You are on page 1of 57

Physiol Rev 98: 419 – 475, 2018

Published December 20, 2017; doi:10.1152/physrev.00043.2016

UNDERSTANDING KEY MECHANISMS OF


EXERCISE-INDUCED CARDIAC PROTECTION TO
MITIGATE DISEASE: CURRENT KNOWLEDGE AND
EMERGING CONCEPTS
Bianca C. Bernardo, Jenny Y. Y. Ooi, Kate L. Weeks, Natalie L. Patterson,
and Julie R. McMullen

Baker Heart and Diabetes Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne,
Victoria, Australia; Department of Diabetes, Central Clinical School, Monash University, Victoria, Australia;
Department of Medicine, Central Clinical School, Monash University, Victoria, Australia; and Department of
Physiology, School of Biomedical Sciences, Victoria, Australia

Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding

L
Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current
Knowledge and Emerging Concepts. Physiol Rev 98: 419 – 475, 2018. Published
December 20, 2017; doi:10.1152/physrev.00043.2016.—The benefits of exercise
on the heart are well recognized, and clinical studies have demonstrated that exercise
is an intervention that can improve cardiac function in heart failure patients. This has led to
significant research into understanding the key mechanisms responsible for exercise-induced
cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced
cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other
cardiac cells, organelles, and systems that have received less or little attention and require further
investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular
stress responses to promote survival (heat shock response, ubiquitin-proteasome system, au-
tophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage re-
sponse), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summa-
rize therapeutic strategies targeting known regulators of exercise-induced protection and the
challenges translating findings from bench to bedside. We conclude that technological advance-
ments that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome,
combined with animal and human studies, provide new opportunities for comprehensively defining
the signaling and regulatory aspects of cell/organelle functions that underpin the protective
properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic
targets for heart disease.

I. INTRODUCTION, CLINICAL ... 419 exercise in clinical trials in heart failure patients have been
II. EFFECTS OF EXERCISE ON THE ... 420 well described (72, 88, 172, 298, 327, 449). Low-, moder-
III. GENOME, TRANSCRIPTOME, ... 448 ate-, and vigorous-intensity exercise have all been shown to
IV. THERAPEUTIC STRATEGIES: ... 454 provide some degree of benefit (68, 123, 225, 403).
V. OBSTACLES AND CHALLENGES 460 Whether a dose-response effect exists based on exercise du-
VI. PERSPECTIVES AND CONCLUSIONS 461 ration and/or intensity is debatable but has been reported
by some studies (68, 123, 218, 225, 403). Of note, there are
I. INTRODUCTION, CLINICAL also reports of very intense/extreme exercise causing dam-
PERSPECTIVE, AND OVERVIEW age to the human heart. This has recently been reviewed and
will not be discussed here (109). For the purpose of this
The benefits of regular exercise or physical activity in hu- review, the term exercise has largely been used to reflect
mans on the heart and other organs in settings of health and regular aerobic or endurance exercise, unless otherwise
disease are well recognized (322). Exercise is known to stated.
reduce the risk of cardiovascular disease and cardiac events
in males and females of the young and aged. The survival Given many individuals do not exercise or are unable to
rate after a cardiovascular event is also greater in physically exercise, there has been intense investigation into delineat-
active individuals compared with more sedentary individu- ing the key mechanisms responsible for exercise-induced
als (39, 68, 205, 224, 291, 436), and the beneficial effects of protection. Exercise represents a very complex stimulus af-

0031-9333/18 Copyright © 2018 the American Physiological Society 419


BERNARDO ET AL.

fecting numerous organs and cell types. Exercise has direct and the challenges translating findings from bench to bed-
beneficial effects on multiple cell types within the heart, but side.
also has effects on other systems (e.g., vascular system, skel-
etal muscle, brain, adipose tissue) which can have second-
ary effects on the heart (4, 151, 198, 206, 371). The cross-
II. EFFECTS OF EXERCISE ON THE
CARDIOVASCULAR SYSTEM
talk between numerous organs and tissues in health and
disease has emerged as an important piece of the puzzle
when understanding disease processes, progression, preven- To meet the oxygen demands of muscle in response to ex-
tion, and treatment (188, 321). The aim of harnessing or ercise, a number of changes take place to allow the heart to
exploiting the beneficial effects of exercise represents an pump more blood each minute to increase cardiac output
three- to sixfold. During settings of acute exercise, the body
active and exciting area of research. By comprehensively
initially responds with activation of the sympathetic ner-
understanding the factors underlying the protective proper-
vous system and downregulation of the parasympathetic
ties of exercise, the hope is that we can develop improved
nervous system. Release of catecholamines [epinephrine
therapies including preventative and treatment strategies.
(E), norepinephrine (NE)] increase heart rate and cardiac
The goal of this review is to highlight known underlying contractility, while blood flow to the heart and exercising
mechanisms responsible for exercise-induced cardiac pro- skeletal muscle is increased via local vasodilator mecha-
tection, as well as to highlight new avenues for research. nisms (FIGURE 1A). This together with vasoconstriction in
Studies in genetic mouse models have been invaluable in the periphery to maintain/increase blood pressure leads to
allowing investigators to use a reductionist approach to an increase in blood flow back to the heart (venous return).
identify key mediators of exercise-induced protection, and a Collectively, the increase in heart rate and stroke volume
description of these studies represents a large portion of this increases cardiac output (159) (FIGURE 1A). The increased
review. While different modes and intensity of exercise have blood flow to the heart during exercise stretches the walls of
been tested in humans with respect to cardiovascular dis- the heart, leading to an increase in the force of contraction
ease, the most commonly used exercise rodent models to (Frank-Starling mechanism) and the quantity of blood leav-
understand the beneficial effects of exercise at the cellular ing the heart. In response to chronic or long-term regular
and molecular level include chronic aerobic models such as exercise which is accompanied by increased sympathetic
swim training (2–5 wk: a ramp protocol from 10 to 90 min activity, and release of hormones and growth factors, the
sessions twice a day, 5–7 days/week), treadmill running heart walls thicken and the size of the heart increases (FIG-
(4 – 6 wk: a ramp protocol from 10 min/day to 60 min/day, URE 1B). The process of cardiac enlargement normalizes
5–7 days/week; ⬍70% VO2 maximal oxygen uptake), and wall stress/tension [Laplace’s law: wall stress ⫽ (pressure ⫻
voluntary free wheel running (4 – 8 wk) (29). Unless other- radius)/(2 ⫻ wall thickness)], allowing the heart to main-
wise stated, these or comparable protocols have been used tain normal heart function. This type of cardiac enlarge-
when referring to swim, treadmill, or voluntary running ment is referred to as physiological cardiac hypertrophy
throughout the text. and is very different at the functional, histological, and
molecular level to pathological cardiac hypertrophy,
In this review we discuss in detail the effects of exercise on which occurs in cardiac disease settings (35). Physiolog-
cardiomyocytes as well as other cardiac cell types, organ- ical cardiac hypertrophy is characterized by increased
elles, and systems that have received less or little attention heart mass, normal/enhanced cardiac function, and no
evidence of cardiac fibrosis [excessive deposition of ex-
and require much further investigation. This includes the
tracellular matrix (ECM) proteins] or cell death (FIGURE
impact of exercise on cardiac excitation and contraction,
1B). In contrast, pathological cardiac hypertrophy is typ-
mitochondrial adaptations, cellular stress responses to pro-
ically characterized by depressed heart function, cardiac
mote survival (heat shock response, ubiquitin-proteasome
fibrosis, and cell death; increased expression of cardiac
system, autophagy-lysosomal system, endoplasmic reticu-
stress markers [e.g., atrial natriuretic peptide (ANP) and
lum unfolded protein response, DNA damage response), B-type natriuretic peptide (BNP)]; and a fall in proteins
extracellular matrix, inflammatory response, and organ to critical for cardiac contraction [e.g., ␣-myosin heavy
organ crosstalk. We summarize technological advance- chain (␣MHC) and sarco/endoplasmic reticulum Ca2⫹-
ments that allow for in-depth profiling of the genome, tran- ATPase (SERCA2a)]. Pathological cardiac hypertrophy
scriptome, proteome, and metabolome in response to exer- is also an independent risk factor for heart failure (35).
cise. We also describe how these methodologies combined
with animal and human studies provide new opportunities
for comprehensively defining cellular and molecular mech- A. Exercise-Induced Cardiac Remodeling
anisms that underpin the protective properties of exercise.
Finally, this is followed by a description of therapeutic Exercise-induced cardiac remodeling is a complex process
strategies targeting known regulators of exercise-induced that involves exercise-induced stimuli (catecholamines, me-
protection in small animals, large animals, and clinical trials chanical stretch, hormones, and growth factors) regulating

420 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

A ACUTE B CHRONIC
EXERCISE EXERCISE

Sympathetic activity
Hormones
Muscular &
respiratory pumps ↑ Sympathetic ↓ Parasympathetic
engaged activity activity Growth factors

AD

↑E Physiological cardiac hypertrophy


↑ NE &
↑ NE

↑ Venous return Sinoatrial node ↑ Heart mass


Normal/enhanced heart function
SAN
↑ EDV Protection against fibrosis, cell death &
heart failure

↑ Cardiac contractility
Frank Starling
mechanism

↑ Stroke volume ↑ Heart rate

↑ Cardiac output

FIGURE 1. Systemic and cardiac effects of acute and chronic exercise. A: an overview of the effects of acute
exercise on the cardiovascular, sympathetic nervous system, and adrenal medulla, which together increase
cardiac output. B: chronic or long-term exercise is associated with increased sympathetic activity as well as
release of hormones and growth factors that act as stimuli to regulate physiological cardiac hypertrophy.
Physiological heart growth protects the heart from fibrosis, cell death, and heart failure. E, epinephrine; EDV,
end-diastolic volume; NE, norepinephrine. This figure provides a general overview of acute and chronic changes
with aerobic exercise such as jogging, swimming, and cycling. However, it is noteworthy that exercise of
different types, duration, and intensity can have varying effects.

numerous processes within cardiac cells. The adult mam- (328). In the subsequent section we discuss the effects of
malian heart contains multiple cell types including cardiac exercise on cell types, organelles, and key processes within
myocytes, fibroblasts, endothelial cells, leukocytes, and mu- the heart including 1) adaptations to cardiac myocytes in-
ral cells (vascular smooth muscle cells and pericytes). To cluding hypertrophy and proliferation, 2) vascular adapta-
date, mechanisms within cardiac myocytes are best de- tions, 3) electrical activity and excitation-contraction (E-C)
scribed. It is generally accepted that cardiac myocytes rep- coupling, 4) mitochondrial adaptations (energy metabolism
resent 30 – 40% of the cell population in the adult rodent and redox status), 5) cellular stress responses (survival and
and human heart, and this represents 70 – 85% of the death), 6) maintenance of the ECM, and 7) communication
heart’s volume. The percentage and contribution of non- with other organs (FIGURE 2).
myocytes has remained debatable, largely due to the use of
different analytical methods and inadequate tools/markers
(328, 465). However, based on a recent study using a com- B. Adaptations to Cardiac Myocytes:
bination of genetic tools and cellular markers, the adult Hypertrophy and Proliferation
mouse heart consists of ~30% cardiac myocytes, 45% en-
dothelial cells (majority representing vascular endothelial In the adult heart, cardiac enlargement occurs largely due
cells), 11% fibroblasts, 8% mural cells, and 6% leukocytes to a parallel increase in cardiac myocyte size because the

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 421


BERNARDO ET AL.

VASCULAR ADAPTATIONS

CARDIAC HYPERTROPHY
EXCITATION-CONTRACTION COUPLING ↑eNOS ↑NO MYOCYTE GROWTH & PROLIFERATION

Hypertrophy
Angiogenesis

Myocyte
Myocyte

EXERCISE Proliferation

Myocyte

MITOCHONDRIAL ADAPTATIONS

Differentiation &
↑ Sympathetic activity Myocytes
Proliferation
eCSC

↑ Hemodynamic load &


mechanical stretch
ANTI-FIBROTIC ACTIONS
Release of hormones &
CELLULAR STRESS RESPONSE growth factors
Extracellular matrix in disease settings
Proteasome-ubiquitin ↑ Collagen ↑ TIMPs ↓ MMPs
system

Misfolded protein COMMUNICATION


WITH OTHER ORGANS
ER stress Fibrosis
Myocytes
unfolded protein Liver
response

Adipose
tissue
Skeletal
muscle
FIGURE 2. Effects of exercise on cardiac and vascular cells, and cross talk with other organs. In response
to exercise, there are direct effects on different cell types, organelles, and biological processes within the heart
as well as other organs which may have secondary effects on the heart and the rest of the body. These include
myocyte hypertrophy and proliferation, vascular adaptations, excitation-contraction coupling, mitochondrial
adaptations, cellular stress responses, antifibrotic actions, and communication with other organs. The illus-
tration of the female jogging in this figure and subsequent figures has been used as a general symbol to
represent “exercise.” Specific details regarding the type of exercise, duration, and intensity are provided within
the text or TABLE 1.

heart has a very limited capacity for myocyte prolifera- liferation in mice (via administration of an antineoplastic
tion or renewal (331, 390, 391). In response to exercise, agent 5-fluorouracil) before swim training did not pre-
hypertrophy of cardiac myocytes also appears to be the vent exercise-induced heart growth, suggesting that car-
predominant factor responsible for heart enlargement diac cell proliferation is not required for physiological
(62, 269, 446). However, there are also studies to sup- cardiac hypertrophy (19). However, cardiac cell prolifer-
port the existence of cardiomyocyte proliferation in the ation does appear to be essential for exercise-induced
adult mammalian heart and enhancement in a setting of protection against ischemia-reperfusion injury because
exercise. Exercise training in mice (treadmill running or the same agent (5-fluorouracil) abolished the cardiopro-
swimming) was reported to induce cardiac myocyte pro- tective effect of exercise (19). The molecular pathways
liferation and renewal (up to ~7%), potentially from en- associated with cardiac myocyte hypertrophy and prolif-
dogenous cardiac stem/progenitor cells (eCSCs) or pre- eration are discussed below.
existing cardiac myocytes (25, 44, 373, 445, 452). How-
ever, further studies examining the significance of Numerous growth factors are released in response to
exercise-induced cardiomyocyte proliferation are re- exercise and have the capacity to induce cardiac enlarge-
quired. A recent study showed that inhibition of cell pro- ment (i.e., physiological cardiac hypertrophy). These in-

422 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

clude insulin-like growth factor 1 (IGF1), hepatocyte 1. IGF1-PI3K-Akt signaling axis


growth factor (HGF), platelet-derived growth factor
(PDGF), vascular endothelial growth factor (VEGF), and IGF1 is a growth factor released in response to exercise in
neuregulin-1 (NRG1) (83, 445, 459). Of these, IGF1 and animal models and elite athletes (212, 460), and the cardiac
signaling downstream of the IGF1 receptor (IGF1R), i.e., formation of IGF1 was elevated in male soccer players with
phosphoinositide 3-kinase (PI3K; class 1A: p110␣ and exercise-induced hypertrophy in comparison to healthy in-
␤)-Akt signaling, is the best-characterized pathway for dividuals (301). Based on studies utilizing gain- and loss-of-
mediating exercise-induced myocyte hypertrophy. How- function genetically modified mouse models, it is now well
ever, many of the other aforementioned growth factors recognized that the IGF1-IGF1R- insulin receptor substrate
are also able to activate PI3K-Akt signaling (FIGURE 3). It (IRS)-PI3K (class 1A)-phosphoinositide-dependent protein
kinase-1 (PDK1)-Akt1 axis plays a key role in regulating
has been suggested that exercise promotes cardiac cell
exercise-induced heart growth (FIGURE 3 AND TABLE 1).
proliferation and renewal via 1) sympathetic activity and
This section is focused on mechanisms within cardiac myo-
growth factors (IGF1, NRG1, and HGF) inducing eCSC cytes, but it is noteworthy that PI3K-Akt signaling and
proliferation (431, 452), and 2) bone morphogenetic downstream regulators also play a role in endothelial cells
protein 10 (BMP-10) and transforming growth factor ␤1 in response to exercise, as described in section IIC. IGF1
(TGF-␤1) stimulating the differentiation of eCSCs into activates the IGF1R upon binding. This subsequently re-
cardiac lineages (431, 445) (FIGURE 3). cruits the IRS adaptor proteins (IRS1/IRS2) which then ac-

EXERCISE DISEASE SETTINGS


e.g. hypertension
↑ Sympathetic
activity
Growth
Thyroid Growth Factors
hormone NRG1 Synd-4
Growth IGF1 hormone e.g. VEGF, PDGF, HGF
factors
β2-AR IGF1R

ErbB4 β-AR AT-R


eCSCs
IRS1/2 PI3K
p110α/β
P
Signaling contributing
AMPK
to pathological hypertrophy
PGC-1α PDK1 PAK1
P e.g. Calcineurin
JAK2 PKCβ
↑ Survival MAPKs
↑ Proliferation PHLPP1 P P

(IGF1, NRG1, HGF) P P AKT1 GSK3β


PP2A STAT3
Physiological
SIX1 signaling ↑ ROS
mTOR
BMP-10 Thyroid
hormone EYA2
Raptor
PRAS40
[mTORC1 eIF2Bε ↑ Fibrosis
↑ Apoptosis
TGF-β1
Myocyte
cytosol 4E-BP1 S6K1
Differentiation into
myocytes, capillaries HEART FAILURE
PHYSIOLOGICAL
HYPERTROPHY

P Nucleus
AKT1
SIX1
Expression of Cardiomyocyte
mTOR exercise/adaptive survival, PHYSIOLOGICAL
EYA2 PI3K? SRF C/EBPβ genes e.g. proliferation HYPERTROPHY
Gata4, αMHC and growth

Growth Thyroid Expression of


IGF1 CITED4 ? cardiac genes
hormone hormones
e.g. MHCs, Serca2a

FIGURE 3. A schematic of the major signaling pathways involved in exercise-induced cardiac hypertrophy.
The major signaling pathway implicated in exercise-induced cardiac hypertrophy is the IGF1-PI3K-Akt pathway.
Thyroid hormones, NRG1 signaling, growth factors, and Synd4 have also been shown to regulate the IGF1-
PI3K-Akt pathway. Physiological signaling has been shown to inhibit pathological cascades (shown in red).
Dashed lines indicate translocation to a different intracellular compartment, and dotted lines indicate mech-
anisms that are not yet well understood. Signaling is complex, and there is cross talk between various
components of the pathways (not all illustrated).

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 423


BERNARDO ET AL.

Table 1. Genes regulating exercise-induced cardiac remodeling


Reference
Gene and Model Exercise Model Observation Nos.

