner DA, Apstein CS, Saupe KW. Exercise training attenu-

62. Leosco D, Rengo G, Iaccarino G, Filippelli A, Lymperopoulos A,
ates age-associated diastolic dysfunction in rats. Circulation.
Zincarelli C, Fortunato F, Golino L, Marchese M, Esposito G, Rapacciuolo 2001;104:221–226.
A, Rinaldi B, Ferrara N, Koch WJ, Rengo F. Exercise training and beta- 83. Iemitsu M, Miyauchi T, Maeda S, Tanabe T, Takanashi M, Matsuda M,
blocker treatment ameliorate age-dependent impairment of beta-adrener- Yamaguchi I. Exercise training improves cardiac function-related gene
gic receptor signaling and enhance cardiac responsiveness to adrenergic levels through thyroid hormone receptor signaling in aged rats. Am J
stimulation. Am J Physiol Heart Circ Physiol. 2007;293:H1596–H1603. Physiol Heart Circ Physiol. 2004;286:H1696–H1705. doi: 10.1152/
doi: 10.1152/ajpheart.00308.2007. ajpheart.00761.2003.
63. Bhella PS, Hastings JL, Fujimoto N, Shibata S, Carrick-Ranson G, Palmer 84. Thomas MM, Vigna C, Betik AC, Tupling AR, Hepple RT. Cardiac

MD, Boyd KN, Adams-Huet B, Levine BD. Impact of lifelong exer- calcium pump inactivation and nitrosylation in senescent rat myocar-
cise “dose” on left ventricular compliance and distensibility. J Am Coll dium are not attenuated by long-term treadmill training. Exp Gerontol.
Cardiol. 2014;64:1257–1266. doi: 10.1016/j.jacc.2014.03.062. 2011;46:803–810. doi: 10.1016/j.exger.2011.06.005.
64. Wisløff U, Ellingsen Ø, Kemi OJ. High-intensity interval training to 85. Walton RD, Jones SA, Rostron KA, Kayani AC, Close GL, McArdle A,
maximize cardiac benefits of exercise training? Exerc Sport Sci Rev. Lancaster MK. Interactions of short-term and chronic treadmill training
2009;37:139–146. doi: 10.1097/JES.0b013e3181aa65fc. with aging of the left ventricle of the heart. J Gerontol A Biol Sci Med Sci.
65. Kemi OJ, Haram PM, Loennechen JP, Osnes JB, Skomedal T, Wisløff 2015. doi: 10.1093/gerona/glv093
U, Ellingsen Ø. Moderate vs. high exercise intensity: differential ef- 86. Gerstenblith G, Frederiksen J, Yin FC, Fortuin NJ, Lakatta EG, Weisfeldt
fects on aerobic fitness, cardiomyocyte contractility, and endothe- ML. Echocardiographic assessment of a normal adult aging population.
lial function. Cardiovasc Res. 2005;67:161–172. doi: 10.1016/j. Circulation. 1977;56:273–278.
cardiores.2005.03.010. 87. Levy D, Anderson KM, Savage DD, Kannel WB, Christiansen JC, Castelli
66. Scarpace PJ, Shu Y, Tümer N. Influence of exercise training on myocar- WP. Echocardiographically detected left ventricular hypertrophy: preva-
dial beta-adrenergic signal transduction: differential regulation with age. lence and risk factors. The Framingham Heart Study. Ann Intern Med.
J Appl Physiol (1985). 1994;77:737–741. 1988;108:7–13.
292  Circulation Research  January 22, 2016

88. de Simone G, Gottdiener JS, Chinali M, Maurer MS. Left ventricular 108. Jackson MF, Luong D, Vang DD, Garikipati DK, Stanton JB, Nelson OL,
mass predicts heart failure not related to previous myocardial infarction: Rodgers BD. The aging myostatin null phenotype: reduced adiposity,
the Cardiovascular Health Study. Eur Heart J. 2008;29:741–747. doi: cardiac hypertrophy, enhanced cardiac stress response, and sexual dimor-
10.1093/eurheartj/ehm605. phism. J Endocrinol. 2012;213:263–275. doi: 10.1530/JOE-11-0455.
89. Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Prognostic 109. Rodgers BD, Interlichia JP, Garikipati DK, Mamidi R, Chandra M,
implications of echocardiographically determined left ventricular mass Nelson OL, Murry CE, Santana LF. Myostatin represses physiological
in the Framingham Heart Study. N Engl J Med. 1990;322:1561–1566. hypertrophy of the heart and excitation-contraction coupling. J Physiol.
doi: 10.1056/NEJM199005313222203. 2009;587:4873–4886. doi: 10.1113/jphysiol.2009.172544.
90. Boyle AJ, Shih H, Hwang J, Ye J, Lee B, Zhang Y, Kwon D, Jun K, 110. Heineke J, Auger-Messier M, Xu J, Sargent M, York A, Welle S,

Zheng D, Sievers R, Angeli F, Yeghiazarians Y, Lee R. Cardiomyopathy Molkentin JD. Genetic deletion of myostatin from the heart prevents
of aging in the mammalian heart is characterized by myocardial hy- skeletal muscle atrophy in heart failure. Circulation. 2010;121:419–425.
pertrophy, fibrosis and a predisposition towards cardiomyocyte apop- doi: 10.1161/CIRCULATIONAHA.109.882068.
tosis and autophagy. Exp Gerontol. 2011;46:549–559. doi: 10.1016/j. 111. Egerman MA, Cadena SM, Gilbert JA, et al. GDF11 Increases with Age
exger.2011.02.010. and Inhibits Skeletal Muscle Regeneration. Cell Metab. 2015;22:164–
91. Olivetti G, Melissari M, Capasso JM, Anversa P. Cardiomyopathy of the 174. doi: 10.1016/j.cmet.2015.05.010.
aging human heart. Myocyte loss and reactive cellular hypertrophy. Circ 112. DeMaria AN, Neumann A, Lee G, Fowler W, Mason DT. Alterations in
Res. 1991;68:1560–1568. ventricular mass and performance induced by exercise training in man
92. Anversa P, Hiler B, Ricci R, Guideri G, Olivetti G. Myocyte cell loss evaluated by echocardiography. Circulation. 1978;57:237–244.
and myocyte hypertrophy in the aging rat heart. J Am Coll Cardiol. 113. Ellison GM, Waring CD, Vicinanza C, Torella D. Physiological car-
1986;8:1441–1448. diac remodelling in response to endurance exercise training: cellu-
93. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider lar and molecular mechanisms. Heart. 2012;98:5–10. doi: 10.1136/
F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, heartjnl-2011-300639.
Frisén J. Evidence for cardiomyocyte renewal in humans. Science. 114. Kong SW, Bodyak N, Yue P, Liu Z, Brown J, Izumo S, Kang PM. Genetic
2009;324:98–102. doi: 10.1126/science.1164680. expression profiles during physiological and pathological cardiac hyper-
94. Terman A, Brunk UT. Autophagy in cardiac myocyte homeostasis, ag- trophy and heart failure in rats. Physiol Genomics. 2005;21:34–42. doi:
ing, and pathology. Cardiovasc Res. 2005;68:355–365. doi: 10.1016/j. 10.1152/physiolgenomics.00226.2004.
cardiores.2005.08.014. 115. Neri Serneri GG, Boddi M, Modesti PA, Cecioni I, Coppo M, Padeletti
95. Frey N, Katus HA, Olson EN, Hill JA. Hypertrophy of the heart: a new L, Michelucci A, Colella A, Galanti G. Increased cardiac sympathetic
therapeutic target? Circulation. 2004;109:1580–1589. doi: 10.1161/01. activity and insulin-like growth factor-I formation are associated with
CIR.0000120390.68287.BB. physiological hypertrophy in athletes. Circ Res. 2001;89:977–982.
96. van Empel VP, De Windt LJ. Myocyte hypertrophy and apoptosis: 116. Kim J, Wende AR, Sena S, Theobald HA, Soto J, Sloan C, Wayment
a balancing act. Cardiovasc Res. 2004;63:487–499. doi: 10.1016/j.
BE, Litwin SE, Holzenberger M, LeRoith D, Abel ED. Insulin-like
cardiores.2004.02.013.
growth factor I receptor signaling is required for exercise-induced car-
97. Domenighetti AA, Wang Q, Egger M, Richards SM, Pedrazzini
diac hypertrophy. Mol Endocrinol. 2008;22:2531–2543. doi: 10.1210/
T, Delbridge LM. Angiotensin II-mediated phenotypic cardio-
me.2008-0265.
myocyte remodeling leads to age-dependent cardiac dysfunc-
117. McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Kang PM,
tion and failure. Hypertension. 2005;46:426–432. doi: 10.1161/01.
Izumo S. Phosphoinositide 3-kinase(p110alpha) plays a critical role for
Downloaded from http://ahajournals.org by on November 15, 2020