IGF1R: cardiac myocyte Swim exercise: ramp protocol followed Hypertrophic growth blunted in IGF1R 203
specific KO of IGF1R by 90 min sessions twice/day for 4 KO mice.
wk (total ~5 wk)
IGF1R: cardiac myocyte Swim exercise: ramp protocol followed Augmented cardiac hypertrophic 267
specific transgenic by 90 min sessions twice/day for 3 response in IGF1R transgenic.
overexpression of wk (total ~4 wk)
IGF1R
PI3K: cardiac myocyte Swim exercise: ramp protocol followed dnPI3K transgenic mice displayed a 269, 310
specific by 90 min sessions twice/day for blunted hypertrophic response to
dnPI3K(p110␣) 2–3 wk (total ~3–4 wk) exercise compared with Ntg.
transgenic Mitochondrial adaptations also
attenuated.
PI3K: muscle specific Swim exercise: ramp protocol followed Exercise-induced hypertrophy 247
KO of PI3K (p110␣ by 90 min sessions twice/day for 3 attenuated in KO.
and p110␤) wk (total ~4 wk)
IRS1: cardiac myocyte Swim exercise: ramp protocol followed No response to exercise-induced 347
specific deletion by 90 min sessions twice/day for 4 hypertrophy in KO.
knockout of IRS1 wk (total ~5 wk)
IRS2: cardiac myocyte Swim exercise: ramp protocol followed KO mice exhibit baseline cardiac 347
specific deletion by 90 min sessions twice/day for 4 hypertrophy and no response to
knockout of IRS2 wk (total ~5 wk) exercise-induced hypertrophy.
PDK1/PDPK1: cardiac Swim exercise: ramp protocol followed Exercise-induced hypertrophy 307
myocyte specific by 90 min sessions twice/day for 4 prevented in cPDPk1⫹/–. No effect
heterozygote wk (total ~5 wk) on mitochondrial adaptations
knockout of PDPK1 associated with exercise.
(cPDPk1⫹/–)
PAK1/(potential Treadmill endurance running for 6 wk Exercise-induced hypertrophy, cardiac 85
PDK2): global KO (55–60% maximal aerobic velocity, contractility, myofilament calcium
15% incline, 60 min/day, 5 sensitivity, and posttranslational
days/week) modification of sarcomeric proteins
were blunted in PAK1 KO mice.
Akt1: global KO Swim exercise: 90 min sessions Exercise-induced hypertrophy 91
twice/day for total ~3 wk attenuated in KO.
Akt1 KO and kdAkt Swim exercise: ramp protocol followed Exercise-induced hypertrophy blunted 310
by 90 min sessions twice/day for 2 but no effect on mitochondrial
wk (total ~3 wk) adaptations.
HSF1: HSF1-deficient Voluntary wheel running for 4 wk Equivalent hypertrophy in WT and 359
heterozygote HSF1⫹/–. Cardiac dysfunction in
(HSF1⫹/–) mice HSF1⫹/–.
Heterozygote C/EBP␤ Swim exercise: ramp protocol followed Under basal condition, heterozygote 44
(complete KO not by 90 min sessions twice/day for 2 mice display cardiac hypertrophy
viable) wk (total ~3 wk) associated with myocyte
hypertrophy and proliferation.
There is a minimal increase in
heart size in response to swim
training.
Protein phosphatase Swim exercise: ramp protocol followed Exercise-trained PHLPP1 KO mice 282
PHLPP1: KO by 90 min sessions twice/day for 2 showed an exaggerated
wk (total ~3 wk) physiological hypertrophic
response.
Activation of Akt by PHLPP1 KO mice had elevated
PHLPP1 deletion angiopoietin-2 and VEGF-A levels
and increased myocardial capillary
density compared with control
mice.
PRAS40 4 wk of voluntary wheel running Physiological hypertrophy was not 429
blocked by PRAS40 overexpression
but was inhibited by phospho-dead
mutant PRAS40TA.
Continued

424 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

Table 1.—Continued
Reference
Gene and Model Exercise Model Observation Nos.

eNOS KO 8 wk of voluntary wheel running (~7 Exercise was unable to attenuate MI- 90
km/day) induced ventricular remodeling,
hypertrophy, fibrosis, and apoptosis
in eNOS KO mice.
Of note,eNOS KO mice Skeletal muscle may be the source of 59
were reported to eNOS.
exercise to a lesser
degree than WT in
other references but
not this reference
␤3-AR KO 4 wk of voluntary wheel running Exercise was less effective in 58
exercise protecting KO mice from IR injury.
Of note, ␤3-AR KO
exercise to a lesser
degree than WT
(~50% less)
␤1-AR KO Graded treadmill exercise (speed and The normal exercise-induced increase 351
angle inclination were progressively in heart rate was blunted in ␤1-AR
increased) KO, but this did not impact on
maximal exercise capacity.
RyR2-S2808A Swimming in untrained mice for 5 min Mutant mice had impaired exercise 378
mutation (knock-in capacity.
mouse-global)
Mouse lacks the major Note: Authors cannot exclude the
PKA phosphorylation possibility that extracardiac factors
site on RyR2 contributed to the impaired
exercise capacity.
NOS1-KO mice 8 wk aerobic interval training program Exercised NOS1-KO mice did not have 354
(treadmill) cardiac hypertrophy or increased
VO2max compared with sedentary
NOS1-KO mice.
nNOS-KO mice 8 wk aerobic interval training program Exercised nNOS-KO mice have 353
(treadmill) decreased ejection fraction and
increased ROS production.
nNOSOE No exercise protocol–goal to nNOSOE myocytes exhibited 353
(overexpression) mice determine if overexpression could enhanced contraction and
recapitulate the exercise training relaxation, and a higher VO2max.
state in sedentary lifestyle
Synd4 KO mice Swim exercise: ramp protocol followed KO mice exhibited a blunted cardiac 453
by 90 min sessions twice/day for 3 hypertrophic response compared
wk (total ~4 wk) with WT mice following 4 wk of
swim training.
Male global AMPK␣2 Swim training for 4 wk in total (50 Exercise training in control mice 248
KO min/day, 6 days/week following a attenuated isoproterenol (␤-AR
ramp protocol) agonist)-induced ROS and fibrosis
(isoproterenol administered in the
last 2 wk of swim training).
Protection was not observed in
AMPK␣2 KO mice. Note: AMPK ␣2
is abundant in skeletal muscle.
Comprehensive evidence
demonstrating KO mice exercised
to the same degree as control
mice was not included.
Continued

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 425


BERNARDO ET AL.

Table 1.—Continued
Reference
Gene and Model Exercise Model Observation Nos.

Cardiac-specific Swim training for 22 days in total (90 Gata4-deleted mice developed 313
deletion of Gata4 min/twice a day, 7 days/week hypertrophy in response to swim
(reduced Gata4 by following a ramp protocol) training, but the magnitude was
E17.5 and into blunted compared with control
adulthood by crossing mice (36% increase in VW/BW in
floxed mice with Cre control mice, 26% increase in
from the ␤MHC Gata4-deleted mice). Note: Gata4-
promoter) deleted mice display cardiac
dysfunction under basal conditions
by 12 wk of age and apoptosis. It
was not clear what age mice began
swimming, but it is stated that
mice were monitored continuously
to ensure equal exertion.
Inducible, cardiac Basal phenotype 15–20% increase in heart weight, 37
specific CITED4 concentric hypertrophy without an
transgenic mice increase in fibrosis, no difference in
lung weight, no difference in
fractional shortening. This
phenotype is similar to what is seen
after endurance training in mice.

BW, body weight; KO, knockout; MI, myocardial infarction; VW, ventricular weight; WT, wild type. Unless stated otherwise, most swim studies
have used a ramp protocol typically starting at 10 min/twice a day and increasing 10 min/day ⱕ90 min sessions/twice a day.

tivates the PI3K-PDK1-Akt1 pathway. The p110␣ isoform as a linear pathway for simplicity, it is recognized that sub-
of PI3K (class 1A PI3K) was the first gene shown to play a stantial crosstalk between other pathways exist. For exam-
critical role for exercise-induced cardiac hypertrophy. Car- ple, exercise-induced cardiac hypertrophy was attenu-
diac-myocyte specific transgenic mice expressing a domi- ated in cardiac-specific KO models of IGF1R, IRS1, IRS2,
nant negative PI3K mutant (dnPI3K) displayed a markedly and cardiac-specific dnPI3K mice, but different down-
reduced hypertrophic response to 4 wk of chronic swim stream mechanisms have been implicated [e.g., decreased
training compared with nontransgenic littermates (269). In pAkt, decreased pJAK2, increased dephosphorylation of
contrast, dnPI3K transgenic mice developed a robust hyper- Akt, and altered regulation of adenosine monophos-
trophic response to the pathological stimulus of chronic phate-activated protein kinase (AMPK)] (203, 269, 310,
pressure overload (269). Subsequent to this discovery, the 347) (FIGURE 3).
critical role of PI3K has been confirmed by other studies,
and the role of genes upstream and downstream of PI3K
2. Other regulators of Akt signaling
have been interrogated. Swim exercise training in mice for
3–5 wk induces a reproducible increase in heart size that
was blunted or prevented in mice with deletion or reduced As described earlier, growth factors in addition to IGF1 are
activity of IGF1R, IRS1, IRS2, class IA PI3K(p110␣ ⫹/⫺ elevated in response to exercise and have the potential to act
p110␤), PDK1, and Akt1 (91, 203, 247, 269, 307, 310, via other transmembrane receptors to activate Akt signaling
347). Regulators of Akt and the mammalian target of rapa- (FIGURE 3). The contribution of these alternate upstream
mycin complex 1 (mTORC1), P21-activated kinase (PAK1; regulators in mediating different aspects of exercise-in-
252), and proline-rich Akt substrate of 40 kDa (PRAS40) duced remodeling are not well defined, and some redundan-
have also been identified as key regulators of exercise-in- cies may exist. Syndecan-4 (a transmembrane proteogly-
duced physiological heart growth (85, 429) (FIGURE 3, TA- can) was upregulated in rat hearts following 4 wk of high-
BLE 1). intensity aerobic swim training, and swim-induced cardiac
hypertrophy was largely prevented in Syndecan-4 KO mice
Distal to Akt, signaling and crosstalk become more com- (453). However, this was a global KO mouse model, and
plex. For instance, deletion of ribosomal S6 kinases (S6K1 direct measures of exercise capacity between groups (con-
and S6K2; downstream effectors considered important for trol and KO) were not presented (453). Akt was implicated
protein synthesis) had no effect on swim-induced cardiac in the blunted hypertrophic response because 1) the phos-
hypertrophy (268). It remains unclear whether compensa- phorylation of Akt was lower in swim-trained Syndecan-4
tory mechanisms were responsible for exercise-induced car- KO mice, and 2) in cultured cardiac myocytes, Syndecan-4
diac enlargement in S6K1/2 knockout (KO) mice. In addi- overexpression induced myocyte enlargement, which was
tion, while the IGF1 signaling cascade is typically presented blocked with an Akt inhibitor (453).

426 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

A novel Akt phosphatase, PHLPP1 (PH domain leucine- with the regression of pressure-overload induced hypertro-
rich repeat protein phosphatase), has also been shown to phy in rats. Eya2 was confirmed as an antihypertrophic
regulate exercise-induced hypertrophy. PHLPP1 dephos- gene in neonatal cardiomyocytes (454). Cardiac-specific
phorylates Akt, which in turn terminates Akt signaling. Un- transgenic expression of Eya2 in mice (⬃20- to 30-fold
der basal conditions, PHLPP1 KO mice had increased Akt higher protein than control) was reported to induce a mild
activity but normal heart size and function. However, when hypertrophic response (~5– 8% increase in heart and myo-
PHLPP1 KO mice underwent forced swim training for 20 cyte size) which was characteristic of physiological hyper-
days, cardiac hypertrophy was accentuated compared with trophy (normal heart function) (227). In a later study from
wild-type exercised mice (282). the same investigators, it was shown that Eya2 protein ex-
pression was increased in hearts of mice subjected to swim
3. Transcription factors regulated by exercise and exercise for 4 wk (226). However, in this subsequent report
Akt signaling which further characterized the same Eya2 transgenic mice,
a mild basal hypertrophic phenotype was not observed
CCAAT/enhancer binding protein ␤ (C/EBP␤; encoded by (226). However, it was demonstrated that both swim train-
CEBPB) and Cbp/p300-interacting transactivator with ED- ing in wild-type mice and transgenic expression of Eya2
rich carboxy-terminal domain 4 (CITED4; encoded by were associated with increased expression of a physiologi-
CITED4) have been identified as critical regulators of phys- cal regulator in the heart, mammalian target of rapamycin
iological cardiac hypertrophy and cardiomyocyte prolifer- (mTOR). In addition, Eya2 was shown to act as a transcrip-
ation (44) (FIGURE 3). C/EBP␤ expression was downregu- tional coactivator in complex with another transcription
lated in hearts of mice following an acute bout or 2 wk of factor, Six1, to directly upregulate mTOR. Finally, thyroid
aerobic exercise training, but was unchanged in hearts of hormone (T3; inducer of physiological cardiac growth) was
mice with pathological cardiac hypertrophy induced by shown to increase Eya2 and Six1 protein levels in isolated
aortic banding (44) and upregulated in the rat heart 3, 6, neonatal cardiac myocytes (226) (FIGURE 3).
and 12 h post-myocardial infarction (MI) (97). siRNA
knockdown of CEBPB in neonatal rat cardiomyocytes in- 4. Benefits of physiological heart growth and
duced hypertrophy and increased cell number, demonstrat- components of IGF1-PI3K-Akt signaling in disease
ing that C/EBP␤ is a negative regulator of cardiomyocyte settings
hypertrophy and proliferation. Additionally, heterozygote
C/EBP␤ mice displayed many of the cardiac phenotypes Studies in exercise-trained rodents and genetic models
seen in exercised mice, i.e., increased heart and cardiomyo- where components of the IGF1-PI3K-Akt pathway have
cyte size and improved cardiac function. Although mice been regulated show that physiological growth/signaling is
with genetically reduced C/EBP␤ levels displayed increased able to attenuate pathological heart enlargement/remodel-
exercise capacity under basal conditions, their hypertrophic ing and/or cardiac myocyte hypertrophy in cardiac disease
response following swim exercise training was not further models (181, 266, 349, 415, 434, 437, 446). The molecular
elevated. C/EBP␤ expression was shown to be regulated by mechanisms responsible will require further study but in-
Akt1, as adenoviral overexpression of Akt1 in neonatal rat clude the ability of the IGF1-PI3K-Akt axis to inhibit sig-
cardiomyocytes reduced C/EBP␤ mRNA expression, and naling cascades downstream of GPCR, which have been
adenoviral transfer of a dominant negative Akt1 mutant implicated in inducing pathological cardiac hypertrophy
increased C/EBP␤ mRNA levels (44). C/EBP␤ was shown to (35, 439) (FIGURE 3). IGF1R and constitutively active (ca)
interact with serum response factor (SRF), a transcription PI3K transgenic mice displayed a blunted hypertrophic re-
factor that promotes transcription of ␣MHC and GATA sponse to pressure overload, whereas dnPI3K transgenic
binding protein 4 (GATA4) (44). Boström et al. (44) postu- mice and Akt1 KO mice showed an exaggerated hypertro-
lated that C/EBP␤ sequesters SRF; thus reductions in phic response; regulation of extracellular signal-regulated
C/EBP␤ expression would lead to the release of SRF, in turn kinase (ERK) activation was implicated (266, 267, 446).
enhancing binding of SRF to serum response elements. Furthermore, heterozygote C/EBP␤ mice were resistant to
Downregulation of C/EBP␤ induces cardiomyocyte prolif- pathological remodeling and cardiac dysfunction induced
eration via upregulation of CITED4 expression, a transcrip- by aortic banding (44), and CITED4 transgenic mice were
tion factor that is upregulated in the heart by exercise (44). protected in a model of ischemia-reperfusion injury via a
In the adult heart, inducible cardiac specific expression of mechanism mediated by mTORC1 (37). Eya2 transgenic
CITED4 resulted in a heart phenotype reminiscent of what mice were also protected against pressure overload-induced
was seen after endurance training in mice, that is, increased pathological remodeling due to aortic constriction. How-
heart weight by 15–20%, preserved cardiac function, and ever, the degree of protection differed in two studies. In the
no evidence of fibrosis (37). initial report, Eya2 transgenic mice displayed a blunted hy-
pertrophic response to aortic constriction (227), but a later
Another transcription factor regulated by exercise is the report identified no significant difference in heart size (226).
eyes absent 2 (Eya2) gene. The potential role of Eya2 in the However, in both studies, Eya2 transgenic mice showed
heart was first identified when profiling genes associated evidence of protection compared with control mice based

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 427


BERNARDO ET AL.

on cardiac function, fibrosis, or molecular profile. In addi- 6. Essential processes accompanying exercise-
tion, there was evidence of preserved physiological signal- induced cardiac hypertrophy
ing in the disease model (226, 227). That is, in Eya2 trans-
genic hearts subjected to aortic-constriction, SERCA2a For the maintenance of cardiac function with exercise-in-
gene expression was preserved, and there was increased or duced physiological hypertrophy, cardiac growth must be
preserved expression of regulators induced with physiolog- accompanied by numerous events including vascular adap-
ical hypertrophy (IGF1R, PI3K, Akt, gp130 and Hsp70) tations, regulation of E-C coupling, mitochondrial adapta-
and genes involved with cell proliferation and cell cycle tions to regulate cellular energy and reactive oxygen species
regulation (e.g., CREB1, CDK5, GATA4) (226, 227). The (ROS), cellular stress responses, and regulation of the ECM.
cardioprotective effects of Eya2 are considered to be medi- These factors are described in section II, C–G.
ated via the Eya2/Six1-mTOR signaling axis, and possibly
PI3K (226). C. Adaptations to Endothelial Cells

5. Relevance of the IGF1 pathway in humans: Endothelial cells line the heart chambers (endocardium)
exercise and disease settings and the small intramyocardial coronary arteries/capillaries.
The endothelium acts as a structural barrier but also re-
There is also evidence from human studies linking IGF1 leases a number of substances that can regulate cardiac
with physiological cardiac hypertrophy and protection structure and function, e.g., growth factors, vasocon-
against cardiovascular disease. Cardiac formation of IGF1 strictors, vasodilators (52, 182). The vascular endothe-
was elevated in trained competitive soccer players com- lium of the coronary circuit controls coronary blood sup-
ply to the heart via interactions with smooth muscle cells
pared with healthy controls, which was positively corre-
(FIGURE 4A). In contrast, endothelial cells in the myocar-
lated with left ventricular (LV) mass index, LV end diastolic
dial capillaries and in the endocardial endothelium are in
dimension index, and LV posterior wall thickness (301).
close proximity to cardiac myocytes, allowing for direct
Furthermore, genotyping studies have identified a number
communication between cardiac myocytes and endothe-
of polymorphisms in the genes encoding IGF1 and the
lial cells (FIGURE 4B) (52).
IGF1R that correlate with LV mass index (187). For in-
stance, a genotyping study of elite endurance athletes found 1. Vascular adaptations
that polymorphisms in the genes encoding IGF1, IGF1R,
and myostatin (a negative regulator of IGF1 signaling) cor- Vascular adaptations can broadly be classified as functional
relate with LV mass (187). The effects of these polymor- (changes in vasomotor control) and structural [angiogene-
phisms on serum IGF1 levels and, more specifically, cardiac sis (proliferation of blood vessels by sprouting from existing
formation of IGF1 are unclear or have not been assessed (9, vessels) and vascular remodeling]. For a comprehensive and
125, 187, 355, 421). Associations between lower serum detailed review on this topic, the reader is referred to Ref-
IGF1 levels with diabetes or cardiac-related disease have erence 222. The subsequent text summarizes some of the
been reported in some, but not all, studies (41, 421), possi- key changes observed with exercise, i.e., vasodilation of the
bly due to environmental factors (125). In a large Dutch coronary vasculature and angiogenesis. The oxygen con-
cohort, absence of the wild-type IGF1 allele was associated sumption requirements of the heart increase with exercise
with lower serum IGF1 levels and an increased risk of type due to the faster heart rate, wall stress, and contractility.
2 diabetes, MI, and LV pathological hypertrophy (41, 421). The oxygen supply to the heart is predominantly enhanced
A polymorphism in IRS1 that reduces PI3K activity in vitro by an increase in coronary blood flow via vasodilation of
was associated with increased MI severity scores in patients the coronary arterioles (159). In response to exercise, re-
with acute MI (242). lease of catecholamines as well as activation of the renin-

FIGURE 4. Role of exercise-induced NO in the endothelium (vascular and cardiac), vascular smooth muscle cells, and cardiac myocytes. A:
vascular endothelium and vascular smooth muscle cells. NO produced from eNOS in the coronary vascular endothelium diffuses into vascular
smooth muscle cells where it causes relaxation and results in vasodilation for increased blood flow. B: cardiac endothelium and cardiac
myocytes. eNOS and nNOS have distinct subcellular locations. eNOS is located in the endothelium and caveolae within cardiac myocytes, and
nNOS is preferentially localized in the SR. Exercise-induced shear stress, catecholamine release (acting on ␤3-AR), and release of growth factors
(e.g., VEGF) from cardiac myocytes (acting via the VEGFR-PI3K-Akt pathway in endothelial cells) contribute to an increase in NO in cardiac
endothelial cells. NO produced in cardiac endothelial cells can promote angiogenesis. The role of eNOS in cardiac myocytes appears to be derived
mainly from the coronary microvascular endothelium. Once NO has diffused into cardiac myocytes, it can inhibit ␤1-AR contractility [i.e.,
␤1-AR-cAMP-PKA induced phosphorylation of the L-type Ca2⫹ channel (LTCC), RyR2 and PLB], reduce mitochondrial respiration and O2
consumption, and induce myocyte relaxation (via cGMP). eNOS in caveolae plays a role in the positive inotropic response to sustained stretch
by increasing the RyR2 open probability. nNOS-derived NO may inhibit Ca2⫹ influx through the L-type Ca2⫹ channels and stimulate Ca2⫹ reuptake
via phosphorylation of PLB.