HYP.0000173069.53699.d9.
the induction of physiological, but not pathological, cardiac hypertro-
98. Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracel-
phy. Proc Natl Acad Sci U S A. 2003;100:12355–12360. doi: 10.1073/
lular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600. doi:
pnas.1934654100.
10.1038/nrm1983.
118. McMullen JR, Shioi T, Huang WY, Zhang L, Tarnavski O, Bisping E,
99. Liao PH, Hsieh DJ, Kuo CH, Day CH, Shen CY, Lai CH, Chen RJ,
Schinke M, Kong S, Sherwood MC, Brown J, Riggi L, Kang PM, Izumo
Padma VV, Kuo WW, Huang CY. Moderate exercise training attenu-
S. The insulin-like growth factor 1 receptor induces physiological heart
ates aging-induced cardiac inflammation, hypertrophy and fibrosis in-
growth via the phosphoinositide 3-kinase(p110alpha) pathway. J Biol
juries of rat hearts. Oncotarget. 2015;6:35383–35394. doi: 10.18632/
oncotarget.6168. Chem. 2004;279:4782–4793. doi: 10.1074/jbc.M310405200.
100. Manne ND, Kakarla SK, Arvapalli R, Rice KM, Blough ER. Molecular 119. DeBosch B, Treskov I, Lupu TS, Weinheimer C, Kovacs A, Courtois M,
mechanisms of age-related cardiac hypertrophy in F344XBN rat model. Muslin AJ. Akt1 is required for physiological cardiac growth. Circulation.
J Clin Exp Cardiolog. 2014;5:1–6. 2006;113:2097–2104. doi: 10.1161/CIRCULATIONAHA.105.595231.
101. Ceylan-Isik AF, Dong M, Zhang Y, Dong F, Turdi S, Nair S, Yanagisawa 120. Matsui T, Li L, del MonteF, Fukui Y, Franke TF, Hajjar RJ, Rosenzweig
M, Ren J. Cardiomyocyte-specific deletion of endothelin receptor A A. Adenoviral gene transfer of activated phosphatidylinositol 3’-ki-
rescues aging-associated cardiac hypertrophy and contractile dysfunc- nase and Akt inhibits apoptosis of hypoxic cardiomyocytes in vitro.
tion: role of autophagy. Basic Res Cardiol. 2013;108:335. doi: 10.1007/ Circulation. 1999;100:2373–2379.
s00395-013-0335-3. 121. Matsui T, Tao J, del Monte F, Lee KH, Li L, Picard M, Force TL, Franke
102. Engelhardt S, Hein L, Wiesmann F, Lohse MJ. Progressive hypertrophy TF, Hajjar RJ, Rosenzweig A. Akt activation preserves cardiac function
and heart failure in beta1-adrenergic receptor transgenic mice. Proc Natl and prevents injury after transient cardiac ischemia in vivo. Circulation.
Acad Sci U S A. 1999;96:7059–7064. 2001;104:330–335.
103. Inuzuka Y, Okuda J, Kawashima T, et al. Suppression of phosphoinositide 122. Boström P, Mann N, Wu J, Quintero PA, Plovie ER, Panáková D, Gupta
3-kinase prevents cardiac aging in mice. Circulation. 2009;120:1695– RK, Xiao C, MacRae CA, Rosenzweig A, Spiegelman BM. C/EBPβ
1703. doi: 10.1161/CIRCULATIONAHA.109.871137. controls exercise-induced cardiac growth and protects against patho-
104. Flynn JM, O’Leary MN, Zambataro CA, Academia EC, Presley MP, logical cardiac remodeling. Cell. 2010;143:1072–1083. doi: 10.1016/j.
Garrett BJ, Zykovich A, Mooney SD, Strong R, Rosen CJ, Kapahi P, cell.2010.11.036.
Nelson MD, Kennedy BK, Melov S. Late-life rapamycin treatment re- 123. Carè A, Catalucci D, Felicetti F, et al. MicroRNA-133 controls cardiac
verses age-related heart dysfunction. Aging Cell. 2013;12:851–862. doi: hypertrophy. Nat Med. 2007;13:613–618. doi: 10.1038/nm1582.
10.1111/acel.12109. 124. Wang Y, Wisloff U, Kemi OJ. Animal models in the study of exercise-
105. Loffredo FS, Steinhauser ML, Jay SM, et al. Growth differentiation fac- induced cardiac hypertrophy. Physiol Res. 2010;59:633–644.
tor 11 is a circulating factor that reverses age-related cardiac hypertro- 125. Liu X, Xiao J, Zhu H, Wei X, Platt C, Damilano F, Xiao C, Bezzerides
phy. Cell. 2013;153:828–839. doi: 10.1016/j.cell.2013.04.015. V, Boström P, Che L, Zhang C, Spiegelman BM, Rosenzweig A. miR-
106. Smith SC, Zhang X, Zhang X, et al. GDF11 Does Not Rescue Aging- 222 is necessary for exercise-induced cardiac growth and protects against
Related Pathological Hypertrophy. Circ Res. 2015;117:926–932. doi: pathological cardiac remodeling. Cell Metab. 2015;21:584–595. doi:
10.1161/CIRCRESAHA.115.307527. 10.1016/j.cmet.2015.02.014.
107.
Morissette MR, Stricker JC, Rosenberg MA, Buranasombati 126. Wright KJ, Thomas MM, Betik AC, Belke D, Hepple RT. Exercise train-
C, Levitan EB, Mittleman MA, Rosenzweig A. Effects of myo- ing initiated in late middle age attenuates cardiac fibrosis and advanced
statin deletion in aging mice. Aging Cell. 2009;8:573–583. doi: glycation end-product accumulation in senescent rats. Exp Gerontol.
10.1111/j.1474-9726.2009.00508.x. 2014;50:9–18. doi: 10.1016/j.exger.2013.11.006.
Roh et al   Exercise and Cardiac Aging   293

127. Choi SY, Chang HJ, Choi SI, Kim KI, Cho YS, Youn TJ, Chung WY, 145. Lenk K, Erbs S, Höllriegel R, Beck E, Linke A, Gielen S, Winkler
Chae IH, Choi DJ, Kim HS, Kim CH, Oh BH, Kim MH. Long-term SM, Sandri M, Hambrecht R, Schuler G, Adams V. Exercise train-
exercise training attenuates age-related diastolic dysfunction: association ing leads to a reduction of elevated myostatin levels in patients with
of myocardial collagen cross-linking. J Korean Med Sci. 2009;24:32–39. chronic heart failure. Eur J Prev Cardiol. 2012;19:404–411. doi:
doi: 10.3346/jkms.2009.24.1.32. 10.1177/1741826711402735.
128. Wang W, Zhang H, Xue G, Zhang L, Zhang W, Wang L, Lu F, Li H, Bai S, 146. Morissette MR, Cook SA, Foo S, McKoy G, Ashida N, Novikov

Lin Y, Lou Y, Xu C, Zhao Y. Exercise training preserves ischemic precon- M, Scherrer-Crosbie M, Li L, Matsui T, Brooks G, Rosenzweig
ditioning in aged rat hearts by restoring the myocardial polyamine pool. A. Myostatin regulates cardiomyocyte growth through modula-
Oxid Med Cell Longev. 2014;2014:457429. doi: 10.1155/2014/457429. tion of Akt signaling. Circ Res. 2006;99:15–24. doi: 10.1161/01.
129. Derumeaux G, Ichinose F, Raher MJ, Morgan JG, Coman T, Lee C, RES.0000231290.45676.d4.
Cuesta JM, Thibault H, Bloch KD, Picard MH, Scherrer-Crosbie M. 147. Molkentin JD. Parsing good versus bad signaling pathways in the