428 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

A VASCULAR ENDOTHELIUM

EXERCISE &
SHEAR STRESS

Coronary vascular
endothelial cells eNOS NO

Diffusible NO

GTP cGMP
Vascular smooth
muscle cells
RELAXATION

B CARDIAC ENDOTHELIUM

EXERCISE &
SHEAR STRESS

eNOS Akt
Cardiac eNOS NO
endothelial cells
ANGIOGENESIS PI3K
NO

β3 VEGFR
Extracellular matrixFibrosis ROS NO EXERCISE
Myocyte Sympathetic VEGF
EXERCISE Diffusible activity
Sympathetic hypertrophy
activity NE

NE Myocyte
size
Myocyte β1 β3 Caveolae

cAMP NO eNOS
Mitochondrial
T-tubule
respiration
PKA ROS
LTCC cGMP
NO

NO P
nNOS
Stretch PLB
eNOS
SERCA2a
Caveolae RyR2

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 429


BERNARDO ET AL.

angiotensin-aldosterone system and endothelin system all i.e., ⬎3 sessions per week of vigorous aerobic-endurance
act on receptors on the vasculature to cause vasoconstric- exercise for ⱖ 2 yr) (58, 59).
tion to contribute to the redistribution of blood flow to-
wards the heart and exercising muscles. The coronary vas- Voluntary running wheel exercise in mice for 4 wk was
culature of the heart escapes these vasoconstrictor actions associated with increased circulating levels of cat-
via the production of factors such as nitric oxide (NO) echolamines and increased protein expression of cardiac
(159). Exercise is associated with an increase in vascular ␤3-ARs (expression in endothelial cells vs. cardiac myocytes
shear stress due to the increase in heart rate which increases was not specifically assessed) (58). Followup studies in
blood flow, and this is associated with the release of factors ␤3-AR KO mice showed that ␤3-ARs play a critical role in
from the endothelium including NO (18). The sympathetic regulating eNOS/NO levels in response to exercise, and for
nervous system is also activated with exercise, and this also mediating the protective effects of exercise. In a cardiac
contributes to NO production via catecholamines acting on stress model of ischemia-reperfusion injury, 4 wk of volun-
␤3-adrenoceptors (␤3-ARs) (FIGURE 4). tary free wheel running provided protection. The mecha-
nism implicated was exercise-induced catecholamines caus-
NO is generated by NO synthases (NOSs). There are three ing eNOS-NO generation via ␤3-AR stimulation (58). Pro-
NOS isoforms: endothelial NOS (eNOS, NOS3), neuronal tection may also be mediated via mechanisms involving
␤3-AR NO production in cardiac myocytes (described in
NOS (nNOS, NOS1), and inducible NOS (iNOS, NOS2).
section IID1A).
Under physiological settings and in response to exercise,
eNOS and nNOS appear to be the predominant isoforms
responsible for NO generation. iNOS is elevated in the 2. Angiogenesis
heart in settings of cardiac pathology (251, 372). The best
recognized role of NO under basal conditions and in re- In a setting of cardiac myocyte hypertrophy, angiogenesis is
sponse to exercise is the regulation of vascular homeostasis a key response for maintaining perfusion and an adequate
via eNOS. However, more recently, nNOS has also been nutrient supply (50, 442). Evidence from human and ani-
shown to play an important role in exercise-induced cardiac mals studies suggests that angiogenesis/capillary density in-
protection. Small amounts of NO are produced in the heart creases at a similar rate as exercise-induced cardiac enlarge-
by the constitutive expression of eNOS and nNOS. eNOS is ment (104, 158). This is in contrast to cardiac-disease-in-
present in coronary and endocardial endothelial cells, as duced pathological hypertrophy where angiogenesis is
well as cardiac myocytes (FIGURE 4). nNOS is present in inadequate (35). Mechanisms implicated for exercise-in-
cardiac autonomic nerves and ganglia, and preferentially duced angiogenesis and endothelial repair include increased
localized in the sarcoplasmic/endoplasmic reticulum (SR) of VEGF, PI3K-Akt signaling, stem cell mobilization, and
cardiac myocytes (FIGURE 4B) (372). The subsequent sec- miRNAs (e.g., miRNA-126) (371). In addition to commu-
tion describes the role of eNOS related mainly to the vas- nication from endothelial cells to cardiac myocytes, there is
also communication from cardiac myocytes to endothelial
culature. The role of eNOS and nNOS related to calcium
cells. For example, in response to exercise, VEGF is secreted
handling in cardiac myocytes and redox balance is dis-
from cardiac myocytes that signals to endothelial cells to
cussed in section IID1F.
promote angiogenesis via PI3K-Akt signaling (21, 445,
463) (FIGURE 4B). Genetically modified mice with reduced
Molecular mediators and mechanisms implicated or associ-
VEGF in heart and skeletal muscle had reduced muscle
ated with increased eNOS expression and NO production
capillary density, and this was associated with cardiac dys-
in endothelial cells include ␤3-ARs, PI3K-Akt signaling, and
function and reduced exercise capacity on a treadmill (en-
microRNAs (miRNAs; e.g., miRNA-21), described in detail durance and exhaustion tests) (314). However, because
in previous reviews (18, 159, 371). There is substantial VEGF was also reduced in skeletal muscle and this was
communication from endothelial cells to vascular smooth associated with exercise intolerance, interpretation is more
muscle cells and cardiac myocytes, including the diffusion complicated than a cardiac-specific mouse model. Chronic
of NO from endothelial cells to both cell types (463) (FIG- cardiac myocyte selective activation of PI3K via gene deliv-
URE 4). The production of endothelial NO causes relax- ery was associated with enhanced capillary density in nor-
ation of vascular smooth muscle cells (via cGMP signaling) mal adult mice (8 wk associated with physiological hyper-
(FIGURE 4A). NO-induced vasodilatation allows blood flow trophy) and mice with pathological hypertrophy due to aor-
to be matched to metabolic demands during exercise. Fur- tic-banding (10 wk after gene delivery) (434). Furthermore,
thermore, stored NO metabolites in the circulation and mice with expression of caPI3K specifically in cardiac myo-
heart (nitrite and nitrosothiols) can be reduced to NO dur- cytes display physiological hypertrophy with normal heart
ing cardiac stress to protect the heart from ROS, patholog- function, suggesting that angiogenesis was adequate in
ical hypertrophy, and fibrosis (58, 59). Increased NO me- these mice (302, 382). In contrast, while short-term trans-
tabolites have been identified in exercise-trained rodents genic expression of Akt (2 wk) induced physiological hyper-
and humans (e.g., healthy young male endurance athletes, trophy associated with normal function, longer term ex-

430 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

pression (6 wk) was associated with reduced capillary den- jority of cardiac myocytes (atrial and ventricular), an in-
sity and cardiac dysfunction (383). ward sodium current (INa) leads to the rapid depolarization
phase (phase 0), opening of fast Na⫹ channels (largely
D. Cardiac Excitation and Contraction NaV1.5) (112, 297) (FIGURE 5A). This is followed by a brief
period of repolarization (phase 1) due to voltage-gated tran-
The heart is unique, in that it can contract independently of sient outward currents (Ito; atria and ventricle of human
stimulation from the nervous system. However, in a setting and mouse) and ultrarapid K⫹ currents (IKur; atria of hu-
such as exercise, the autonomic nervous system plays a key man) (49, 178). In mice, Ito plays a more prominent role for
role in allowing the heart to respond to meet the increased repolarization than in humans, reflected by the different
demands of the body (see sect. IID1). The heartbeat origi- shape on the AP (229, 300) (FIGURE 5, A AND B). The hu-
nates from the sinoatrial node (SA) within the right atrium man AP has a long plateau phase (phase 2) followed by a
of the heart which acts as a pacemaker, producing regular rapid repolarization. In contrast, in mice there is an earlier
electrical impulses. The SA node contains autorhythmic rapid repolarization via Ito, leading to a dramatic drop in
cells which spontaneously “fire” (depolarize). The electrical the amplitude of the AP by ~50 – 60% (FIGURE 5B). This is
impulse (action potential, AP) is propagated through the followed by slowing of the mouse AP, represented by a
atria to the atrioventricular node (AV) and then passes via plateau phase shoulder in which another transient outward
the conducting Purkinje fibers to spread through the ventri- current (IKslow) plays a role for repolarization (229). During
cle. Contractile cardiac myocytes are excited by adjoining the plateau phase (phase 2 on human AP), there is a small
cardiac pacemaker cells leading to a coordinated contrac- but constant inward L-type calcium current (ICaL; opening
tion of the atria and ventricle. The electrocardiogram of L-type Ca2⫹ channels). This plays an important role for
(ECG) provides a measure of the electrical activity of the
E-C coupling. E-C coupling refers to the excitation stimuli
heart which represents an integrated readout of APs in dif-
(electrical AP) being linked/coupled to cardiac muscle con-
ferent regions of the heart (FIGURE 5A). On the ECG, the
traction (FIGURE 5C). Calcium is the key mediator that cou-
P-wave represents atrial depolarization followed by atrial
ples electrical excitation to contraction by cycling in and out
contraction, the QRS complex reflects ventricular depolar-
of the cardiac myocyte during each AP (336). The initial
ization followed by ventricular contraction, and the T-wave
entry of Ca2⫹ into cardiac myocytes through L-type Ca2⫹
represents ventricular repolarization followed by ventricu-
channels which occurs during phase 2 of the action poten-
lar relaxation. At the cellular level, the AP represents a
tial (plateau phase) is insufficient to trigger contraction of
sequence of depolarizing inward currents (Na⫹ and Ca2⫹)
myofibrils. However, the ICaL increases the probability of
and repolarizing outward currents (K⫹) moving across the
the ryanodine receptors (RyR) opening (type 2 in the heart,
membrane via channels/pumps of cardiac myocytes (82,
137, 297, 299, 336). In humans, the AP of cardiac myocytes RyR2) and leads to the release of Ca2⫹ from the SR into the
is composed of five phases (phase 0, depolarization; phase cytoplasm (a process known as calcium-induced calcium
1, brief early repolarization; phase 2, plateau phase of main- release). The elevation in free cytoplasmic Ca2⫹ levels re-
tained depolarization; phase 3, repolarization; phase 4, sults in Ca2⫹ binding to troponin C in the myofilaments,
resting) (82) (FIGURE 5A). Ion channels in humans and mice leading to cardiac contraction and mechanical systole (198,
are generally highly conserved. However, there are differ- 336). In diastole, cardiac relaxation occurs in response to
ences in the shape/phases of the AP and surface ECG from removal of intracellular free and troponin C-bound Ca2⫹
humans and mice due to the presence of different channels, by SERCA2a (removes the majority of Ca2⫹) together with
and some differences in the contribution and/or timing of the Na⫹Ca2⫹ exchanger (NCX), the mitochondrial calcium
the opening/closing of channels (300, 360) (FIGURE 5, A uniporter (MCU), and the sarcolemmal Ca2⫹-ATPase (FIG-
2⫹
AND B). This is not surprising, considering the heart rate of URE 5C). As SERCA2a controls the uptake of Ca into the
mice is ~10 times higher than humans, the mouse requiring SR, it plays important roles for cardiac relaxation, Ca2⫹
shorter APs. The subsequent text summarizes the role of loading of the SR, and consequently the amount of Ca2⫹
different currents contributing to the different phases of the available for release during cardiac myocyte contraction
human AP and how these can change with exercise (see sect. (196). SERCA2a activity is regulated by phospholamban
IID1). However, since many of the mechanistic studies have (PLB/PLN). When PLB is unphosphorylated it binds to
been obtained from genetic mouse models, the key differ- SERCA2a and inhibits its activity. However, when PLB is
ence between the contribution of currents in human and phosphorylated, it is removed from SERCA2a and no longer
mouse myocytes have also been highlighted. Despite differ- inhibits its activity, and Ca2⫹ uptake into the SR increases
ences in the AP and ECG of mice and humans, mouse mod- (198). PLB is largely phosphorylated by cAMP-dependent
els have led to a better understanding of electrical remodel- protein kinase A (PKA) and Ca2⫹/calmodulin-dependent ki-
ing in the heart and the cause of arrhythmias in settings of nase II (CaMKII) (196) (FIGURE 5C).
cardiac pathology (180).
The repolarization of the human AP after phase 2 occurs in
Cardiac APs are initiated by a voltage-gated inward current response to delayed outwardly rectifying K⫹ currents (slow,
that causes depolarization of the cell membrane. In the ma- IKs and rapid, IKr; phase 3) (82). The difference in shape of

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 431


BERNARDO ET AL.

PHASE 1
A HUMAN B MOUSE
Ito PHASE 2 R
SAN
IKs

PHASE 0
Atrium ECG
SAN AVN
ICaL IKr PHASE 3 P Q ST

AVN Purkinje Ito


INa IK1
ICaL
IKslow
Ventricle IK1 Ca2+ Na+ IK1
R
PHASE 4 INa
PHASE 4
ECG NCX 3Na+-2K+
PQS T
ATPase
+ +
Na K
C
EXERCISE
ACTION
POTENTIAL
↓ Parasympathetic ↑ Sympathetic
Ito IKs IKr IKur IKslow IK1 IKACh 3Na+-2K+
↓ Ach ↑ NE
ATPase
INa K+ K+ K+ K+ K+
Na+
a Kv4.2 KvLQT1 Kir3.1 M2 β1 AR
Kv4.3 Kv1.5 Kir2.1
Nav1.5 ERG Kir3.4

ATPase K+
Na+
Gαi Gαs
Sarcoplasmic ICaL
reticulum cAMP Ca2+
2+ PKA P Cav1.2
L-type Ca
channel b RyR2 c P
PLB CaMKII h ∆ATP K+
Ca2+
ICaL ATP
IKATP
PLB MCU Kir6.2
Cav1.2 Ca2+ Ca2+ 2+
SERCA2a Ca mPTP
Ca2+ Ca2+
f Ca2+
Ca2+ ATPase
Ca2+
DIASTOLE g
d Ca2+ Mitochondria Ca2+
Ca2+
SYSTOLE NCX

T-tubule Na+ T-tubule


e Myofilaments

FIGURE 5. The action potential (AP) and effects of exercise on E-C coupling. The ventricular AP from human
(A) and mouse (B). The AP represents an interplay of inward and outward currents. INa, sodium current; ICaL,
L-type Ca2⫹ current; Ito, transient outward K⫹ current; IKr, rapid component of the delay rectifier K⫹ current;
IKs, slow component of the delayed rectifier K⫹ current; IK1, inward rectifier K⫹ current. C: currents which
contribute to the AP in A are presented on the cell membrane together with key channel proteins. The AP
depolarizes the cell membrane (a) leading to activation of L-type Ca2⫹ channels in the t-tubule and Ca2⫹ entry
(b). Ca2⫹ then binds to ryanodine receptors (RyR2) on the sarcoplasmic reticulum (SR; c). This is followed by
large amounts of Ca2⫹ from the SR (referred to as Ca2⫹-induced Ca2⫹ release; d) which leads to myofilament
and cardiac contraction (e). For the heart to relax, cytoplasmic Ca2⫹ levels must be returned to diastolic levels,
allowing Ca2⫹ to dissociate from myofilaments. This occurs due to inactivation of extracellular Ca2⫹ entering
through the L-type Ca2⫹ channels, and Ca2⫹ being removed from the cytoplasm via SERCA2a (f), the
Na⫹/Ca2⫹ exchanger (NCX: forward mode), the plasma membrane Ca2⫹-ATPase (g), and the mitochondrial
calcium uniporter (MCU; h). In response to exercise there is 1) increased sympathetic activity, release of
catecholamines, and increased activation of the ␤1-AR-cAMP-PKA pathway which increases cardiac contrac-
tility via phosphorylation of the L-type Ca2⫹ channel, RyR2, and PLB; 2) reduced parasympathetic activity acting
via the M2 receptor; and 3) regulation of the KATP channel.

the mouse AP (i.e., plateau phase shoulder) is because IKs tential. This occurs in response to the extra Na⫹ and Ca2⫹
and IKr are not prominent repolarizing K⫹ currents in the (which entered the cell during the AP) being removed from
adult mouse heart (229, 300, 360) (FIGURE 5, A AND B). the cell and K⫹ entering the cell via the sodium/potassium
Despite the name, non-voltage-gated “inwardly” rectifying pump (Na⫹-K⫹-ATPase pump) and NCX (82, 297, 299)
K⫹ (Kir) currents also contribute to repolarization in hu- (FIGURE 5, A AND B). There are other Kir channels (i.e., in
man and mouse via outward K⫹ currents. IK1 contributes addition to IK1) that are important for normal heart func-
mainly during phase 3 and 4 (299). tion but are not considered to play a major role in AP
repolarization under normal physiological settings. How-
Return to the resting state (phase 4) requires restoration of ever, these currents are known to play an important role in
ion concentrations back to balanced states pre-action po- conditions such as exercise and include 1) acetylcholine-