Myocardial alterations in senescent mice and effect of exercise training: heart: role of calcineurin-nuclear factor of activated T-cells. Circ Res.
a strain rate imaging study. Circ Cardiovasc Imaging. 2008;1:227–234. 2013;113:16–19. doi: 10.1161/CIRCRESAHA.113.301667.
doi: 10.1161/CIRCIMAGING.107.745919. 148. Chung E, Yeung F, Leinwand LA. Calcineurin activity is required for
130. Kwak HB, Song W, Lawler JM. Exercise training attenuates age-induced cardiac remodelling in pregnancy. Cardiovasc Res. 2013;100:402–410.
elevation in Bax/Bcl-2 ratio, apoptosis, and remodeling in the rat heart. doi: 10.1093/cvr/cvt208.
FASEB J. 2006;20:791–793. doi: 10.1096/fj.05-5116fje. 149. Harman D. The biologic clock: the mitochondria? J Am Geriatr Soc.
131. Rossoni LV, Oliveira RA, Caffaro RR, Miana M, Sanz-Rosa D, Koike 1972;20:145–147.
MK, Do Amaral SL, Michelini LC, Lahera V, Cachofeiro V. Cardiac 150. Linnane AW, Marzuki S, Ozawa T, Tanaka M. Mitochondrial DNA mu-
benefits of exercise training in aging spontaneously hypertensive rats. J tations as an important contributor to ageing and degenerative diseases.
Hypertens. 2011;29:2349–2358. doi: 10.1097/HJH.0b013e32834d2532. Lancet. 1989;1:642–645.
132. Iemitsu M, Maeda S, Jesmin S, Otsuki T, Miyauchi T. Exercise training 151. Dai DF, Rabinovitch PS. Cardiac aging in mice and humans: the role of
improves aging-induced downregulation of VEGF angiogenic signaling mitochondrial oxidative stress. Trends Cardiovasc Med. 2009;19:213–
cascade in hearts. Am J Physiol Heart Circ Physiol. 2006;291:H1290– 220. doi: 10.1016/j.tcm.2009.12.004.
H1298. doi: 10.1152/ajpheart.00820.2005. 152. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo ZM, Kewitt K,
133. Huang CY, Yang AL, Lin YM, Wu FN, Lin JA, Chan YS, Tsai FJ, Tsai Walter CA, Richardson A. Does oxidative damage to DNA increase with
CH, Kuo CH, Lee SD. Anti-apoptotic and pro-survival effects of exercise age? Proc Natl Acad Sci U S A. 2001;98:10469–10474. doi: 10.1073/
training on hypertensive hearts. J Appl Physiol (1985). 2012;112:883– pnas.171202698.
891. doi: 10.1152/japplphysiol.00605.2011. 153. Khrapko K, Bodyak N, Thilly WG, van Orsouw NJ, Zhang X, Coller
134. Lai CH, Ho TJ, Kuo WW, Day CH, Pai PY, Chung LC, Liao PH, Lin HA, Perls TT, Upton M, Vijg J, Wei JY. Cell-by-cell scanning of whole
mitochondrial genomes in aged human heart reveals a significant frac-
FH, Wu ET, Huang CY. Exercise training enhanced SIRT1 longev-
tion of myocytes with clonally expanded deletions. Nucleic Acids Res.
ity signaling replaces the IGF1 survival pathway to attenuate aging-
1999;27:2434–2441.
induced rat heart apoptosis. Age (Dordr). 2014;36:9706. doi: 10.1007/
154. Orr WC, Sohal RS. Extension of life-span by overexpression of su-
s11357-014-9706-4.
peroxide dismutase and catalase in Drosophila melanogaster. Science.
135. Putinski C, Abdul-Ghani M, Stiles R, Brunette S, Dick SA, Fernando P,
1994;263:1128–1130.
Megeney LA. Intrinsic-mediated caspase activation is essential for car-
155. Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP,
diomyocyte hypertrophy. Proc Natl Acad Sci U S A. 2013;110:E4079–
Downloaded from http://ahajournals.org by on November 15, 2020

Lanfrancone L, Pelicci PG. The p66shc adaptor protein controls oxida-


E4087. doi: 10.1073/pnas.1315587110.
tive stress response and life span in mammals. Nature. 1999;402:309–
136. Yamashita K, Kajstura J, Discher DJ, Wasserlauf BJ, Bishopric NH,
313. doi: 10.1038/46311.
Anversa P, Webster KA. Reperfusion-activated Akt kinase prevents apop-
156. Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging.
tosis in transgenic mouse hearts overexpressing insulin-like growth fac-
Science. 1996;273:59–63.
tor-1. Circ Res. 2001;88:609–614.
157. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive
137. Weeks KL, Gao X, Du XJ, et al. Phosphoinositide 3-kinase p110α is
and persists longer than nuclear DNA damage in human cells following
a master regulator of exercise-induced cardioprotection and PI3K gene oxidative stress. Proc Natl Acad Sci U S A. 1997;94:514–519.
therapy rescues cardiac dysfunction. Circ Heart Fail. 2012;5:523–534. 158. Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, Bruder
doi: 10.1161/CIRCHEARTFAILURE.112.966622. CE, Bohlooly-Y M, Gidlöf S, Oldfors A, Wibom R, Törnell J, Jacobs HT,
138. Lin CH, Lin CC, Ting WJ, Pai PY, Kuo CH, Ho TJ, Kuo WW, Chang CH, Larsson NG. Premature ageing in mice expressing defective mitochondrial
Huang CY, Lin WT. Resveratrol enhanced FOXO3 phosphorylation via DNA polymerase. Nature. 2004;429:417–423. doi: 10.1038/nature02517.
synergetic activation of SIRT1 and PI3K/Akt signaling to improve the 159. Kujoth GC, Hiona A, Pugh TD, et al. Mitochondrial DNA muta-

effects of exercise in elderly rat hearts. Age (Dordr). 2014;36:9705. doi: tions, oxidative stress, and apoptosis in mammalian aging. Science.
10.1007/s11357-014-9705-5. 2005;309:481–484. doi: 10.1126/science.1112125.
139. Zimmermann S, Moelling K. Phosphorylation and regulation of Raf by 160. Hiona A, Sanz A, Kujoth GC, et al. Mitochondrial DNA mutations in-
Akt (protein kinase B). Science. 1999;286:1741–1744. duce mitochondrial dysfunction, apoptosis and sarcopenia in skeletal
140. Rommel C, Clarke BA, Zimmermann S, Nuñez L, Rossman R, Reid muscle of mitochondrial DNA mutator mice. PLoS One. 2010;5:e11468.
K, Moelling K, Yancopoulos GD, Glass DJ. Differentiation stage- doi: 10.1371/journal.pone.0011468.
specific inhibition of the Raf-MEK-ERK pathway by Akt. Science. 161. Trifunovic A, Hansson A, Wredenberg A, Rovio AT, Dufour E,

1999;286:1738–1741. Khvorostov I, Spelbrink JN, Wibom R, Jacobs HT, Larsson NG. Somatic
141. Gratton JP, Morales-Ruiz M, Kureishi Y, Fulton D, Walsh K, Sessa WC. mtDNA mutations cause aging phenotypes without affecting reactive
Akt down-regulation of p38 signaling provides a novel mechanism of oxygen species production. Proc Natl Acad Sci U S A. 2005;102:17993–
vascular endothelial growth factor-mediated cytoprotection in endo- 17998. doi: 10.1073/pnas.0508886102.
thelial cells. J Biol Chem. 2001;276:30359–30365. doi: 10.1074/jbc. 162. Dai DF, Chen T, Wanagat J, Laflamme M, Marcinek DJ, Emond MJ,
M009698200. Ngo CP, Prolla TA, Rabinovitch PS. Age-dependent cardiomyopa-
142. Lebrasseur NK, Coté GM, Miller TA, Fielding RA, Sawyer DB.
thy in mitochondrial mutator mice is attenuated by overexpression of
Regulation of neuregulin/ErbB signaling by contractile activity in skel- catalase targeted to mitochondria. Aging Cell. 2010;9:536–544. doi:
etal muscle. Am J Physiol Cell Physiol. 2003;284:C1149–C1155. doi: 10.1111/j.1474-9726.2010.00581.x.
10.1152/ajpcell.00487.2002. 163. Rossignol R, Faustin B, Rocher C, Malgat M, Mazat JP, Letellier T.
143. Fukazawa R, Miller TA, Kuramochi Y, Frantz S, Kim YD, Marchionni Mitochondrial threshold effects. Biochem J. 2003;370:751–762. doi:
MA, Kelly RA, Sawyer DB. Neuregulin-1 protects ventricular myocytes 10.1042/BJ20021594.
from anthracycline-induced apoptosis via erbB4-dependent activation of 164. Müller-Höcker J. Cytochrome-c-oxidase deficient cardiomyocytes in the
PI3-kinase/Akt. J Mol Cell Cardiol. 2003;35:1473–1479. human heart–an age-related phenomenon. A histochemical ultracyto-
144. Lenk K, Schur R, Linke A, Erbs S, Matsumoto Y, Adams V, Schuler G. chemical study. Am J Pathol. 1989;134:1167–1173.
Impact of exercise training on myostatin expression in the myocardium 165. Wang J, Wilhelmsson H, Graff C, Li H, Oldfors A, Rustin P, Brüning
and skeletal muscle in a chronic heart failure model. Eur J Heart Fail. JC, Kahn CR, Clayton DA, Barsh GS, Thorén P, Larsson NG. Dilated
2009;11:342–348. doi: 10.1093/eurjhf/hfp020. cardiomyopathy and atrioventricular conduction blocks induced by
294  Circulation Research  January 22, 2016