432 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

activated K⫹ current [IKACh greatest abundance in atria and inhibition of SERCA2a), RyR2, troponin I, and myosin
pacemaker cells but also considered to play a role in the binding proteins (36, 137). The phosphorylation of L-type
ventricle (138, 275)] and 2) ATP-sensitive K⫹ current Ca2⫹ channels leads to an increase in Ca2⫹ entry into the
(IKATP). In contrast to the voltage-gated ion channels that heart which increases Ca2⫹ release by the SR through
are regulated by changes in the electrical membrane poten- RyR2. Increased Ca2⫹, together with enhanced SR Ca2⫹
tial (e.g., fast Na⫹ channels, L-type Ca2⫹ channel), IKACh cycling, results in increased contraction.
and IKATP channels are ligand-gated channels (138, 299).
There is also evidence that CaMKII is activated in response
1. Exercise-induced changes of the AP and E-C to ␤-AR stimulation. Increased intracellular Ca2⫹ in re-
coupling sponse of PKA-induced phosphorylation of L-type Ca2⫹
channels activates CaMKII (FIGURE 5C). CaMKII can also
Exercise has been shown to regulate the activity and/or phosphorylate the L-type Ca2⫹ channels, RyR2, and PLB
density of numerous channels, pumps, and processes asso- and thus, together with PKA, increase entry of Ca2⫹ into the
ciated with the cardiac AP and E-C coupling (summarized SR (78, 141, 441, 451). Treadmill aerobic interval training
in TABLE 2). For the heart to meet the energetic demands of in mice for 6 wk was associated with increased cardiac
exercise, there is an increase in heart rate and cardiac con- myocyte contractility, larger calcium amplitudes, faster re-
tractility. This occurs in response to the autonomic nervous laxation, phosphorylation of PLB, and increased activation
system and hormones, which increase heart rate by acting at of CaMKII␦ (predominant cardiac isoform). Many of these
the SA node, and by enhancing cardiac myocyte contractil- effects were inhibited by a CaMKII inhibitor [autocamtide-
ity by altering ion currents, pumps, and regulators/compo- 2-related inhibitory peptide (AIP), although this inhibitor
nents of E-C coupling (137). Under resting conditions, the
may have some off target actions] (196, 451). In contrast,
parasympathetic nervous system predominates and releases
excessive CaMKII is recognized to contribute to cardiac
acetylcholine which controls the human heart rate at
pathology, heart failure, and arrhythmia (78, 285).
~60 –70 beats/min. During exercise the sympathetic ner-
vous system is activated and NE is released (mainly from the
B) SERCA2A. Exercise such as chronic aerobic running has
postganglionic sympathetic neurons that innervate the
been shown to increase cardiac myocyte contractility by
heart but also the adrenal medulla). NE activates ␤1-ARs
increasing SERCA2a Ca2⫹ uptake/activity or protein in
which are expressed throughout the heart (atrial and ven-
healthy animals as well as rodent models of cardiac disease
tricular myocytes, SA node, AV node). ␤-Adrenergic signal-
(63, 149, 177, 196, 424, 446). Ca2⫹ uptake by SERCA2a
ing plays an important role in coupling heart function with
can be enhanced by the direct regulation of SERCA2a tran-
changing physiological demands. Short-term or acute acti-
vation of ␤-adrenergic signaling in the heart (as occurs with scription and activity, or via the modification of regulatory
exercise) increases heart rate via effects on the SA node, and processes/factors, e.g., PLB phosphorylation or other pro-
enhances systolic and diastolic function (contractility and tein posttranslational modifications (O-GlyNAcylation,
relaxation) by acutely increasing Ca2⫹. In contrast, long- SUMOylation) (149). Furthermore, a number of exercise-
term chronic activation of ␤-adrenergic signaling as occurs induced factors/triggers/signaling pathways including the
in disease settings results in Ca2⫹ overload of the SR, and ␤-adrenergic pathway, thyroid hormone levels (T3/T4), mi-
this leads to myocyte death (apoptosis and necrosis), car- tochondrial biogenesis, miRNAs, and PI3K-Akt signaling
diac dysfunction, and heart failure (441). have been reported to regulate SERCA2a transcription
and/or protein (149).
The mechanisms responsi-
A) ␤-ADRENERGIC-RELATED MECHANISMS.
ble for the exercise-induced shortening of the AP and in- C) MITOCHONDRIA.Mitochondria in cardiac myocytes play a
creased heart rate are complex and not entirely understood; role in maintaining a physiological Ca2⫹ concentration in
some mechanisms may also differ between species (110). the cytoplasm by acting as a buffer for elevated Ca2⫹ that
However, there is evidence to suggest that ␤-adrenergic can have an adverse effect on myocyte function. Cellular
stimulation increases heart rate via cAMP binding to ion Ca2⫹ is largely stored in the SR, and mitochondria located
channels responsible for the funny current [If; hyperpolar- near Ca2⫹-release sites on the SR capture released Ca2⫹.
ization-activated cyclic nucleotide-gated (HCN) channels The MCU is considered an important regulator of how
that conduct an inward pacemaker current] (137, 141, much Ca2⫹ enters the mitochondria (FIGURE 5C). At resting
451). Ca2⫹ levels the activity of the MCU in the heart is low.
However, with increased Ca2⫹ cycling due to inotropic
In both humans and rodents, cardiac contractility is in- drive or augmented work load, MCU activity is enhanced to
creased by ␤-ARs coupling to the stimulatory G protein allow faster Ca2⫹ loading of the mitochondria. In an induc-
(Gs)-cAMP-PKA pathway, and exercise stimulates ␤-AR ible cardiac-specific KO mouse model of MCU, basal mito-
signaling (185, 192, 283, 284) (FIGURE 5C). PKA phosphor- chondrial Ca2⫹ was not affected and mice showed no ob-
ylates many proteins to mediate an increased rate of E-C vious phenotype. However, the KO model displayed intol-
coupling including L-type Ca2⫹ channels, PLB (relieving erance to acute treadmill exercise, suggesting that the MCU

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 433


BERNARDO ET AL.

Table 2. Exercise-mediated regulation of channels/pumps/processes associated with the cardiac AP and E-C coupling
Current/Pump/Process Protein Gene Key Features Impact of Exercise/caPI3K

INa: sodium current Nav1.5 SCN5A Phase 0 of human AP Swim training in mice 1Scn5a (455)†
Voltage activated caPI3K: 1Scn5a (455)†
Ventricle and atria: human and
mouse (299)
Ito (Ito,f and Ito,s) Phase 1 of human AP Swim training in mice 1current amplitude,
1Kv4.2, 1KCND2, 1KCND3 (455)
Ito,f: transient outward current, Kv4.2 KCND2 Voltage activated caPI3K: 1current amplitude, 1Kv4.2,
fast 1KCND2, 1KCND3 (455)
Kv4.3 KCND3 Ventricle and atria: human and Transgenic model of DCM: 2current
mouse (299) amplitude and density, 2KCND2,
2KCND3. Most defects attenuated by
swim training or caPI3K (456)
Ito,s: transient outward current, Kv1.4 KCNA4 Voltage activated TAC model: caPI3K prevented a 2Ito,f
slow current density and 1 current amplitude
(456)‡
Ventricle and atria: human and Ito,s not specifically assessed
mouse (299)
IKur: delayed rectifier, ultra Kv1.5 KCNA5 Phase 1 of human AP
rapid (human)
Voltage activated
Atria: human (299)
IKslow (IKslow1, IKslow2) Same protein and gene as IKur Swim training in mice 1current amplitude
in humans. of IKslow, 1Kv2.1, 1Kcnb1 (455)
IKslow1: rapidly activating, slowly Kv1.5 KCNA5 Ventricle and atria: mouse caPI3K: 1current amplitude and density
inactivating delayed rectifier (299) of IKslow, 1Kv2.1, 1Kcnb1 (455)
current (mouse)
IKslow2: delayed rectifier with Kv2.1 KCNB1 Ventricle and atria: mouse Transgenic model of DCM: 2current
slower inactivation kinetics (299) amplitude and density of IKslow, 2Kcna5
than IKslow1 (mouse) 2Kcnb1. Defects attenuated by swim
training or caPI3K (456)
TAC model: caPI3K prevented a 2IKslow
current density and 1current amplitude
(456)‡
ICaL: calcium current, L-type Cav1.2 CACNA1C Phase 2 of human AP Swim training in mice 1Cacna1c (455)†
Voltage activated caPI3K: 1current amplitude and density
of ICa, 1Cacna1c (455)
Ventricle and atria: human and
mouse (299)
IKr: delayed rectifier, rapid HERG/ KCNH2 Phase 3 of human AP Swim training in mice 1mERG (455)†
(smaller role in mouse) mERG
Voltage activated
Ventricle and atria: human and
mouse (299)
IKs: delayed rectifier, slow (role KvLQT1 KCNQ1 Phase 3 of human AP Swim training in mice 1Kcnq1 (455)†
in mice unclear but gene
present)
Voltage activated caPI3K: 1Kcnq1 (455)†
Ventricle and atria: human.
Not reported in mice but
gene present (299, 455)
IK1: inward rectifier Kir2.1 KCNJ2 Phase 3 and 4 of human AP Swim training in mice 1current amplitude
and density, 1Kir2.2, 1Kcnj2,
1Kcnj12 (455)
Kir2.2 KCNJ12 Voltage activated caPI3K: 1current amplitude and density,
1Kir2.2, 1Kcnj2, 1Kcnj12 (455)
Continued

434 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

Table 2.—Continued
Current/Pump/Process Protein Gene Key Features Impact of Exercise/caPI3K

Ventricle and atria: human and Transgenic model of DCM: no change in


mouse (299) current amplitude but 2density,
2Kcnj2, 2Kcnj12. Exercise and caPI3K
1current amplitude and density. caPI3K
1Kcnj2 and Kcnj12 (not assessed with
exercise) (456)
TAC model: caPI3K prevented a 2IK1
current density and 1current amplitude
(456)‡
Iss: delayed rectifier TASK-1 KCNK3 Ventricle and atria: human and Swim training in mice 1current amplitude
mouse (299) and density, 1TASK1, 1Knk3 (455)
caPI3K: 1current amplitude and density,
1TASK1, 1Kcnk3 (455)
Transgenic model of DCM: no change in
current amplitude or density, 2Kcnk3.
Exercise and caPI3K 1current
amplitude and density. caPI3K 1Kcnk3
(not assessed with exercise) (456)
Currents below do not appear
to contribute directly to the
normal AP based on Ref.
299
IKAch: acetylcholine-regulated Kir3.1 KCNJ3 Activated by acetylcholine Knockout mice with loss of function of
K⫹ current (contribution in Kir3.4 show a delayed recovery of
mouse unclear) resting heart rate after exercise (275)
Kir3.4 KCNJ5 Not assessed in Refs. 455, 456
IKATP: ADP activated K⫹ Kir6.2 KCNJ11 Activated by 1[ADP]/[ATP] Short-term treadmill exercise increased
channel the expression of KATP in the heart
(469)
Not assessed in Refs. 455, 456
Exercise-induced shortening of AP
attenuated in Kir6.2 KO (469)
Depressed cardiac function and
pathological heart growth in Kir6.2 KO
mice subjected to 28 days of moderate
swim training (183)
NCX Treadmill for 11 wk in rats (associated
with 1HW/BW) 1affinity of NCX for
Ca2⫹ (412)
SERCA2a and PLB Aerobic interval treadmill training for 6 wk
in mice was associated with 1SERCA2a
protein and function, i.e., 1Ca2⫹ SR
uptake (195). The same protocol was
associated with 1cardiac myocyte
contractility, larger calcium amplitudes,
faster relaxation, phosphorylation of
PLB, and increased activation of
CaMKII␦ (196). Some of these changes
were prevented with a CaMKII inhibitor
(196) (proposed mechanism via CaMKII
phosphorylating PLB).

*Kv4.3 predominant in larger mammals. †Ionic current not assessed in study. ‡Exercise not assessed. For “protein,” the primary ␣-subunit of
the major ion channel is included. Accessory/␤-subunits have not been listed but can be found in References 299, 300. AP, action potential;
BW, body weight; DCM, dilated cardiomyopathy; E-C, excitation-contraction; HW, heart weight; KO, knockout; TAC, transverse aortic constric-
tion. Reference numbers are given in parentheses.

plays a critical role for the acute regulation of Ca2⫹ into the The KATP channel is distributed in all regions of the heart
mitochondria in response to exercise (217). and is considered to link cellular metabolism with mem-
brane excitability. The activity of KATP is regulated by ad-
I) KATP channels. KATP
D) LIGAND-GATED CHANNELS (KATP AND KACh). enine nucleotides, typically being activated by falling ATP
channels are considered to play a prominent role in re- and rising ADP levels (414). Stress-induced opening of the
sponse to stress or metabolic challenges including exercise. channel has been considered protective because the opening

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 435


BERNARDO ET AL.

of KATP leads to quicker repolarization, shortening of the physiological hypertrophy, chronic changes are required.
AP, and reduced Ca2⫹ influx. This results in the conserva- To maintain cardiac excitability in a setting of exercise-
tion of cellular energy stores and prevents calcium overload induced cardiac hypertrophy, it is important for the density
(184, 414). The KATP channel in cardiac myocytes is en- of ion channels on cardiac myocytes to increase in parallel
coded largely by Kir6.2 (FIGURE 5C). Kir6.2 KO mice have with the increase in cardiac myocyte size. It was shown that
been subjected to treadmill stress exercise testing and mod- cardiac electrical function was maintained in mice with ex-
erate aerobic swimming exercise (183, 468). Under basal ercise-induced hypertrophy due to chronic swim training
conditions, cardiac function was comparable in Kir6.2 KO for 4 wk or in caPI3K mice with physiological hypertrophy.
and wild-type mice. However, Kir6.2 KO mice displayed a ECG traces and AP waveforms were comparable in exercise
reduced capacity to perform a high-intensity treadmill test and caPI3K mice compared with untrained mice and non-
(lower work load and duration), and disruption of cardiac transgenic mice (recordings made under resting conditions
performance was implicated (468). In response to a 28-day not during exercise). The comparable ECG traces and APs
modest swimming protocol, global Kir6.2 KO mice dis- were attributed to an increase in repolarizing outward K⫹
played hypertrophy that was associated with depressed sys- current amplitudes together with an increase in inward
tolic function and myocyte damage (physiological hyper- Ca2⫹ currents (455). This is in contrast to the prolongation
trophy not observed in wild-type mice with this exercise of the AP which is a common feature of pathological car-
protocol). While these results are confounded by the dele- diac hypertrophy and heart failure and can make the heart
tion of Kir6.2 within other organs (e.g., skeletal muscle), it susceptible to arrhythmia (163). Exercise training in mice
was reported that KO mice swam to a similar degree to has also been shown to regulate the protein and gene ex-
wild-type mice (confirmed by similar enhancement in skel- pression of a number of channels responsible for currents
etal muscle aerobic capacity and lower resting heart rates) highlighted on FIGURE 5 (455) (TABLE 2).
(183). Consistent with the KATP channel playing a key role
in the heart in response to exercise, cardiac-specific trans- In cardiac disease rodent models, exercise or increased car-
genic mice with nonfunctional KATP channels displayed an diac PI3K activity has been shown to have favorable out-
attenuated response to short-term exercise. Treadmill exer- comes on electrical activity/AP and E-C coupling. Treadmill
cise at moderate speed for 5 days in wild-type mice was running in rats which began 4 wk post-MI resulted in better
associated with increased cardiac expression of Kir6.2 and intracellular Ca2⫹ handling and sensitivity within cardiac
shortening of the AP in response to an exercise-induced myocytes compared with nontrained MI rats (446). Cardiac
increase in heart rate. These changes were blunted in the disease mouse models including pressure overload-induced
transgenic mice (469). Thus KATP is considered to be re- pathological hypertrophy (transverse aortic constriction,
quired for the beneficial cardiac effects of exercise (184). TAC) and dilated cardiomyopathy with heart failure (trans-
genic model) are characterized by QT prolongation, de-
II) KACh channels. After exercise has ceased, it is important creased K⫹ current densities, and impaired repolarization.
for the heart rate to return to normal levels within a short Transgenic expression of caPI3K was able to attenuate or
time frame. A delay in heart rate recovery after exercise has prevent ECG and AP abnormalities in these disease models,
been shown to be an independent predictor of mortality in and protection was associated with increased repolarizing
healthy individuals as well as those with underlying cardiac K⫹ current amplitudes (e.g., IK1, Ito) and increased expres-
disease (74, 75, 179, 305). Release of acetylcholine from sion of K⫹ channel subunits (e.g., Kir2.1, Kir2.2, Kv4.2,
cardiac vagal nerves is responsible for the heart rate recov- Kv4.3) (456). Swim training for 4 wk also upregulated re-
ery after exercise. Acetylcholine binds to muscarinic recep- polarizing K⫹ currents in the model of dilated cardiomyop-
tors in the heart (M2) which are abundant in the atria, SA athy and heart failure (456) (TABLE 2).
node, and AV node. Binding to M2 receptors activates in-
hibitory G proteins (Gi) that 1) inhibit PKA activity and As de-
F) NO-RELATED MECHANISMS ASSOCIATED WITH E-C COUPLING.
downstream effects on contractility and 2) promote the scribed in section IIC1, NO is increased in response to ex-
opening of KACh channels (increases in IKACh) (138, 275) ercise and has been shown to play a key role for exercise-
(FIGURE 5C). KO mice with loss of function of Kir3.4, which induced cardiac protection via vascular adaptations (58,
encodes an integral subunit of the IKACh channel, show a 332). NO derived from eNOS and nNOS within cardiac
delayed recovery of resting heart rate after an acute 5 min myocytes can also regulate numerous steps of E-C coupling
swim or treadmill test (275). (259) (FIGURE 4B). NO derived from eNOS within the
plasma membrane of cardiac myocytes (e.g., within cave-
E) LONG-TERM CHANGES TO CARDIAC EXCITABILITY AND E-C COUPLING WITH olae activated by ␤3-AR or stretch) can potentially regulate
PHYSIOLOGICAL CARDIAC HYPERTROPHY AND PROTECTION FROM DIS- ROS production, myofilament sensitivity, PLB, and RyR2
EASE. During exercise, there are acute changes to the conduc- (251, 372) (FIGURE 4B). Exercise-induced ␤1-AR signaling
tion system and E-C coupling to increase heart rate and results in a positive inotropic effect via cAMP-PKA signal-
contractility. However, with regular long-term exercise and ing phosphorylating the L-type Ca2⫹ channel and enhanc-
the development of exercise-induced adaptions including ing SERCA2a (via the phosphorylation of PLB). It has been

436 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

suggested that ␤3-AR-induced NO production may blunt mitochondrial fusion-related proteins (e.g., Mfn2 and
␤-adrenergic inotropic responses in disease settings (58, 59, OPA1) and fission-related proteins (DRP1) has been con-
372). sidered a target for cardiac therapies (100, 368). Studies
have suggested that inhibition of excessive/pathological mi-
NO derived from nNOS in the SR (FIGURE 4B) may also tochondrial fission is beneficial in the heart (100), and ex-
provide benefit in a setting of exercise by 1) inhibiting Ca2⫹ ercise has been associated with changes in fusion- and fis-
entering myocytes via the L-type Ca2⫹ channels, 2) stimu- sion-related proteins. For instance, aerobic interval training
lating reuptake of Ca2⫹ into the SR by phosphorylating in rats (treadmill for 8 wk) provided protection against
PLB, 3) modulating ␤-adrenergic inotropic responses, and MI-induced impairment of mitochondrial respiratory func-
4) reducing oxidative stress (372). A favorable effect of tion, and exercise prevented the MI-induced decrease in
exercise on the balance of ROS and NO in the heart (ni- mitochondrial fusion (Mfn2 and OPA1) and increased fis-
troso-redox balance) is thought to contribute to exercise- sion (DRP1) (175).
induced cardiac protection; attributed to NO “buffering”
ROS (353). nNOS is elevated in the heart in response to Mitochondria play key roles for energy production (ATP),
exercise and is reported to be the major NOS responsible redox control (producing and scavenging ROS), calcium
for a positive shift in the nitroso-redox balance in ventric- homeostasis, contractility, ion gradients, nuclear gene ex-
ular myocytes. By subjecting NOS1 global KO mice (gene pression, and cell fate (survival/cell death). In this section
encoding nNOS) to an 8-wk aerobic interval treadmill we have focused on the role of mitochondria for energy
training program, it was shown that nNOS is critical for production and redox status. Calcium homeostasis was de-
exercise-induced physiological cardiac hypertrophy and ex- scribed in section IID and cell fate/survival in section IIF.
ercise-induced antioxidant properties (353, 354). In cardiac
myocytes, exercise was associated with increased nNOS 1. Mitochondrial energy metabolism
protein, enhanced myocyte contraction (Ca2⫹ transients),
and increased SR Ca2⫹ cycling (load and release). These The heart has high energy demands having to continuously
adaptations did not occur in myocytes from NOS1 KO contract to provide oxygen and nutrients to the body. Mi-
mice. While it is noted that global NOS1 KO mice may have tochondria generate ATP through oxidative phosphoryla-
reduced exercise capacity due to deletion within skeletal tion, a process which generates ⬎90 –95% of the ATP in the
muscle, the investigators consider this unlikely to have con- heart (144, 240). With limited ATP reserves (nearly com-
tributed to the phenotype because training intensity was plete turnover of the ATP stores every 10 s), the heart relies
matched to similar levels in control and KO mice (354). on a continuous energy supply that can be generated from a
number of substrates including carbohydrates (glucose, lac-
tate, and pyruvate) and noncarbohydrates (fatty acids,
E. Mitochondrial Adaptations: Energy amino acids, and ketones) (38, 171, 240). In the normal
Production and Redox Status healthy heart under resting conditions, oxygen and fuel sup-
ply is relatively abundant. Fatty acids are the preferred fuel
Overview: the volume density for mitochondria in the hu- source (60 – 80%), an energy rich substrate. Glucose and
man and mouse heart has been estimated to represent 23 lactate represent the other major fuel sources under resting
and 32%, respectively (368). Within cardiac myocytes, mi- conditions (38, 171) (FIGURE 6). Upon entry of free fatty
tochondria are located just below the plasma membrane acids into the cell, the majority are transported into the
where they provide ATP to membrane ion pumps, and be- mitochondria and undergo fatty acid oxidation (␤-oxida-
tween myofibrils where they deliver energy to support con- tion). Glucose enters the cell via glucose transporters
traction (12, 144). Mitochondria size and number are reg- (GLUT) and undergoes glycolysis. Glycolysis converts glu-
ulated by the process of mitochondria fusing (fusion) and cose to pyruvate under aerobic conditions (lactate under
dividing (fission). A balance of mitochondrial fission/fusion anaerobic conditions). Pyruvate is transported into the mi-
is critical for mitochondrial functions, and loss of this bal- tochondria and undergoes oxidation (i.e., glucose oxida-
ance can lead to cardiac disease. Thus the regulation of tion). Both pathways (free fatty acids and glucose) con-

FIGURE 6. Effects of exercise on energy metabolism. During exercise, circulating substrate levels (fatty acids, glucose, and lactate) are
regulated dependent on exercise type (e.g., increased fatty acids with aerobic exercise, increased lactate with anaerobic exercise). Substrates
enter cardiac myocytes via active and passive transport (e.g., fatty acids via CD36, glucose via GLUT4). Fatty acids enter the mitochondria and
are converted to acetyl-CoA via fatty acid oxidation (␤-oxidation). Glucose and lactate are converted to pyruvate and undergo glucose oxidation
and conversion to acetyl-CoA. Acetyl-CoA enters the TCA cycle, and reducing equivalents deliver electrons to the electron transport chain,
leading to ATP generation via oxidative phosphorylation. AMPK, PGC-1␣, and PI3K are increased in the heart in response to exercise and can
regulate components of energy metabolism by affecting fatty acid oxidation and gene expression. PGC-1␣ is an important transcription
coactivator; when in combination with other transcription factors (TF), it regulates the expression of genes related to fatty acid oxidation and
mitochondrial biogenesis.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 437


BERNARDO ET AL.

Fatty acids Lactate Glucose

Circulation
CD36 GLUT4

Cardiomyocyte
AMPK AMPK
<5% ATP

EXERCISE Fatty acids


Glycolysis DISEASE
Inadequate
PGC-1α glucose oxidation
Lactate Pyruvate
+
PI3K + +
AMPK +
↑ Proton production

Mitochondria

Fatty acid Glucose


oxidation Acetyl- oxidation
CoA

ATP
e-
TCA e- e-
cycle
>95% ATP
AMPK e-

EXERCISE
Electron transport chain Cardiac contraction

PGC-1α
Oxidative phosphorylation

Nucleus PGC-1α
Fatty acid oxidation Glycolytic
PPARα genes PPARγ genes

PGC-1α
Fatty acid oxidation genes
TF Mitochondrial biogenesis
TCA

EXERCISE DISEASED HEART

↔ / ↑ Fatty acid oxidation ↔ / ↓ Fatty acid oxidation


↔ / ↑ glucose oxidation: glycolysis ↓ glucose oxidation: glycolysis
↑ ATP ↓ ATP
↑ proton production
↑ efficiency ↓ efficiency

438 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

verge to produce acetyl-CoA which undergoes oxidation protects the heart in settings of cardiac stress. One expla-
in the TCA cycle and the production of ATP (via reducing nation for the improved outcome in cardiac stress settings is
equivalents, delivery of electrons to the electron trans- the lower net proton production of glucose oxidation com-
port chain, and generation of a proton gradient across pared with glucose fermentation (glycolysis and reoxida-
the inner mitochondrial membrane) (171, 240, 395). The tion of NADH produced by glycolysis). Lower proton pro-
balance of fatty acid oxidation, glucose oxidation, and duction is associated with better recovery of intracellular
glycolysis has an impact on cardiac efficiency and energy pH which would be expected to limit calcium overload
production (FIGURE 6). (H⫹/Na⫹ and Na⫹/Ca2⫹ exchange) and reduce the ener-
getic cost of maintaining ion homeostasis (53). Higher fatty
During exercise, energy expenditure of the heart increases acid oxidation in the exercise-trained heart may also help
severalfold compared with resting conditions. This is asso- limit the toxic effects of high free fatty acids on the cytosol
ciated with a rise in oxygen consumption and an increased and mitochondria (53). In another study, swim exercise
rate of mitochondrial ATP production that matches the rate training for 3 wk in mice before acute MI was associated
of ATP breakdown by the working muscles (12). It is gen- with reduced infarct size, and exercise prevented the MI-in-
erally accepted that the metabolic phenotype of the exer- duced decrease in fatty acid metabolism-related genes [e.g.,
cise-trained heart and diseased heart are different (FIGURE those encoding carnitine palmitoyltransferase 1 (CPT-1), me-
6). While varying views exist regarding the key metabolic dium chain acyl-CoA dehydrogenase (MCAD), peroxisome
changes that occur in each state, which may differ depend- proliferator-activated receptor ␣ and ␥ (PPAR-␣ and -␥)]
ing on disease or exercise type and/or duration, some com- (405).
mon findings are emerging. In the diseased heart, one long-
standing view has been that the heart becomes energetically Substrate availability is one key factor which “dictates” the
starved due to inadequate rates of energy substrate metab- choice of substrate used by the heart to produce energy, but
olism and ATP production. A more recent view is that the other mechanisms include the activation of kinases/signal-
chronically stressed heart uses energy inefficiently due to ing cascades [e.g., PI3K, AMPK, and proliferator-activated
mismatched glucose metabolism (i.e., where glycolysis is receptor ␥ co-activator 1␣ (PGC-1␣)] and transcriptional
uncoupled from glucose oxidation). Mismatched glucose regulation of genes involved in metabolism. The PPAR fam-
metabolism causes accelerated proton production that can ily of transcription factors (PPAR-␣ and PPAR-␥) regulate
lead to acidosis and impaired myocyte contractility (171, the expression of genes encoding enzymes of fatty acid and
258) (FIGURE 6). Lopaschuk and colleagues (258) demon- glucose metabolism. Models of chronic exercise training in
strated that in the failing mouse heart due to MI, the hearts rodents (e.g., swim training or treadmill) have been shown
were inefficient in energy utilization for mechanical func- to increase AMPK, PGC-1␣, and PI3K in the heart and
tion, i.e., mismatch in glucose metabolism due to alterations promote fatty acid oxidation, ATP generation, and mito-
in glucose oxidation not matching the changes in glycolysis. chondrial biogenesis (2) (FIGURE 6). AMPK is considered
As in the diseased heart, the effect of acute and endurance beneficial by increasing fatty acid uptake and oxidation,
exercise on cardiac metabolism requires further study accelerating glucose uptake via translocation of GLUT4 to
(427). However, in contrast to a disease setting, there is the plasma membrane, and inhibiting energy-consuming
evidence to suggest that exercise is associated with en- pathways (e.g., protein synthesis) (101). The critical role of
hanced fatty acid and glucose oxidation in the heart or no PI3K for mitochondrial adaptations was demonstrated us-
significant change (FIGURE 6) (427). In healthy trained male ing mice with decreased cardiac PI3K activity (dnPI3K
subjects, myocardial oxidation of exogenous glucose in- mice). In response to chronic swim training, dnPI3K mice
creased during moderate-intensity exercise (cycle ergometer did not display an exercise-induced increase in mitochon-
exercise at 40% of the subject’s maximal O2 uptake 25–50 drial ATP production and fatty acid oxidation (310). IRS1
min) (133). The increased use of glucose with exercise has and IRS2 also play critical roles for chronic swim exercise-
been considered a more efficient energy source than fatty induced mitochondrial adaptations (e.g., measures of mito-
acids (producing more ATP per O2 molecules consumed) chondrial fatty acid metabolism) (310, 347). In contrast,
(121, 427). There is also a high use of fatty acids during Akt and PDK1 do not appear to play an essential role for
exercise as a result of elevated circulating triglycerides and chronic swim exercise-induced mitochondrial adaptations
free fatty acids due to increased lipolysis in adipose tissue. (307, 310).
Treadmill exercise in female rats (10 wk, 4 days/week) was
associated with cardiac hypertrophy, lower rates of glycol- Of note, this section has focused on the production of ATP
ysis, higher glucose oxidation, and higher rates of fatty acid generated from fatty acids and glucose because these are
oxidation. Rats subjected to this training protocol were also considered the main substrates during aerobic exercise.
protected against ischemia-reperfusion injury (improved re- However, during intense anaerobic exercise, lactate con-
covery of function associated with lower glycolysis, and centrations in the blood rise, and the heart predominantly
higher fatty acid oxidation) (53). Thus it was concluded uses lactate (240). In addition, while fatty acids and carbo-
that exercise induces a protective metabolic phenotype that hydrates have been considered the key substrates for car-

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 439


BERNARDO ET AL.

diac ATP production, it is now emerging that ketones and myocytes from mice subjected to 8 wk of aerobic treadmill
branched chain amino acids can play an important role in interval training compared with untrained mice (353).
contributing to energy production in some settings such as Treadmill running for 10 days in rats was reported to pro-
heart failure and can compete for acetyl-CoA (244). To tect against arrhythmia by better maintaining the mito-
date, the significance of these substrates for the heart in a chondrial membrane potential, lowering mitochondrial
setting of exercise is less clear, but it is noteworthy that ROS production, and preserving redox homeostasis (8).
ketone bodies are oxidized as a fuel source during exercise Mice with chronic elevation of PI3K in the heart (caPI3K
and can act as signaling metabolites (114). mice) were protected against maladaptive ROS in a setting
of diabetic cardiomyopathy (349). Finally, as described in
2. Redox control section IID1F, an 8-wk aerobic interval treadmill training
program in mice increased NO which is thought to buffer
Oxidative stress is a term typically used to describe elevated excess ROS in settings of cardiac pathology (353, 354).
intracellular levels of ROS due to an imbalance between free
radicals, ROS, and antioxidants which contribute to pa-
thology by causing damage to DNA, proteins, and lipids F. Cellular Stress Responses: Survival and
(369). ROS can be produced from a number of sites includ- Death
ing the cytosol, plasma membrane, nucleus, and peroxi-
somes. However, in cardiac myocytes, mitochondria are Since the majority of adult cardiac myocytes are unable to
considered the predominant source of ROS. Under normal/ proliferate or be renewed (331, 390, 391), systems to pro-
basal conditions, ROS are produced as byproducts of mito- mote myocyte survival and prevent cell death are critical for
chondrial electron transfer activity and are buffered by an- maintaining cardiac function. Cells have a number of sys-
tioxidant systems. These “physiological” ROS act as signal- tems/strategies to maintain normal physiological functions
ing molecules to regulate physiological processes (7, 357, and to monitor and cope in conditions of stress including 1)
369). In cardiac disease settings, chronic elevation of ROS heat shock response, 2) ubiquitin-proteasome system, and
can lead to mitochondrial dysfunction, cell death, inflam- 3) organelle-specific stress responses [e.g., mitochondria,
mation, and fibrosis (357). However, elevated ROS in re- endoplasmic reticulum (ER), autophagic-lysosomal path-
sponse to exercise can have beneficial effects in the heart (7, way, nuclear DNA response] (211) (FIGURE 7). Many of
357, 369). The distinct outcomes appear to be dependent on these systems work together to form a multilayered system
the exposure time to ROS (e.g., persistent vs. pulsatile), the to protect against pathological insults. Exposure to a mild/
proximity of ROS to moieties most susceptible to damage, intermittent stress such as exercise has been shown to acti-
and the capacity of the endogenous cellular ROS scavenging vate a number of stress responses that are considered to
mechanisms (i.e., antioxidant capacity). Exercise is associ- better prepare the cell against stronger/chronic stress stim-
ated with pulsatile, compensated oxidative stress (e.g., aug- uli (as may occur in a cardiac disease setting). Thus the
mentation of antioxidant capacity), whereas disease is as- maintenance of mechanisms for counterbalancing stress is
sociated with persistent and uncompensated oxidative considered a potential strategy for preventing cell death and
stress (i.e., the endogenous protective mechanisms are over- pathology (160, 211).
whelmed) (7). Excessive ROS is also known to activate
signaling cascades associated with cardiac disease including 1. Heat shock response
chronic CaMKII activation (112).
Heat shock proteins (Hsps) are molecular chaperones that
Exercise-induced mechanical loading/stretch of the heart, contribute to a number of protein quality control mecha-
as well as neurohumoral activation, has been implicated for nisms including the folding of newly synthesized proteins,
increasing ROS generation and the subsequent upregula- and the clearance/removal of misfolded proteins and aggre-
tion of antioxidant defense mechanisms in the recovery gates (47, 160). In a setting of increased misfolded proteins,
phase (e.g., glutathione reductase, thioredoxin reductase, refolding by Hsps and co-chaperones is considered the pref-
and manganese superoxide dismutase) (7, 159). ROS act- erential response. However, if the protein cannot be re-
ing as second messengers to alter redox-sensitive enzymes folded to its native form, it can be degraded by the ubiquitin
is considered to contribute to exercise-induced adaptive proteasome system (UPS), a process also involving chaper-
signaling. Adaptive responses to ROS also include mito- ones and co-chaperones (FIGURE 7). If the chaperone and/or
chondrial adaptations (e.g., increased biogenesis, en- UPS machineries become rate limiting, misfolded proteins
hanced mitochondrial phosphorylation capacity and ef- are likely to aggregate and can be removed via the process of
ficiency) and activity of cell quality control systems (e.g., autophagy (381). The Hsp70 family, and in particular the
autophagy/mitophagy; discussed in further detail in sec- inducible form of Hsp70, is considered primarily responsi-
tion IIF3A) (7). ble for the protective effects of the Hsp response induced by
exercise. Hsp70 is rapidly upregulated in response to cellu-
Of note, while ROS is elevated intermittently with exercise, lar stresses, such as heat, oxidative stress, alterations in pH,
basal ROS levels have been shown to be reduced in cardiac and increased Ca2⫹, many of which occur in response to

440 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

HEAT SHOCK
RESPONSE
Protein
synthesis AUTOPHAGY
Cathepsins Lysosome LYSOSOMAL DEGRADATION
Beclin 1
Protein Vps34 Bcl-2 EXERCISE
Hsp70 EXERCISE
folding

Cellular Cellular
Beclin 1
p62

7 Atg5
stress p62 p62 p62
stress Ub
Ub Ub
-II
Ub
-II
Ub ↑ Protein
LC3
Ub Ub
LC3 LC3
Ub Ub Ub Ub
Ub Ub
Ub Ub Ub
Degradation

A tg
Native Misfolded Protein
protein protein aggregate
Hsp70 MACROAUTOPHAGY Autophagosome LAMP-2A
Protein
E3 ligases Ub
refolding Hsp70 Ub
Ub
eg. CHIP HSF1 Ub ± E3:CHIP
HSPs
Nucleus
Hsc70
HSF1 MITOPHAGY + complex
HSP70 ATF(N)
Chaperones
HSPs Activated e.g. BiP
HSF1
Misfolded
HSPs XBP1s Chaperones protein
ER homeostasis
CHAPERONE-MEDIATED
EXERCISE ATF4 Chaperones AUTOPHAGY
FOXO Inhibition XBP1
of growth Antioxidants
Ub CHOP
Ub E3 ligases FOXO
Ub ATF(N)
Ub DNA
Hsp70 MAFbx K63 Golgi
Telomere
DAMAGE XBP1s Apparatus
E3 ligases MAFbx K48 dysfunction
MuRF1 ATF4
DNA damage
ROS Spliced ATF6
P BiP
eIF2α P
BiP
mPTP opening ↓ Protein IRE1
synthesis BiP
P
EXERCISE PERK BiP
Degradation BiP
26S Proteasome Mild UPR ER
BiP
Protective
UBIQUITIN- ER STRESS
Disease
PROTEASOME MITOCHONDRIA UNFOLDED PROTEIN
induced UPR
SYSTEM EXERCISE RESPONSE

FIGURE 7. Effects of exercise on stress response pathways in the heart. Exercise is able to regulate
components of the 1) heat shock response, 2) ubiquitin-proteasome system, 3) autophagy lysosomal degra-
dation, 4) the opening of the mPTP in mitochondria, 5) ER stress unfolded protein response, and 6) the DNA
damage response. Due to the complex nature of each process and multiple interactions between processes,
only those most relevant to exercise are shown.

exercise (202). Numerous studies have shown that exercise improvements in functional recovery in isolated rat hearts
upregulates Hsp70 in the rodent heart, in some cases in an subjected to ischemia-reperfusion injury (320). However,
intensity-dependent manner, and in response to acute and rats trained in the cold to prevent the induction of Hsp70
long-term exercise adaptations (67, 94, 160, 194, 276, 306, displayed a similar degree of cardioprotection to rats
406). Exercise-induced increases in myocardial Hsp70 con- trained in ambient temperatures in ex vivo and in vivo mod-
tent appear to occur primarily in response to the exercise- els of ischemia-reperfusion injury (340, 406). Furthermore,
induced increase in core body temperature, as there was no Hsp70 is not required for PI3K(p110␣)-induced cardiac
upregulation of Hsp70 in the hearts of rats trained in the protection, yet PI3K(p110␣) is a critical regulator of exer-
cold (148, 152, 406). Expression of Hsp70 is regulated by cise-induced hypertrophy and protection (434).
the transcription factor heat shock factor 1 (HSF1). Under
basal conditions, HSF1 is in an inactive form in the cyto- 2. Ubiquitin-proteasome system
plasm in a complex with Hsp70 and other Hsps. In the
presence of cellular stress, misfolded proteins competitively The UPS plays a major role for the routine turnover of
bind Hsps, and HSF1 is released from the complex. HSF1 proteins and the degradation of damaged proteins (211).
then translocates to the nucleus where it binds to heat shock The accumulation of damaged/misfolded proteins, referred
elements and upregulates the expression of Hsps including to as proteotoxicity, has an adverse effect on organelles and
Hsp70 (444) (FIGURE 7). cells. Proteotoxicity has been implicated in settings of car-
diac pathology, and dysfunction of the UPS has been re-
There is a large body of evidence suggesting that Hsp70 is ported in settings of human cardiomyopathy (86, 430).
critical for mediating protection in acute cardiac stress set- The UPS is responsible for degrading ~90% of proteins in
tings (444). However, the role of Hsp70 in chronic cardiac the majority of mammalian cells, within multiple com-
disease settings (34) and exercise is less clear. Silencing of partments (cytoplasm, nucleus, ER). UPS-induced pro-
Hsp70 with oligonucleotides attenuated exercise-induced tein degradation typically consists of two steps. First, a

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 441


BERNARDO ET AL.