heart-specific inactivation of mitochondrial DNA gene expression. Nat trained human skeletal muscle. Am J Physiol Regul Integr Comp Physiol.
Genet. 1999;21:133–137. doi: 10.1038/5089. 2010;298:R912–R917. doi: 10.1152/ajpregu.00409.2009.
166. Baris OR, Ederer S, Neuhaus JF, von Kleist-Retzow JC, Wunderlich 185. Chinsomboon J, Ruas J, Gupta RK, Thom R, Shoag J, Rowe GC,
CM, Pal M, Wunderlich FT, Peeva V, Zsurka G, Kunz WS, Hickethier T, Sawada N, Raghuram S, Arany Z. The transcriptional coactivator
Bunck AC, Stöckigt F, Schrickel JW, Wiesner RJ. Mosaic deficiency in PGC-1alpha mediates exercise-induced angiogenesis in skeletal mus-
mitochondrial oxidative metabolism promotes cardiac arrhythmia during cle. Proc Natl Acad Sci U S A. 2009;106:21401–21406. doi: 10.1073/
aging. Cell Metab. 2015;21:667–677. doi: 10.1016/j.cmet.2015.04.005. pnas.0909131106.
167. Rossi DJ, Jamieson CH, Weissman IL. Stems cells and the path-
186. Ling C, Poulsen P, Carlsson E, Ridderstråle M, Almgren P, Wojtaszewski
ways to aging and cancer. Cell. 2008;132:681–696. doi: 10.1016/j. J, Beck-Nielsen H, Groop L, Vaag A. Multiple environmental and ge-
cell.2008.01.036. netic factors influence skeletal muscle PGC-1alpha and PGC-1beta gene
168. Ahlqvist KJ, Hämäläinen RH, Yatsuga S, et al. Somatic progenitor cell expression in twins. J Clin Invest. 2004;114:1518–1526. doi: 10.1172/
vulnerability to mitochondrial DNA mutagenesis underlies progeroid JCI21889.
phenotypes in Polg mutator mice. Cell Metab. 2012;15:100–109. doi: 187. Dillon LM, Rebelo AP, Moraes CT. The role of PGC-1 coactivators in
10.1016/j.cmet.2011.11.012. aging skeletal muscle and heart. IUBMB Life. 2012;64:231–241. doi:
169. Menshikova EV, Ritov VB, Fairfull L, Ferrell RE, Kelley DE, Goodpaster 10.1002/iub.608.
BH. Effects of exercise on mitochondrial content and function in aging 188. Dillon LM, Williams SL, Hida A, Peacock JD, Prolla TA, Lincoln J,
human skeletal muscle. J Gerontol A Biol Sci Med Sci. 2006;61:534–540. Moraes CT. Increased mitochondrial biogenesis in muscle improves
170. Viña J, Gomez-Cabrera MC, Borras C, Froio T, Sanchis-Gomar F,
aging phenotypes in the mtDNA mutator mouse. Hum Mol Genet.
Martinez-Bello VE, Pallardo FV. Mitochondrial biogenesis in exercise 2012;21:2288–2297. doi: 10.1093/hmg/dds049.
and in ageing. Adv Drug Deliv Rev. 2009;61:1369–1374. doi: 10.1016/j. 189. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jäger S, Handschin
addr.2009.06.006. C, Zheng K, Lin J, Yang W, Simon DK, Bachoo R, Spiegelman BM.
171. Eisele JC, Schaefer IM, Randel Nyengaard J, Post H, Liebetanz D, Brüel Suppression of reactive oxygen species and neurodegeneration by the
A, Mühlfeld C. Effect of voluntary exercise on number and volume of PGC-1 transcriptional coactivators. Cell. 2006;127:397–408. doi:
cardiomyocytes and their mitochondria in the mouse left ventricle. Basic 10.1016/j.cell.2006.09.024.
Res Cardiol. 2008;103:12–21. doi: 10.1007/s00395-007-0684-x. 190. Wenz T, Rossi SG, Rotundo RL, Spiegelman BM, Moraes CT. Increased
172. Safdar A, Bourgeois JM, Ogborn DI, Little JP, Hettinga BP, Akhtar M, muscle PGC-1alpha expression protects from sarcopenia and metabolic
Thompson JE, Melov S, Mocellin NJ, Kujoth GC, Prolla TA, Tarnopolsky disease during aging. Proc Natl Acad Sci U S A. 2009;106:20405–20410.
MA. Endurance exercise rescues progeroid aging and induces systemic doi: 10.1073/pnas.0911570106.
mitochondrial rejuvenation in mtDNA mutator mice. Proc Natl Acad Sci 191. Kang KW, Lee SJ, Kim SG. Molecular mechanism of nrf2 activation
U S A. 2011;108:4135–4140. doi: 10.1073/pnas.1019581108. by oxidative stress. Antioxid Redox Signal. 2005;7:1664–1673. doi:
173. Linke A, Adams V, Schulze PC, Erbs S, Gielen S, Fiehn E, Möbius- 10.1089/ars.2005.7.1664.
Winkler S, Schubert A, Schuler G, Hambrecht R. Antioxidative effects 192. Cherry AD, Suliman HB, Bartz RR, Piantadosi CA. Peroxisome prolif-
of exercise training in patients with chronic heart failure: increase in erator-activated receptor γ co-activator 1-α as a critical co-activator of
radical scavenger enzyme activity in skeletal muscle. Circulation. the murine hepatic oxidative stress response and mitochondrial biogen-
2005;111:1763–1770. doi: 10.1161/01.CIR.0000165503.08661.E5. esis in Staphylococcus aureus sepsis. J Biol Chem. 2014;289:41–52. doi:
174. Yamashita N, Hoshida S, Otsu K, Asahi M, Kuzuya T, Hori M. Exercise 10.1074/jbc.M113.512483.
provides direct biphasic cardioprotection via manganese superoxide dis- 193. Aquilano K, Baldelli S, Pagliei B, Cannata SM, Rotilio G, Ciriolo MR.
Downloaded from http://ahajournals.org by on November 15, 2020