protein is tagged with a polyubiquitin chain (four or dation and involves heat shock cognate protein 70
more ubiquitin molecules), a step involving three en- (Hsc70) and lysosomal membrane-bound lysosomal-as-
zymes: 1) ubiquitin-activation enzyme (E1), 2) ubiquitin- sociated membrane protein 2A (LAMP2a)] (211, 430)
conjugating enzyme (E2), and 3) ubiquitin ligase (E3) (FIGURE 7; microautophagy not presented). Most cardiac
(270, 318, 420). Second, the tagged protein is degraded research has focused on macroautophagy, and this has
by the proteasome (a large multisubunit protease com- typically been referred to as “autophagy” (430).
plex, 26S) (FIGURE 7). The proteasome is primarily a
cytosolic enzyme but is also located in nuclei and the ER Under basal conditions, autophagy functions as an intracel-
membrane. In most cases, chains of polyubiquitin are lular recycling system for protein and organelle quality con-
linked to the targeted protein through lysine 48 (K48; trol and is required for cardiac myocyte function and sur-
“canonical” ubiquitination). However, linkage can also vival. Deletion of autophagy specific genes (Atgs; e.g., Atg5)
occur with other lysine residues such as lysine 63 (K63; led to pathological cardiac hypertrophy and dysfunction in
“noncanonical” ubiquitination). As described below, mice (295, 404). In response to cellular stress, autophagy
ubiquitination also has roles independent of the protea- levels increase, and this process of degrading proteins is
some including the regulation of transcription factors, accompanied by the release of energy, permitting cells to
gene transcription, and autophagy (270). adapt to changing nutritional and energy demands (156).
Macroautophagy is considered to play a key role for de-
Hundreds of distinct E3 ligases have been identified, but grading aberrant protein aggregates and organelles in car-
only a relatively small number have been found to be muscle diac myocytes under disease settings (430). Fewer studies
specific (heart and skeletal muscle) (270). The role of the have explored the role of autophagy in the exercise-trained
UPS in a setting of cardiac hypertrophy (exercise- or dis- heart. However, a key study from He et al. (156), utilizing
ease-induced) is not well understood, but it has been shown mice that transgenically express a green fluorescent protein
that both protein synthesis and UPS-controlled protein deg- (GFP)-labeled marker of autophagasomes (LC3), demon-
radation are elevated in the hypertrophied heart (318). At strated that autophagy also plays an important role in the
least two muscle-specific E3 ligases have been shown to be heart in response to exercise. It was shown that treadmill
regulated in the heart with exercise, i.e., muscle atrophy
exercise-induced autophagy in the heart in vivo within 30
F-box (MAFbx, also known as atrogin-1) and muscle RING
min, and this was associated with conversion of LC3-I to
finger protein 1 (MuRF1) (5, 270, 318). With the use of
LC3-II (autophagosome-membrane-associated form of
genetic mouse models, it was shown that MAFbx inhibits
LC3), and degradation of p62 (autophagy substrate pro-
exercise- and IGF1-induced cardiac growth via the activa-
tein). It was also shown that exercise-induced disruption of
tion of Forkhead box transcription factors (Foxo1 and
the Bcl-2-beclin 1 complex is essential for exercise-induced
Foxo3a) (231). A mechanism involving the ubiquitination
autophagy in the heart; Bcl-2 is an anti-autophagy protein
of K63 polyubiquitin chains and the regulation of transcrip-
that inhibits autophagy by direct interaction with beclin 1
tion rather than degradation by the proteasome was re-
(FIGURE 7). Mutant mice with normal levels of basal au-
ported (231, 318) (FIGURE 7).
tophagy but deficient in exercise-induced autophagy (due to
3. Organelle-specific stress responses mutations in the phosphorylation sites of Bcl-2) displayed
lower maximal exercise capacity (baseline skeletal muscle
A) AUTOPHAGY-LYSOSOMAL DEGRADATION PATHWAY. In addition to strength and cardiac function were normal) (156). It was
the proteasome, the lysosomal system is the other main not directly assessed whether this was due to defects in
system responsible for the turnover of proteins. In contrast skeletal muscle, heart, or both. Other studies in exercise-
to the proteasome system which selectively degrades nor- trained rodents have also reported an increase in au-
mal proteins (typically with short half-lives) and abnormal tophagy-related and -regulatory genes (e.g., LC3-II, p62,
proteins, the autophagic-lysosomal pathway is considered Bcl-2) in cardiac muscle (312, 402). In addition, Chen et al.
the main mechanism responsible for the degradation of pro- (65) showed a link between autophagy of proteins in the
teins with longer half-lives. The lysosomal system is also heart and exercise-induced cardiac protection in rabbits
able to degrade whole organelles (e.g., mitophagy) and with MI.
large protein aggregates (167, 211, 304) (FIGURE 7). Three
types of autophagy have been described based on the lyso- The carboxy terminus of Hsp70-interacting protein (CHIP)
somal delivery system: 1) macroautophagy [involving fu- regulates a number of quality control pathways and can
sion of an autophagosome (a double membrane vesicle with also affect exercise-induced autophagy. CHIP is a co-chap-
sequestered damaged proteins/cytoplasmic materials/or- erone that interacts with Hsp70 to mediate refolding, and
ganelles) with a lysosome to form an autolysosome, fol- also acts as a ubiquitin ligase (E3 Ub ligase) (444). More
lowed by digestion of the material], 2) microautophagy (ly- recently, CHIP was shown to regulate autophagy via a pro-
sosome directly engulfs cytosolic components), and 3) cess termed “chaperone-assisted autophagy” which is ubiq-
chaperone-mediated autophagy [requires unfolding of the uitin-dependent (different from chaperone-mediated au-
protein and internalization into the lysosome for degra- tophagy which is ubiquitin-independent) (FIGURE 7). The

442 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

role of CHIP was assessed by subjecting CHIP KO mice to B) MITOCHONDRIA. Mitochondria can respond to a variety of
5 wk of voluntary wheel running. In hearts of wild-type stress signals (e.g., oxidative stress, hypoxia, loss of growth
mice, there was no significant effect on autophagy-related factors, and DNA damage) and are able to switch on a cell
genes (e.g., Atg7, Atg5, Vps34). However, each of these death program (necrosis or apoptosis) by opening of the
genes was elevated in hearts from running CHIP KO mice mitochondrial permeability transition pore (mPTP; located
(no change in sedentary mice). After 1 wk of running, CHIP in the inner mitochondrial membrane; FIGURE 7). Under
KO mice were reported to have increased autophagic flux normal healthy conditions, the inner mitochondrial mem-
(based on LC3BII/LC3BI ratio after 1 wk), and LAMP2a brane is largely impermeable, only allowing equilibration of
and cathepsin L expression were elevated after 5 wk. There ions and water and small molecules. This regulation allows
was no significant increase in autophagy regulators in wild- mitochondria to maintain a pH gradient and membrane
type mice, but there was a trend for an increase in LC3BII potential which drives oxidative phosphorylation. In set-
and LAMP2a. CHIP KO mice also displayed an exagger- tings of stress, opening of the mPTP causes collapse of the
ated hypertrophic response to wheel running compared membrane potential, swelling of the mitochondria, and cell
with wild-type mice, but this was not associated with mark- death (144). It has been suggested that acute exercise (60
ers of pathology (e.g., no evidence of fibrosis and cardiac min treadmill bout) and chronic exercise (5 wk treadmill
function was normal; mild nonsignificant increase in ANP) running) have the capacity to decrease mPTP induction/
(443). The enhanced physiological response in the KO was opening and apoptotic signaling in settings of cellular or in
attributed to loss of CHIP being associated with enhanced vivo cardiac stress (e.g., doxorubicin-treated rats). In these
IGF1-Akt signaling and increased autophagic flux. The in- studies, mitochondrial swelling, mitochondrial function,
vestigators speculate that enhanced autophagy in CHIP KO and/or protein expression of apoptotic and mPTP-related
mice after running wheel exercise would allow misfolded proteins were assessed (11, 250).
proteins and/or mitochondria that accumulate during run-
ning to be cleared. However, interpretation of this study is C) ENDOPLASMIC RETICULUM AND UNFOLDED PROTEIN RESPONSE. The en-
complex because the CHIP KO have increased misfolded doplasmic reticulum (ER) controls the synthesis, folding,
proteins under basal conditions and are more susceptible to and assembly of proteins before trafficking in vesicles to the
MI (278, 461). The authors note that the increase in au- Golgi apparatus, cytosol, and plasma membrane. Only cor-
tophagy in running CHIP KO could be due to a compensa- rectly folded proteins are transported out of the ER. Short-
tory mechanism to decrease the burden of unfolded/ term ER stress (as may occur with acute exercise) will trig-
damaged proteins, or CHIP inhibiting autophagy. Hsc70 is ger the unfolded protein response (UPR) which is initially
critical for the chaperone function of CHIP as well as chap- considered adaptive because it involves transduction events
erone-mediated autophagy. Thus it is possible that when which eliminate misfolded proteins and restores ER homeo-
CHIP is present, it targets Hsc70 for ubiquitination and stasis. However, with prolonged ER stress, the UPR can
proteasomal degradation, limiting its availability for au- lead to cell dysfunction and cell death. The ER stress re-
tophagy. While the mechanism will require further exami- sponse begins with the ER chaperone [ER-specific HSP70
nation, it appears CHIP regulates autophagy in response to family member: glucose-regulated protein kinase 78
exercise (443). (Grp78)/BiP] sensing unfolded or damaged proteins and
dissociating from three UPR sensors: 1) inositol requiring
Preliminary data suggest that defective autophagy (due to element-1(IRE-1), 2) PKR-like ER kinase (PERK), and 3)
deletion of Atg7) leads to a maladaptive cardiac phenotype activating transcription factor 6 (ATF6) (FIGURE 7). This is
in response to voluntary wheel running in a setting of met- followed by the activation of pathways to reestablish ER
abolic stress (obesity and insulin resistance). Premature homeostasis by inhibiting protein synthesis and increasing
death occurred in 40% of heart and skeletal muscle-specific the capacity of the ER (protein folding and degradation).
Atg7 KO mice that had been placed on a high-fat diet for 12 PERK activation reduces protein synthesis from further
wk and subjected to voluntary wheel running. Death was overwhelming the ER via the inactivation of eukaryotic
attributed to cardiomyopathy associated with cardiac en- translation initiation factor 2␣ (eIF2␣). Activated ATF6
largement and fibrosis (238). shuttles to the Golgi, is cleaved into an active transcription
factor which enters the nucleus, and upregulates the tran-
Finally, when considering upstream mechanisms regulating scription of ER chaperone proteins including Grp78/BiP.
autophagy, it is noteworthy that autophagy is controlled by Activation of IRE-1 leads to splicing and activation of X-
mTOR which is regulated by growth factors including IGF1 box-binding protein 1 (XBP-1), and ultimately increased
which are released from the heart in response to exercise expression of genes involved in ER homeostasis. However,
(301). Collectively, the studies above highlight the complex with prolonged ER stress, the UPR can lead to cell dysfunc-
interactions between protein quality control mechanisms tion and cell death via activation of CHOP (a transcription
within the heart, and further work will be required to un- factor of the C/EBP family) and caspase pathway (211, 241,
derstand how exercise regulates these processes. 279).

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 443


BERNARDO ET AL.

Heart disease is associated with a defective ER stress re- KO mice, and treatment with IGF1 and growth hormone
sponse (142, 143, 235). Cardiac-disease-associated mito- upregulated myocardial telomerase activity and increased
chondrial dysfunction, calcium imbalance, and oxidative pAkt (438). Thus it was concluded that exercise-induced
stress disrupt proper protein folding, resulting in the accu- telomerase activity was mediated by IGF1, Akt, and eNOS.
mulation of misfolded proteins in the ER. One group re- In another study, cardiac telomere length decreased with
ported that short-term exercise (5 days of aerobic treadmill age in the developing rat heart, and this was associated with
running) had no effect on ER stress adaptation in rats sub- an increase in miR-34a gene expression (254), a PI3K-reg-
jected to ischemia-reperfusion (based on the assessment of ulated miRNA (236) (see sect. IIIA2C).
protein expression) (293). More recently, there have been
reports to suggest that exercise can blunt ER stress in a 4. Cell death
setting of ischemia-reperfusion or MI (45, 46). In rats sub-
jected to moderate-intensity treadmill running (60 min, 5 When cellular stress responses are inadequate, cell death
days/week for 8 wk) 4 wk post-MI, cardiac function tended will occur. Within the heart, cell death can occur via apo-
to be better compared with untrained MI rats, and this was ptosis, necrosis, and potentially autophagy (FIGURE 8).
associated with lower expression of UPR markers (e.g., BiP There is also growing evidence of crosstalk between au-
and CHOP which were elevated with MI) and lower mis- tophagy, apoptosis, and necrosis (294). Of these, apoptosis
folded protein (46). In another study, intermittent hypoxia has been best characterized in the heart in response to ex-
for 3 wk induced proapoptotic ER stress in the heart includ- ercise. In the rodent heart, exercise (e.g., voluntary or tread-
ing higher BiP, pPERK, ATF4, a trend for elevated CHOP mill running ranging from 3 wk to 6 mo) has been accom-
and higher cleaved caspase-3. High-intensity exercise on a panied by unchanged or decreased levels/rates of apoptotic
treadmill for 10 days during the last 2 wk of hypoxia was markers (176, 386, 438). Exercise training for 12 wk was
associated with prevention of these proapoptotic ER stress shown to attenuate age-induced cardiac apoptosis
markers and reduced ischemia-reperfusion-induced infarct (TUNEL) in rats, and this was associated with lower protein
size (45). In that study, exercise alone seemed to induce a expression of some components of the death-receptor-
mild UPR including an increase in BiP and trends for higher dependent apoptotic pathway (extrinsic; FADD, caspase-8,
pPERK and ATF4. However, this was not associated with caspase-3) and the mitochondria-dependent apoptotic
an increase in CHOP or cleaved caspase-3. Futhermore, in pathway (intrinsic; caspase-9, caspase-3) (219) (FIGURE 8).
both studies only associations between exercise and ER PI3K-Akt signaling has also been shown to mediate protec-
stress in the heart were presented (i.e., no direct evidence of tion by preserving mitochondrial integrity and function,
cause and effect). Studies in genetic mice specifically target- and inhibiting apoptotic pathways (144). In a study in aged
ing components of ER stress will be required to assess the rats, it was suggested that swim exercise (5 times/week for
precise role of ER stress and exercise-induced cardiac pro- 12 wk) attenuates apoptosis via increasing SIRT1-PGC-1␣
signaling and replaces IGF1-PI3K-Akt signaling. However,
tection. For instance, utilizing ATF6 KO mice, it was shown
components of the IGF1-PI3K-Akt pathway remained ele-
that the UPR plays an important role in mediating the ad-
vated in aged mice with exercise. Thus this conclusion is
aptation to exercise in skeletal muscle (450). In summary,
somewhat questionable and relies on protein expression
an exercise-induced mild UPR may provide protection by
data alone (219).
upregulating defense mechanisms which better prepare the
heart to respond to a more severe disease-induced chronic
There is evidence for substantial crosstalk between au-
UPR.
tophagy and apoptosis (294) (FIGURE 8). For instance, the
interaction between Bcl-2 (anti-apoptotic protein) and be-
D) NUCLEAR DNA RESPONSE-TELOMERE SHORTENING. Cardiac disease
clin 1 (autophagy protein) allows for a switch between au-
and aging are associated with increased ROS that can cause
tophagy and apoptosis. When there is less interaction be-
DNA damage (FIGURE 7). As described earlier, exercise can
tween Bcl-2 and beclin 1 (e.g., in response to exercise),
attenuate disease-induced ROS. Telomere shortening is also autophagy will increase (156). Caspase-3 can inhibit au-
known to contribute to the DNA damage response. Telo- tophagy by cleaving beclin 1, and caspase-9 is reported to
meres are DNA-protein structures at chromosome ends. promote autophagy by a mechanism involving Atg7 and
These sequences protect chromosomal DNA from degrada- LC3-II (232).
tion and shorten with each cell division (377). Shortening of
telomeres has been associated with a range of age-related
diseases including cardiovascular disease. Several studies G. Maintenance of the ECM and the
have shown that chronic exposure to exercise is associated Antifibrotic Properties of Exercise
with telomere length maintenance (161, 246). Voluntary
wheel running for 21 days in mice was shown to decrease The ECM in the heart provides a highly organized struc-
markers of cellular aging, upregulate cardiac telomerase tural framework surrounding cardiac myocytes, allowing
activity, and increase telomere-stabilizing proteins in the for the coordination of mechanical, electrical, and chemical
heart. These exercise-induced changes were absent in eNOS processes so that cardiac myocytes contract in a coordi-

444 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

IGF1
Death
EXTRINSIC receptor

IGF1R NECROSIS
Cytoplasm Opening
or
of mPTP FADD
APOPTOSIS
Caspase-8
EXERCISE
mPTP
PI3K Akt EXERCISE

ROS
INTRINSIC
Bax Bax
Apoptotic Bak
Beclin 1 signal
Bcl-2
Outer mitochondrial membrane
permeabilisation

EXERCISE Bcl-2
Cytochrome C
Beclin 1 Bcl-2
Caspase
activation

Beclin 1
EXERCISE Caspase-9

AUTOPHAGY
EXERCISE Caspase-3

APOPTOSIS
FIGURE 8. Effects of exercise on cell death pathways in the heart. Exercise regulates key players of cell death
pathways (intrinsic/extrinsic apoptosis, necrosis, and autophagy). Each pathway is distinct in their own cellular
processes, but there is crosstalk between apoptosis and autophagy. Akt (downstream of PI3K) phosphorylates
and inhibits the pro-apoptotic factor Bax. Due to the complexities of cell death pathways, only those relevant to
exercise are depicted.

nated way resulting in a uniform and regular cardiac con- blasts (direct interactions, paracrine and mechano-electric
traction (108, 256, 423). The ECM consists of collagens, mechanisms) (215, 256, 416, 423).
glycoproteins (e.g., fibronectin, laminin, elastin), proteogly-
cans, extracellular proteases, and ECM receptors. In the A common feature of the diseased heart and pathological
normal healthy heart, collagens are the most abundant cardiac hypertrophy is cardiac fibrosis, i.e., excessive depo-
structural components of the ECM and are produced sition of ECM proteins by cardiac fibroblasts. Fibrosis
mainly by fibroblasts. Previous studies had suggested that within the heart and other organs is considered one of the
fibroblasts account for as much as two-thirds of the total most serious health problems in current medicine (356,
cell population in the heart (108, 215, 256, 423). However, 407). In the heart, cardiac fibrosis leads to cardiac stiffen-
a more recent study using improved tools and markers sug- ing, impaired electrical conductivity, and cardiac dysfunc-
gests that fibroblasts account for closer to 11% of the cells tion that can lead to arrhythmia and heart failure (416).
in the heart (328). Balance of the ECM via collagen ECM Myofibroblasts (smooth muscle-like fibroblasts) in the dis-
synthesis and degradation is critical for maintaining cardiac eased heart are considered key players responsible for car-
function. This is achieved by matrix metalloproteinases diac fibrosis. In contrast, appreciable expression of myofi-
(MMPs) which are enzymes that degrade ECM proteins and broblast markers has not been detected in normal healthy
tissue inhibitors of metalloproteinases (TIMPs) which pre- adult hearts (215). Myofibroblasts can originate from mul-
vent excessive ECM degradation by MMPs. The ECM, tiple sources including resident fibroblasts (considered the
MMPs, and TIMPs can be regulated by numerous factors main source), smooth muscle cells, perivasculature peri-
and mechanisms including cytokines and growth factors cytes, epithelial-to-mesechymal and endothelial-to-mesen-
[e.g., tumor necrosis factor-␣ (TNF-␣), TGF-␤, angiotensin chymal transition, fibrocytes, and other myeloid progeni-
II, endothelin], inflammatory signaling, the adrenergic sys- tors. In response to a pathological stress, cardiac fibroblasts
tem, and crosstalk between cardiac myocytes and fibro- transform to myofibroblasts and express higher levels of

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 445


BERNARDO ET AL.