mutase activation. J Exp Med. 1999;189:1699–1706. p53 orchestrates the PGC-1α-mediated antioxidant response upon mild
175. Demirel HA, Powers SK, Caillaud C, Coombes JS, Naito H, Fletcher redox and metabolic imbalance. Antioxid Redox Signal. 2013;18:386–
LA, Vrabas I, Jessup JV, Ji LL. Exercise training reduces myocardial 399. doi: 10.1089/ars.2012.4615.
lipid peroxidation following short-term ischemia-reperfusion. Med Sci 194. Muthusamy VR, Kannan S, Sadhaasivam K, Gounder SS, Davidson CJ,
Sports Exerc. 1998;30:1211–1216. Boeheme C, Hoidal JR, Wang L, Rajasekaran NS. Acute exercise stress
176. Kanter MM, Hamlin RL, Unverferth DV, Davis HW, Merola AJ. Effect activates Nrf2/ARE signaling and promotes antioxidant mechanisms in
of exercise training on antioxidant enzymes and cardiotoxicity of doxo- the myocardium. Free Radic Biol Med. 2012;52:366–376. doi: 10.1016/j.
rubicin. J Appl Physiol (1985). 1985;59:1298–1303. freeradbiomed.2011.10.440.
177. Powers SK, Criswell D, Lawler J, Martin D, Lieu FK, Ji LL, Herb RA. 195. He X, Ma Q. Redox regulation by nuclear factor erythroid 2-related
Rigorous exercise training increases superoxide dismutase activity in factor 2: gatekeeping for the basal and diabetes-induced expression of
ventricular myocardium. Am J Physiol. 1993;265:H2094–H2098. thioredoxin-interacting protein. Mol Pharmacol. 2012;82:887–897. doi:
178. Somani SM, Frank S, Rybak LP. Responses of antioxidant system to 10.1124/mol.112.081133.
acute and trained exercise in rat heart subcellular fractions. Pharmacol 196. Gounder SS, Kannan S, Devadoss D, Miller CJ, Whitehead KJ,

Biochem Behav. 1995;51:627–634. Whitehead KS, Odelberg SJ, Firpo MA, Paine R III, Hoidal JR, Abel ED,
179. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Rajasekaran NS. Impaired transcriptional activity of Nrf2 in age-related
Troy A, Cinti S, Lowell B, Scarpulla RC, Spiegelman BM. Mechanisms myocardial oxidative stress is reversible by moderate exercise training.
controlling mitochondrial biogenesis and respiration through the ther- PLoS One. 2012;7:e45697. doi: 10.1371/journal.pone.0045697.
mogenic coactivator PGC-1. Cell. 1999;98:115–124. doi: 10.1016/ 197. Pillai VB, Sundaresan NR, Gupta MP. Regulation of Akt signaling by
S0092-8674(00)80611-X. sirtuins: its implication in cardiac hypertrophy and aging. Circ Res.
180. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, Kelly DP. 2014;114:368–378. doi: 10.1161/CIRCRESAHA.113.300536.
Peroxisome proliferator-activated receptor gamma coactivator-1 pro- 198. Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, Yu X, Tian B, Wagner
motes cardiac mitochondrial biogenesis. J Clin Invest. 2000;106:847– T, Vatner SF, Sadoshima J. Sirt1 regulates aging and resistance to oxida-
856. doi: 10.1172/JCI10268. tive stress in the heart. Circ Res. 2007;100:1512–1521. doi: 10.1161/01.
181. Bayod S, Del Valle J, Lalanza JF, Sanchez-Roige S, de Luxán-Delgado RES.0000267723.65696.4a.
B, Coto-Montes A, Canudas AM, Camins A, Escorihuela RM, Pallàs M. 199. Hsu CP, Odewale I, Alcendor RR, Sadoshima J. Sirt1 protects the heart
Long-term physical exercise induces changes in sirtuin 1 pathway and from aging and stress. Biol Chem. 2008;389:221–231. doi: 10.1515/
oxidative parameters in adult rat tissues. Exp Gerontol. 2012;47:925– BC.2008.032.
935. doi: 10.1016/j.exger.2012.08.004. 200. Hafner AV, Dai J, Gomes AP, Xiao CY, Palmeira CM, Rosenzweig A,
182. Kavazis AN, Smuder AJ, Powers SK. Effects of short-term endurance Sinclair DA. Regulation of the mPTP by SIRT3-mediated deacetylation
exercise training on acute doxorubicin-induced FoxO transcription in of CypD at lysine 166 suppresses age-related cardiac hypertrophy. Aging
cardiac and skeletal muscle. J Appl Physiol (1985). 2014;117:223–230. (Albany NY). 2010;2:914–923.
doi: 10.1152/japplphysiol.00210.2014. 201. Pillai VB, Sundaresan NR, Jeevanandam V, Gupta MP. Mitochondrial
183. Safdar A, Little JP, Stokl AJ, Hettinga BP, Akhtar M, Tarnopolsky MA. SIRT3 and heart disease. Cardiovasc Res. 2010;88:250–256. doi:
Exercise increases mitochondrial PGC-1alpha content and promotes 10.1093/cvr/cvq250.
nuclear-mitochondrial cross-talk to coordinate mitochondrial biogenesis. 202. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP.
J Biol Chem. 2011;286:10605–10617. doi: 10.1074/jbc.M110.211466. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-
184. Little JP, Safdar A, Cermak N, Tarnopolsky MA, Gibala MJ. Acute dependent antioxidant defense mechanisms in mice. J Clin Invest.
endurance exercise increases the nuclear abundance of PGC-1alpha in 2009;119:2758–2771. doi: 10.1172/JCI39162.
Roh et al   Exercise and Cardiac Aging   295