proinflammatory and profibrotic factors and secrete ECM wk after anterior MI had a reduction in collagen content in
proteins to promote wound healing (422). While the initial the left ventricle and also in the scar region (337). Some
transient activation of myofibroblasts is considered an studies have identified mechanisms contributing to the anti-
adaptive response to maintain the structural integrity of the fibrotic effects of exercise, but this represents an area for
heart, persistent myofibroblast activation leads to excessive further investigation. Fibrosis in the aged rat heart was
ECM deposition, and a maladaptive phenotype leading to associated with an imbalance in the MMP/TIMP ratio (e.g.,
the progression of heart failure (422). Factors and signaling decreased MMPs and increased TIMP-1) and increased
pathways involved in early activation of cardiac fibroblasts TGF-␤; exercise training attenuated these changes (215,
and in pathological remodeling have been the focus of nu- 216). With the use of genetic mouse models, it has been
merous investigations and include angiotensin II, endothe- demonstrated that IGF1, IGF1R, and/or PI3K signaling
lin-1, TGF-␤, RhoA, G protein-coupled receptor kinase 2 protects against cardiac fibrosis in cardiac disease settings
(GRK2), Smads, and mitogen-activated protein kinase including pressure overload, dilated cardiomyopathy, MI,
(MAPKs) (256, 416, 422, 423). Therapeutic approaches and atrial fibrillation (266, 269, 335, 415, 434). Further-
that target some of these factors have shown some efficacy, more, approaches that target PI3K-regulated miRNAs also
but additional therapies are greatly needed (223, 407). have been associated with anti-fibrotic properties (see sect.
Since exercise has been shown to have anti-fibrotic proper- IVA2B) for further details). AMPK has also been implicated
ties in preclinical models of heart disease, this represents an in the protective effect of swim training against isoprotere-
active area of investigation. nol-induced cardiac fibrosis by inhibiting ROS production
(248).
Unlike pathological cardiac hypertrophy, physiological car-
diac hypertrophy in response to moderate aerobic exercise H. Exercise-Induced Inflammatory Response
training is not associated with fibrosis (interstitial or
perivascular) in humans or animal models. In male athletes
The effects of exercise on the immune system are complex
(cyclists or weight lifters) with enlarged hearts, there was no
and depend on the intensity and duration of the exercise
evidence of fibrosis based on imaging techniques (98, 221).
performed (122). In general, acute or vigorous/high-inten-
Commonly used exercise training protocols in rodents
sity exercise induces an inflammatory response, character-
(swimming, running) which induce physiological hypertro-
ized by an increase in the number of leukocytes in the cir-
phy are not associated with fibrosis compared with seden-
culation, a process known as leukocytosis (296, 361), and
tary controls (216, 269, 408). For example, it was shown
increases in circulating cytokines, such as interleukin-6
that collagen content of the cardiac ECM in exercised rats
(IL-6; pro- and anti-inflammatory) and TNF-␣ (pro-inflam-
(10 wk of treadmill running) was not increased, and colla- matory) (48, 122). The demargination and mobilization of
gen I/III ratio was maintained (54). Gene expression of leukocytes during exercise arises from the increase in hemo-
collagen I and III in the hearts of rats was not significantly dynamics (which increases shear stress), as well as increased
changed following a 13-wk treadmill training protocol activation of the sympathetic nervous system (385). The
(176). No alterations in collagen deposition or osteopon- inflammatory response to acute exercise is transient, return-
tin-1 protein expression were detected in hearts of rats ing to baseline within a few days (189). In contrast, regular
following 8 wk of treadmill training (289). Furthermore, physical activity or exercise training is associated with
expression of ␣-smooth muscle actin, a marker of myo- reduced levels of inflammation. This may provide benefit
fibroblast activation contributing to collagen remodel- in settings of chronic inflammation, such as insulin resis-
ing, was unchanged in exercise-trained rat hearts after 13 tance, obesity, metabolic syndrome, and type 2 diabetes,
wk of treadmill running (176). However, while fibrosis is conditions that increase cardiovascular disease risk
not a feature of the exercise-trained heart, it is notewor- (113), as well as in aging (131, 345). Exercise training
thy that cardiac hypertrophy in response to exercise or reduces inflammatory markers in patients with chronic
activation of the IGF1R-PI3K pathway is associated with heart failure (3, 76, 338, 339, 419), a syndrome charac-
changes in the collagen profile (215, 267, 409). The func- terized by low-grade chronic inflammation (99). Whether
tional significance of these changes is currently not well the anti-inflammatory effects of exercise directly contrib-
understood. ute to cardioprotection in these settings requires further
investigation.
Exercise training has been shown to attenuate fibrosis in
diseased or aging hearts. Swim training in mice was able to The molecular mechanisms responsible for the anti-inflam-
attenuate fibrosis induced by pressure overload or isopro- matory effects of exercise are still unclear. IL-6 is a cytokine
terenol (248, 434). Similarly, exercise training (involuntary with both pro- and anti-inflammatory properties (325). In-
treadmill) had no discernible effects on fibrosis and cardiac creased IL-6 synthesis and secretion from contracting skel-
remodeling in young rats, but mitigated age-related eleva- etal muscle during exercise may provide benefit by promot-
tion of fibrosis in old rats (216, 408). Furthermore, mice ing the secretion of anti-inflammatory cytokines (such as
that underwent endurance training for 6 wk before and 4 IL-10 and IL-1 receptor agonist), reducing TNF-␣ levels

446 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

and upregulating glucose and lipid metabolism (51, 321). teins and nucleic acids that can be transported between cells
However, long-term aerobic exercise training reduces basal and tissues (358). There is some evidence to suggest that
IL-6 levels in humans (122) and reduces IL-6 levels in exercise can increase the abundance of small extracellular
experimental models of cardiac disease/remodeling. For vesicles in humans (126), and it was reported that exosomes
example, in a rat model of cardiac hypertrophy induced were released from the hearts of type 2 diabetic (db/db)
by sustained ␤-adrenoceptor stimulation (isoproterenol mice subjected to acute exercise (64). A recent study dem-
administration), treadmill running for 12 wk prevented onstrated that an exercise stress test in at-risk human
isoproterenol-induced increases in myocardial IL-6 and subjects was associated with an increased quantity of
TNF-␣ expression (375). In an exercise intervention plasma extracellular vesicles (assessed using nano-flow
study, MI caused significant increases in plasma concen- cytometry; no significant change in extracellular vesicle
trations of IL-6 and TNF-␣ in sedentary rats, but not in size) (20). The same investigators showed that swim
rats that underwent 8 wk of treadmill running, 4 wk after training in mice for 3 wk was associated with increased
the induction of MI (309). Downregulation of Toll-like
levels of serum extracellular vesicles (~1.85-fold). Injec-
receptor 4 (TLR4) may also contribute to the anti-inflam-
tion of the same quantity of extracellular vesicles from
matory effects of exercise. TLR4 is a transmembrane
exercised and sedentary mice into the left ventricle before
receptor that induces inflammatory cytokine production
ischemia-reperfusion injury provided comparable pro-
when activated, and TLR4 expression was reduced in
trained subjects compared with sedentary subjects in two tection based on infarct area and markers of apoptosis.
aged cohorts (8 wk/6 mo of resistance training) (263, However, injection of exercise-induced extracellular ves-
350). icles in a quantity similar to that observed with swim
exercise (1.85-fold) significantly reduced infarct area and
Aerobic exercise also indirectly protects the heart via its apoptosis further (20). Using a cardiomyoblast cell line
effects on the vasculature. During aging, increased oxida- (H9C2 cells) in combination with siRNA and chemical
tive stress and inflammation lead to stiffening of the large inhibitors, it was mechanistically shown that 1) IGF1 was
elastic arteries and vascular endothelial dysfunction, which sufficient to increase the release of extracellular vesicles
increases the risk of atherosclerosis and other cardiovascu- into the cell culture medium, and this was due in part to
lar diseases. Studies in humans and experimental animals increased expression of extracellular vesicle biogenesis-
have demonstrated that aerobic exercise training improves and secretion-related genes (ALIX and RAB35) in IGF1-
endothelial function and that the pertinent mechanism in- treated cells; and 2) exercise-derived extracellular vesi-
volves reduced expression of the pro-inflammatory tran- cles protect H9C2 cells from H2O2-induced apoptosis via
scription factor NF-KB and reductions in oxidative stress ERK1/2 and HSP27. Data from this study suggest that
(362). the exercise-induced increase in the number of extracel-
lular vesicles rather than content mediate cardiac protec-
tion. However, it is noteworthy that there is also evidence
I. Communication Between the Heart and
to suggest that exercise can change the contents of extra-
Other Organs/Systems in Response to
cellular vesicles (64). Additional studies will be required
Exercise to identify the major source of exercise-induced extracel-
lular vesicles.
Communication between multiple organs/systems in the
body in settings of health and disease has emerged as an
In response to exercise, heart rate increases and blood
important factor contributing to the final phenotype/pre-
flow going to skeletal muscle and the heart increases
sentation observed in animal models and patients (188,
significantly. It is recognized that in response to exercise,
213, 321). Exercise is recognized to affect numerous sys-
contracting skeletal muscle acts as an endocrine organ to
tems within the body, but the underlying mechanisms re-
communicate with other tissues/organs such as adipose
sponsible for the crosstalk between organs, including con-
tracting organs (heart and skeletal muscle) and noncon- tissue, liver, and brain to control energy metabolism
tracting organs, is not well understood. It has been (188, 321). The concept of the heart as an endocrine
proposed that exercise-induced release of factors (e.g., pep- organ was recognized in the 1980s with the identification
tides, metabolites, DNA, mRNA, miRNAs, and other RNA that ANP and BNP are expressed in the heart and se-
species) from organs may play a role in mediating the mul- creted upon stimuli such as mechanical and hemody-
tisystemic effects of exercise. These factors have been namic stress. ANP and BNP control cardiovascular func-
termed “exerkines” and may have effects on distant organs tion via actions on distal tissues including the kidneys,
and cells (i.e., endocrine signaling), in addition to effects on adrenal, and brain (374). Factors released from the heart
cells within the same organ (paracrine signaling) or the cell in response to cardiac insults/stress have been identified
itself (autocrine signaling). Interest in this research field has (102). To date, factors released specifically from the
grown with the increased knowledge regarding extracellu- heart in response to exercise remain relatively un-
lar vesicles (exosomes, microvesicles) which contain pro- known.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 447


BERNARDO ET AL.

III. GENOME, TRANSCRIPTOME, ular pathways, and phenotypes. Proteomic and metabolo-
PROTEOME, AND METABOLOME: mic measurements have become more reliable with ad-
FROM EXERCISE-INDUCED vances in mass spectrometry. Thus it is now possible to
SIGNATURES TO THERAPIES AND/OR identify thousands of factors that are regulated in response
BIOMARKERS to stimuli such as exercise. The current challenge is the
analysis of huge data sets to delineate the key drivers and
The identification of exercise-induced molecular signatures complex processes underlying exercise-induced cardiac
may hold promise for the development of new therapies or protection (119). In this section, we briefly describe some
biomarkers for cardiac pathology and heart failure. Heart studies that have explored the effects of exercise on the
failure was recently classified as the most rapidly growing cardiac genome, transcriptome, proteome, and metabo-
cardiovascular condition globally (466). This has been at- lome.
tributed to an increased aging population and preventable
childhood diseases transitioning to cardiovascular diseases.
The majority of cases are considered to be preventable by A. Genome and Transcriptome
targeting lifestyle factors (466). However, this approach is
best implemented by identifying individuals at risk as early
Protein coding genes are the most well-studied sequence,
as possible. Regular physical activity is associated with re-
duced incident heart failure (106). but only account for ⬍2% of the genome (292). In the
last decade, previously unknown noncoding RNA species
Under basal, exercise, or disease settings, cell functions are have been discovered and add a new dimension to the
a consequence of complex interactions between networks regulation of transcription (292). Here, we highlight
of the genome, transcriptome, proteome, and metabolome both coding and noncoding RNAs dynamically regulated
(397) (FIGURE 9). Exercise represents a dynamic and com- in the heart with exercise training (swimming, treadmill
plex process. Early studies largely used reductionist meth- running, voluntary free wheel), as well as genetic-based
ods/approaches to target individual genes or proteins regu- rodent models with physiological hypertrophy (e.g.,
lated by exercise. However, significant improvements in transgenic activation of the IGF1-PI3K-Akt pathway)
“omics” technologies have substantially enhanced the ca- or artificially selected rats with high running perfor-
pacity to define the relationships between genetics, molec- mance.

EXERCISE

Genomic
coding DNA PHENOTYPE
GENE ENZYMATIC
TRANSLATION
EXPRESSION FUNCTIONS

GENOME TRANSCRIPTOME PROTEOME METABOLOME


gtcg tacggg g
ccg atgatc
acac ggatcccc tagct
a tg
Next generation Proteins Lipids
ctcgta tcagatac tagcc
ga c c ca a g tt
ccctt ttagga sequencing
tttttaa g P C G A T
Nucleic
Epigenetic acids
factors Gene array Exercise trained
e.g. Methyl group mRNA
Proteins with Amino acids heart
post-translational Sugars
miRNA modifications
e.g. phosphorylation
Genomic Physiological
O-GlcNAc
non-coding cardiac
DNA hypertrophy
Genetic
REGULATORY
variant
RNAs
EXERCISE
miRNA

FIGURE 9. The use of multi-omics platforms to identify novel mechanisms and uncover exercise signatures.
Integrating data from multi-omics systems to understand genetic variants and epigenetic marks (genome and
epigenome), gene expression and miRNAs (transcriptome), proteins (proteome), and metabolites (metabo-
lome) during exercise to define molecular pathways of exercise.

448 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

1. Gene expression profiling higher in hearts from sedentary HCR (56). The authors
conclude this suggests a switch from normal fatty acid ox-
In the early-mid 2000s, several groups utilized microarray idation to carbohydrate metabolism in the LCR. Genes re-
technology to investigate gene expression changes in the lated to cellular stress (e.g., DNA damage, inflammation)
hearts of rodents in response to endurance training (swim- and pathological growth signaling (e.g., genes associated
ming and running) (69, 129, 136, 208, 396). These analyses with embryonic growth) were also higher in the LCR (56).
identified the differential expression of genes involved in
glucose/insulin signaling, protein biosynthesis, epidermal Few investigators have conducted profiling studies in large
growth factor signaling, fatty acid oxidation/lipid metabo- animal models subjected to exercise. In a swine model that
lism and cell survival (69, 129, 136, 208, 396). Galindo et had undergone treadmill running for 3– 6 wk, the investi-
al. (129) compared 8 different microarray data sets from gators reported that changes in gene expression observed in
models of exercise-induced hypertrophy and identified a set rodent models were not necessarily observed in swine.
of 59 genes with functions related to cell growth, heart However, a common feature in the swine and previous ro-
contraction, cell death, and ECM organization. Further- dent studies was the regulation of genes related to PI3K-Akt
more, the PI3K-Akt signaling pathway was statistically signaling (214). In this study, transcription factors differen-
overrepresented in the identified genes across all models. tially regulated between physiological and pathological hy-
These data sets were also compared against rodent models pertrophy were also identified including the glucocorticoid
of pathological hypertrophy and human cardiac disease. receptor and paired box 6 (Pax6; functional significance in
The value of identifying genes differentially regulated in the heart remains unclear) (214).
models of exercise-induced physiological hypertrophy and
models of pathological hypertrophy was highlighted by More recent studies have used RNA sequencing (RNA-seq)
Sakamoto et al. (359). Using a DNA chip array, they iden-
which allows for higher sensitivity, accuracy, reproducibil-
tified genes that were upregulated in hearts of rats with
ity, and the capacity to identify splice variants (253). RNA-
exercise-induced physiological hypertrophy (8 wk of volun-
seq performed in hearts of swim-trained mice confirmed
tary running) but not pressure overload-induced patholog-
known pathways reported from microarray studies, and
ical hypertrophy (5 wk). Approximately 100 genes were
identified novel pathways (e.g., suppression of autoim-
differentially elevated in each model compared with con-
mune-related pathways) and alternative splice variants
trol. Of these, expression of Hsp70 and HSF1 was higher in
(389). Interestingly, the investigators detected more alter-
the physiological model but not the pathological model. To
native splice isoform changes compared with gene expres-
assess the role of HSF1 in the exercise-trained heart, the
sion changes after exercise, suggesting that exercise-in-
investigators subjected HSF1-deficient heterozygote mice
(HSF1⫹/⫺) to voluntary wheel running. The exercise-in- duced cardiac remodeling may be mediated by modification
duced hypertrophic response was not blunted in HSF1⫹/⫺ of critical domains via alternative splicing without quanti-
mice, but cardiac function was impaired, and the typical tative changes in mRNA expression (389). Identified path-
exercise-induced increase in Hsp70 was prevented. This re- ways related to alternative splicing included cell signaling/
sult is consistent with HSF1 and Hsp70 playing a role for transduction (calcium signaling, insulin signaling) as well as
exercise-induced adaptations (see sect. IIF1). Furthermore, pathways related to cardiac disease.
mice with constitutive activation of HSF1 in the heart were
protected in a setting of pressure overload-induced patho- To identify key or new regulators of physiological cardiac
logical hypertrophy (359). hypertrophy and protection, we and others have under-
taken profiling studies in IGF1R, PI3K, and Akt transgenic
Gene expression studies have also been undertaken in mice (236, 267, 370, 384). A large-scale computational
hearts from rats artificially selected for high and low run- analysis integrating transcriptomic data from in vivo mu-
ning capacity (HCR and LCR, respectively) (448). By 35 rine models of physiological cardiac hypertrophy (short-
wk, LCR display evidence of cardiac pathological remodel- term cardiac-specific Akt transgenic induction, cardiac ac-
ing (myocyte hypertrophy, fibrosis, increased fetal gene ex- tivation of PI3K:caPI3K transgenic, swimming) identified a
pression) (93, 348) and by 15–20 mo show defects in myo- conserved core network involved in processes such as coor-
cyte function (depressed cardiomyocyte contractile func- dinated regulation of angiogenesis, induction of autophagy
tion and relaxation as well as impaired Ca2⫹ handling) compatible with cell survival, the TSC2/mTOR pathway,
(165, 204). LCR are also more susceptible to risk factors of and mitochondrial cytochrome c oxidase (last component
cardiovascular disease (high blood pressure, hyperlipid- to the electron transport chain that facilitates the aerobic
emia, insulin resistance), whereas HCR are protected and production of ATP) (103). In another profiling study we
have better myocyte function (448). Between the two compared gene expression in hearts from caPI3K mice
groups, 1,540 of 28,000 screened genes were found to be with physiological hypertrophy (due to increased PI3K),
differentially expressed. In sedentary LCR, genes involved hearts from dnPI3K mice (with smaller hearts due to
in glucose metabolism were more highly expressed in the decreased PI3K), and hearts from a cardiac disease model
heart. In contrast, genes related to lipid metabolism were (MI) (236). Using bioinformatics we filtered for genes

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 449


BERNARDO ET AL.

that were differentially regulated in the different models, (292). To date, the best studied noncoding RNAs are miR-
correlated with heart function, and selected genes with NAs. miRNAs are small noncoding RNAs (~19 –24 bp) that
relatively high expression in the heart. Growth factor have been shown to regulate gene/protein expression (26).
receptor-bound 14 (Grb14) represented one of the top miRNAs can be regulated by exercise, as well as by signal-
candidate genes, but the role in the heart was largely ing cascades in the heart which induce exercise-induced
unknown. We subsequently studied Grb14 KO mice and physiological heart growth (e.g., PI3K).
showed that Grb14 protects the heart against cardiac
pathology under basal conditions and in a setting of MI. A) EXERCISE-REGULATED miRNAs. miRNAs dynamically regulated
Loss of Grb14 in mice induced a pathological cardiomy- after swimming or running in rodents and mouse models of
opathy characterized by an increase in heart and atrial physiological hypertrophy have been reviewed in detail in
weight, depressed cardiac function, interstitial fibrosis, References 315 and 117. Three cardiac specific miRNAs
and decreased activation of Akt (236). This strategy (miR-1, miR-133a, miR-133b) were the first miRNAs to be
highlights the potential of identifying novel downstream studied in exercise models. Two separate groups confirmed
functional targets that may be manipulated/targeted as downregulation of these miRNAs in swim-trained and
therapeutic approaches. interval-trained rats (61, 388). However, these miRNAs
were also decreased in a model of pathological hypertro-
2. Noncoding RNAs phy induced by pressure overload (61). Subsequently, a
number of groups have tried to identify miRNAs that
Noncoding RNAs include miRNAs, PIWI-interacting play distinct roles in the heart in a setting of exercise and
RNAs, small nucleolar RNAs, and long noncoding RNAs disease.

miR-134 miR-21 miR-206


miR-1
miR-208a miR-146a miR-208b
miR-222
miR-133a miR-20a
miR-126 miR--221
Circulation
miR-499 EXERCISE miR-149*

Cardiac tissue: myocyte, fibroblast etc. miR-99b miR-222


miR-29a,c miR-100 miR-126
miR-17
miR-143 miR-144
miR-154 miR-124
miR-21 ↓ p27
↑ IGF1R miR-19b
miR-27a,b ↓ HIPK1
↓ TIMP3 ↓ Spred-1
↑ ACE2 ↑ p15 miR-223
↑ PI3K ↓ PTEN
↓ Collagen miR-34 ↓ PI3KR2
genes miR-145 (p85β)
↓ ACE miR-208a/b
↑ Ang (1-7) ↑ Akt
↓ TSC2 miR-652 Cell cycle/
↓ Ang II proliferation ↑ MAPK
↑ αMHC
miR-26b
miR-27a
↓ GSK3β ↓ βMHC
↑ Jagged1 ↑ PI3K-
↑ Vinculin ↑ Vegf Akt-
miR-214 eNOS
Anti-fibrotic
↓ Pathological actions
remodeling Physiological
↑ Serca2a
heart growth & Anti-apoptotic
cell survival Preservation of
cardiac contraction & Angiogenesis
electrical conduction

FIGURE 10. miRNAs regulated by exercise or PI3K in the heart and circulation. An overview of cardiac and
circulating miRNAs regulated by exercise or PI3K and changes in their target genes (direct/indirect) that may
contribute to characteristic features associated with physiological cardiac hypertrophy. miRNAs that are
downregulated are highlighted in green, and miRNAs upregulated are highlighted in blue. miR-17 in this figure
is the miR-17-3p strand.