203. Ferrara N, Rinaldi B, Corbi G, Conti V, Stiuso P, Boccuti S, Rengo 213. Ristow M, Zarse K, Oberbach A, Klöting N, Birringer M, Kiehntopf M,
G, Rossi F, Filippelli A. Exercise training promotes SIRT1 activ- Stumvoll M, Kahn CR, Blüher M. Antioxidants prevent health-promot-
ity in aged rats. Rejuvenation Res. 2008;11:139–150. doi: 10.1089/ ing effects of physical exercise in humans. Proc Natl Acad Sci U S A.
rej.2007.0576. 2009;106:8665–8670. doi: 10.1073/pnas.0903485106.
204. Planavila A, Iglesias R, Giralt M, Villarroya F. Sirt1 acts in association 214. Forman DE, Manning WJ, Hauser R, Gervino EV, Evans WJ, Wei JY.
with PPARα to protect the heart from hypertrophy, metabolic dysregula- Enhanced left ventricular diastolic filling associated with long-term en-
tion, and inflammation. Cardiovasc Res. 2011;90:276–284. doi: 10.1093/ durance training. J Gerontol. 1992;47:M56–M58.
cvr/cvq376. 215. Arbab-Zadeh A, Dijk E, Prasad A, Fu Q, Torres P, Zhang R, Thomas JD,
205. Sugden MC, Caton PW, Holness MJ. PPAR control: it’s SIRTainly as easy Palmer D, Levine BD. Effect of aging and physical activity on left ven-
as PGC. J Endocrinol. 2010;204:93–104. doi: 10.1677/JOE-09-0359. tricular compliance. Circulation. 2004;110:1799–1805. doi: 10.1161/01.
206. Palacios OM, Carmona JJ, Michan S, Chen KY, Manabe Y, Ward JL III, CIR.0000142863.71285.74.
Goodyear LJ, Tong Q. Diet and exercise signals regulate SIRT3 and ac- 216. Prasad A, Popovic ZB, Arbab-Zadeh A, Fu Q, Palmer D, Dijk

tivate AMPK and PGC-1alpha in skeletal muscle. Aging (Albany NY). E, Greenberg NL, Garcia MJ, Thomas JD, Levine BD. The ef-
2009;1:771–783. fects of aging and physical activity on Doppler measures of dia-
207. Lanza IR, Short DK, Short KR, Raghavakaimal S, Basu R, Joyner MJ, stolic function. Am J Cardiol. 2007;99:1629–1636. doi: 10.1016/j.
McConnell JP, Nair KS. Endurance exercise as a countermeasure for ag- amjcard.2007.01.050.
ing. Diabetes. 2008;57:2933–2942. doi: 10.2337/db08-0349. 217. Edelmann F, Gelbrich G, Düngen HD, Fröhling S, Wachter R,

208. Rodgers JT, Lerin C, Gerhart-Hines Z, Puigserver P. Metabolic adap- Stahrenberg R, Binder L, Töpper A, Lashki DJ, Schwarz S, Herrmann-
tations through the PGC-1 alpha and SIRT1 pathways. FEBS Lett. Lingen C, Löffler M, Hasenfuss G, Halle M, Pieske B. Exercise training
2008;582:46–53. doi: 10.1016/j.febslet.2007.11.034. improves exercise capacity and diastolic function in patients with heart
209. Anderson R, Prolla T. PGC-1alpha in aging and anti-aging interven- failure with preserved ejection fraction: results of the Ex-DHF (Exercise
tions. Biochim Biophys Acta. 2009;1790:1059–1066. doi: 10.1016/j. training in Diastolic Heart Failure) pilot study. J Am Coll Cardiol.
bbagen.2009.04.005. 2011;58:1780–1791. doi: 10.1016/j.jacc.2011.06.054.
210. Shefer G, Rauner G, Yablonka-Reuveni Z, Benayahu D. Reduced satel- 218. Fujimoto N, Prasad A, Hastings JL, Arbab-Zadeh A, Bhella PS, Shibata
lite cell numbers and myogenic capacity in aging can be alleviated by S, Palmer D, Levine BD. Cardiovascular effects of 1 year of progressive
endurance exercise. PLoS One. 2010;5:e13307. doi: 10.1371/journal. and vigorous exercise training in previously sedentary individuals older
pone.0013307. than 65 years of age. Circulation. 2010;122:1797–1805. doi: 10.1161/
211. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C.
CIRCULATIONAHA.110.973784.
Mortality in randomized trials of antioxidant supplements for primary 219. Stratton JR, Cerqueira MD, Schwartz RS, Levy WC, Veith RC, Kahn
and secondary prevention: systematic review and meta-analysis. JAMA. SE, Abrass IB. Differences in cardiovascular responses to isoproterenol
2007;297:842–857. doi: 10.1001/jama.297.8.842. in relation to age and exercise training in healthy men. Circulation.
212. Gomez-Cabrera MC, Domenech E, Romagnoli M, Arduini A, Borras 1992;86:504–512.
C, Pallardo FV, Sastre J, Viña J. Oral administration of vitamin C 220. McGuire DK, Levine BD, Williamson JW, Snell PG, Blomqvist CG,
decreases muscle mitochondrial biogenesis and hampers training- Saltin B, Mitchell JH. A 30-year follow-up of the Dallas Bedrest and
induced adaptations in endurance performance. Am J Clin Nutr. Training Study: II. Effect of age on cardiovascular adaptation to exercise
2008;87:142–149. training. Circulation. 2001;104:1358–1366.
Downloaded from http://ahajournals.org by on November 15, 2020

You might also like