450 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

Several groups have discovered miRNA signatures in hearts cise, serum levels of miR-17 were increased in chronic heart
of exercise rodent models (swim training and strength train- failure patients following an exercise test. Inhibition of
ing in rats) which target genes associated with collagen miR-17 with an antagomiR in mice partially blocked exer-
deposition/fibrosis (miR-29) (388), angiogenesis (miR-126) cise-induced heart growth in vivo. This was associated with
(84), the renin-angiotensin system (miR-143, miR-27a, attenuation of myocyte hypertrophy, reduced markers of
miR-27b) (118), the PI3K/Akt/mTOR pathway (miR-21, cardiomyocyte proliferation, and regulation of TIMP3,
miR-144, miR-124, miR-145, miR-19b, miR-99b, miR- PTEN, and Akt signaling (380).
100) (249), Akt/GSK3␤ (miR-26b, miR-27a) (257),
SERCA2a (miR-214) (272), and ␣/␤ MHC ratio (miR- B) EXERCISE-INDUCED CIRCULATING miRNAs.The expression of
208a/b by targeting Med13, Pur␤, Sox6, Sp3, HP1␤) (387) miRNAs also change in the circulation in response to
(FIGURE 10). exercise (FIGURE 10). The exact source of these miRNAs
is unclear, but they have the potential to act on the heart
miRNAs are also regulated in rodent hearts in response to and other organs. Circulating miRNAs (c-miRNAs) are
exercise in a heart disease setting (273, 274, 393). Three protected from degradation through inclusion in extra-
miRNAs (miR-1, miR-29, and miR-24) were dysregulated cellular vesicles or association with RNA-binding protein
in a setting of MI, and exercise training prevented this dys- complexes (80). The regulation of c-miRNAs in response
regulation, at least in part (273, 274). Another group iden- to different modes of exercise (marathon running, row-
tified a set of miRNAs (miR-598, miR-429, miR-224, miR- ing, resistance training) have recently been reviewed
425, miR-221) that were regulated by treadmill exercise in (330) and are considered to have potential value as exercise
a rat model of heart failure due to aortic stenosis. It was biomarkers and markers of fitness capacity.
suggested that regulation of these miRNAs may provide
protection because they target genes related to cell death, Baggish et al. (15) reported a unique signature set of
TGF-␤ signaling, and metabolic processes (393). c-miRNAs that were dynamically regulated in response to
acute exhaustive cycling exercise and sustained aerobic
To date, the functional significance of the majority of exer- rowing exercise training in healthy competitive athletes
cise-regulated miRNAs in directly mediating exercise-in- (miR-21, miR-146a, miR-221, and miR-222; miR-20a was
duced remodeling and protection has not been comprehen- upregulated only in response to sustained exercise training).
sively assessed in vivo. Examples of miRNAs that have been However, this study only investigated c-miRNAs with
examined and shown to have a biological role are miRNA- known roles related to exercise adaptations such as angio-
222, miR-223, and miR-17–3p. miR-222 was robustly up- genesis, exercise-induced inflammation, and skeletal and
regulated in hearts of mice in response to exercise (volun- cardiac muscle contractility. Since this study, Sawada et al.
tary wheel-running or swim training), and miR-222 was (364) performed microarray analysis of c-miRNAs and
shown to target cell proliferative genes (243). Inhibition of their response to acute resistance training. Circulating levels
miR-222 prevented the increase in exercise-induced heart of miR-149/miR-149-3p were increased following resis-
growth; however, transgenic cardiac overexpression of tance training, whereas miR-146a and miR-221 were sig-
miR-222 was not sufficient to induce an increase in heart nificantly reduced post resistance training. Investigation
size with exercise training (243). miR-223 was also found to of heart/muscle specific miRNAs (myomiRs) and inflam-
be elevated in hearts of treadmill trained mice, and cardiac mation-related miRNAs in plasma of male endurance
overexpression of miR-223 induced physiological heart athletes following a marathon run revealed increased ex-
growth (enhanced cardiac function, no fibrosis) (457). The pression levels of the myomiRs miR-1, miR-133a, miR-
key direct targets of miR-223 responsible for inducing phys- 206, miR-208b, and miR-499, whereas the fibrosis/in-
iological heart growth are not entirely clear as miR-223 was flammation-related miRNAs (miR-21 and miR-155)
shown to regulate both pro-hypertrophic and anti-hyper- were not affected by exercise (288). Similarly, another
trophic signaling pathways. The investigators postulate study identified a robust increase in circulating levels of
that the pro-hypertrophic pathways outweigh the anti-hy- miR-1, miR-133a, miR-208a, and c-miR-146a in athletes
pertrophic pathways, and this leads to secondary Akt acti- immediately following prolonged aerobic exercise (i.e., a
vation and physiological hypertrophy (457). A recent study marathon run); miR-134, miR-126, and miR-499 were also
demonstrated that the expression of miR-17 (miR-17–3p) altered but to a smaller degree. miR-1 and miR-208a were
was upregulated in hearts from mice after voluntary wheel not affected after acute exhaustive exercise (upright cycling
running for 21 days and in isolated adult cardiomyocytes of 30 min duration). Thus the duration and type of exercise
from mice that had undergone swim training for 21 days appears to affect the regulation of muscle specific c-
(380). Conversely, the expression of miR-17 was decreased miRNAs (16).
in cardiac tissue under pathological settings (a mouse model
of pressure overload induced by transverse aortic constric- C) PI3K-REGULATED miRNAs. Using the same approach utilized to
tion and in human dilated cardiomyopathy heart samples) identify Grb14 (see sect. IIIA1), we identified a set of 62
(380). Further supporting the role of this miRNA in exer- miRNAs (e.g., miR-34, miR-652, miR-154) differentially

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 451


BERNARDO ET AL.

regulated in caPI3K and dnPI3K hearts and diseased hearts components of the HDAC complex in mice (271), but an-
due to MI (236). These miRNAs have mRNA targets that other study showed increased cardiac HDAC2 activity fol-
can regulate angiogenesis, cardiac contraction, electrical lowing 3 days of forced swimming (194). Findings with
conduction, apoptosis, and fibrosis (FIGURE 10). PI3K-reg- chronic exercise training have also been variable. While one
ulated miRNAs (miR-34, miR-652, and miR-154) and an study reported no significant change in the activity of
exercise-regulated miRNA (miR-222) have subsequently HDAC enzymes in the hearts of mice that underwent
been targeted in cardiac disease settings and shown to pro- chronic exercise (voluntary wheel running for 3 wk; trend
vide benefit (see sect. IVA2B). for an increase in class IIa and b in males) (209), another
study in high-volume swim-trained rats (10 wk) reported
3. Genetic variants increased protein expression of HDAC1 and decreased
HDAC3 and HDAC4 protein levels (387).
Whole genome sequencing (WGS) provides the most com-
prehensive coverage of an individual’s genetic variation. Several studies have documented exercise-induced epige-
WGS allows for sequencing of the entire genome including netic changes in endocrine tissues/cells (fat, blood, brain,
1) protein coding genes, 2) noncoding intronic regions of skeletal muscle) that may directly or indirectly exert down-
genes, and 3) large tracts of intergenic sequences. With a stream effects on the heart (reviewed in detail in Refs. 96,
substantial reduction in DNA sequencing costs over the last 237, 308, 467). In human adipose tissue biopsies, individ-
decade (cost per genome in 2006 ⬎$10M vs. 2015–2016
uals involved in a 6-mo moderate-intensity exercise pro-
~$1K) (296a), and improvements in technology and bioin-
gram displayed hypermethylation of genes (including
formatics, WGS has become a realistic alternative to tar-
HDAC4) with parallel changes in gene expression (352).
geted panel screens (e.g., 50 –100 cardiac disease-related
After a single bout of exercise, skeletal muscle biopsies from
genes) and exome sequencing (captures 1% of the genome
normal individuals demonstrated changes in genome-wide
that codes for proteins). Over 150 genetic markers have
been linked to athletic performance (6). It will be important histone acetylation and DNA methylation levels (264). The
to confirm these genetic variants in larger cohorts. How- authors showed augmented nucleo-cytoplasmic shuttling of
ever, it is of interest that some of the variants have been HDAC4 and elevated ubiquination levels of HDAC5
identified in genes involved in processes associated with through exercise (264). Furthermore, disruption of class IIa
exercise-induced cardiac adaptation including cardiac HDAC corepressor complexes with the HDAC inhibitor
growth (IGF1R), angiogenesis (NOS3/eNOS, VEGF), Scriptaid recapitulated the effects of exercise in mice, in-
and E-C coupling (KCNJ11, encodes Kir6.2). With fu- creasing the expression of metabolic genes in skeletal mus-
ture studies utilizing WGS, the discovery of additional cle when administered acutely, and enhancing whole-body
genetic variants is anticipated. In combination with in energy expenditure, increasing lipid oxidation, and reduc-
silico modeling, this may provide important insight on ing fasting blood glucose levels when administered chroni-
gene variants associated with differential responses to cally (130).
exercise (adaptive/maladaptive and/or responders/nonre-
sponders) (6). Translocation of class IIa HDACs (e.g., HDAC4 and
HDAC5) from the nucleus to the cytosol promotes cardio-
myocyte hypertrophy in vitro (153, 287, 426) and has been
B. Epigenetic Adaptations
documented in the failing myocardium (164). However, the
effects of exercise on the subcellular localization and func-
Epigenetics (histone acetylation, histone methylation, and
tion of cardiac class IIa HDACs require further investiga-
DNA methylation) has emerged as a major mechanism reg-
tion. An elegant study by Backs et al. (13) demonstrated
ulating gene expression. One well-known histone modifica-
that acute ␤-adrenergic activation (as occurs during ex-
tion is histone acetylation, tightly regulated by two families
ercise) leads to PKA-dependent proteolytic cleavage of
of enzymes known as histone acetyltransferases (HATs)
and histone deacetylases (HDACs). Enzymes involved in HDAC4 and accumulation of the resulting NH2-terminal
histone acetylation have been extensively studied in patho- fragment (termed HDAC4-NT) in the nucleus.
logical cardiac remodeling; class I HDACs (HDAC 1,2,3) HDAC4-NT contains a myocyte enhancer factor-2
promote transcription of hypertrophic genes, while class IIa (MEF2) binding domain and selectively represses MEF2
HDACs (HDAC 4,5) repress hypertrophy (433). Another activity, preventing the transcription of genes involved in
widely studied epigenetic mark is DNA methylation. A hy- pathological remodeling; the activity of other transcrip-
permethylated DNA region is strongly correlated with gene tion factors, such as SRF (which is associated with cell
suppression. survival) was unaffected. It has been proposed that this
mechanism protects the heart from undergoing maladap-
Exercise-induced epigenetic adaptations in the heart have tive remodeling in physiological stress settings (such as
not been examined extensively. A single bout of acute ex- during exercise, which leads to repetitive transient eleva-
ercise did not alter the protein levels of HDACs 1–5 and tions in catecholamines).

452 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

C. Proteome 376). Since metabolites are downstream of transcriptional,


translational, and posttranslational processes, they are con-
Significant developments in the field of proteomics includ- sidered to serve as the most proximal readouts of changed
ing sample preparation and enrichment strategies, high-res- conditions (e.g., exercise or disease) (FIGURE 9). Conse-
olution mass spectrometers, and high-throughput analysis quently, metabolomics may have potential for identifying
tools have allowed investigators to study proteins in much novel biomarkers with clinical utility, as well as mecha-
greater depth, including the identification and role of pro- nisms of cardiovascular disease development and exercise-
tein posttranslational modifications (PTMs) (120, 401). induced protection (376), particularly in individuals who
PTMs including phosphorylation, N-glycosylation, are well phenotyped and serve as their own biological con-
O-GlyNAcylation, proteolytic cleavage, redox, deamida- trols (346). Another potential advantage of profiling the
tion, sumoylation, citrullination, methylation, and lysine metabolome is the more manageable number of metabolites
acetylation have been identified within numerous compart- (estimated ⬍10,000) in comparison to the genome
ments/organelles within cardiac myocytes including the cell (⬎20,000 genes), transcriptome (⬎106 mRNAs), and pro-
membrane, cytoplasm, mitochondria, SR, Golgi apparatus, teome (⬎106 proteins). Mass spectrometry and nuclear
nucleus proteasome, and sarcomere (234, 401). Conse- magnetic resonance spectroscopy have been the two main
quently, PTMs have the potential to affect numerous pro- technologies used for metabolite profiling.
cesses within the heart including E-C coupling, metabolism,
gene transcription, and cell survival/death. Data are emerg- Metabolomics has already shown its utility as a tool in
ing to show that acute and/or chronic exercise can regulate humans for assessing the effect of exercise on cardiac risk
total protein expression and PTMs in the rodent heart from markers such as insulin resistance (376). Exercise inter-
healthy animals and models of disease. Ferreira et al. (119) ventions (low-, moderate-, and high-intensity exercise:
integrated cardiac proteomics data from eight studies in cycle ergometer, treadmill, elliptical trainers) in seden-
which rats had been subjected to exercise training (swim- tary, overweight, or mildly obese subjects (STRRIDE
ming or treadmill) for different time periods (2, 6, 8, or 54 trial) were associated with alterations in metabolic pro-
wk). Some common proteomic signatures included upregu- files. These profiles were indicative of more efficient mi-
lation of antioxidant systems, activation of kinases (e.g., tochondrial fatty acid oxidation and were associated
PKA), fatty acid oxidation, vasculogenesis, and tissue re- with reduced cardiac risk markers (e.g., improved insulin
generation (119). One study identified changes in the phos- sensitivity) (166, 376). It was also shown that metabolic
phorylation profile of mitochondrial proteins that was in- signatures obtained during exercise (treadmill or cycle
dicative of increased glucose oxidation, protein translation, ergometer) in human subjects with normal exercise ca-
and redox protection (120). Exercise (acute or chronic) has pacity (no evidence of myocardial ischemia) provided
also been associated with decreased cardiac protein O- signatures of exercise performance and susceptibility to
GlcNAc by a number of groups (22, 24, 271); an exception cardiovascular disease (230). Changes in metabolites re-
to this was in a mouse model of type 2 diabetes, in which flected alterations in glycolysis, lipolysis, adenine nucle-
moderate treadmill exercise increased protein O-GlcNAc in otide catabolism, amino acid catabolism, glycogenolysis,
the heart (79). Increased levels of O-GlcNAc are a feature of
oxidative stress, and insulin sensitivity. Furthermore, a
many pathologies including diabetes, aging, ischemia, and
set of metabolites that were identified in humans during
heart failure. However, whether this increase contributes to
exercise were able to regulate the expression of transcrip-
declining heart function or is an adaptive mechanism in
tional regulators of metabolism in cell culture (cultured
response to stress, is unclear (255). The therapeutic poten-
myotubes), and one was confirmed in skeletal muscle of
tial of targeting proteins that play roles in mediating exer-
mice (230).
cise-induced physiological hypertrophy is highlighted by
the identification of Hsp20. Hsp20 was detected by mass
Metabolic signatures in response to exercise have also
spectrometry and two-dimensional electrophoresis exclu-
been identified in animal studies. Monleon et al. (286)
sively in hearts from exercise-trained rats (6 wk of progres-
performed metabolic profiling on plasma from mice sub-
sive treadmill running) (42, 55), and cardiac-specific trans-
jected to 18 mo of spontaneous exercise (running wheels
genic overexpression of Hsp20 was able to protect the heart
against cardiac injury (115). provided to mice at 3 mo of age). Voluntary exercise for
18 mo was accompanied by increased maximal running
capacity and grip strength. Alterations in plasma metab-
D. Metabolome olites in the exercise group (at rest) were indicative/sug-
gestive of increased lipolysis (increased total fatty acids,
Metabolomics or metabolite profiling refers to the system- unsaturated fatty acids, glycerol), altered glucose metab-
atic analysis of metabolites within a biological sample (e.g., olism (decreased alanine and lactate), altered catechol-
blood, urine, biological tissues). Metabolites are defined as amine synthesis (elevated tyrosine and phenylalanine),
low molecular weight biochemicals and include nucleo- and increased phospholipid metabolism (lower choline).
tides, sugars, amino acids, organic acids, and lipids (346, Heart glucose consumption (assessed by positron emis-

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 453


BERNARDO ET AL.

sion tomography) was also lower in the exercise group, IV. THERAPEUTIC STRATEGIES:
possibly due to increased fatty oxidation in the heart TARGETING THE PROTECTIVE ACTIONS
(although not confirmed in this study) (286). OF EXERCISE

Lai et al. (220) performed metabolic profiling and transcrip- Earlier sections of this review highlight the wide range of effects
tomic profiling in hearts from mice with compensated hy- that exercise has on a number of cell types, organelles, and pro-
pertrophy due to pressure overload (TAC) and decompen- cesses within the heart, as well as other systems/organs that can
sated hypertrophy (TAC together with a small MI), and then have secondary effects on the heart. It is unlikely a single
compared this to hearts from mice with physiological hy- therapeutic intervention would meet the criteria of recapitulating
pertrophy (8 wk of voluntary wheel running; less cardiac all the beneficial effects of exercise. However, a number of ap-
enlargement than the pathological models). The metabolo- proaches that target key mechanisms responsible for exercise-
mics signatures were shown to more robustly distinguish induced remodeling have been targeted and shown some degree
the exercise and pathological models than the transcrip- of protection (FIGURE 11).
tomic results. In the decompensated heart failure model, the
metabolite profile (e.g., increased acylcarnitine species) sug- A. Approaches Targeting the IGF1-PI3K-Akt
gested a bottleneck in carbon substrate flux (fatty acids and Pathway
glucose) into the TCA cycle which could limit ATP produc-
tion and cause contractile dysfunction. In contrast, many of The IGF1-PI3K(p110␣) pathway is elevated in the heart
these metabolites were decreased in the exercise-trained in response to exercise in animal models and humans
heart, suggesting matched carbon substrate flux and no (refer to sect. IIB1; Refs. 33, 35, 435), and numerous
bottlenecking. groups have demonstrated that various IGF1, IGF1R,

EXERCISE
NATURAL PRODUCTS
Flavonoids NH2 OH

O N
Icaritin N

Benfotiamine N SMALL
MOLECULE
Ca2+ ISCHEMIC IGF1
BGP-15
CONDITIONING
ICaL IGF1R

Cytoplasm

Ca2+ RNAi BASED THERAPIES &


OLIGONUCLEOTIDES miR-222
PI3K-regulated miR-17-92
PI3K
miRNAs cluster
Sarcoplasmic antimiR-34a/34
RyR2
reticulum
antimiR-652
Ca2+ SMALL
antimiR-154 MOLECULE
SERCA2a GENE THERAPY
X SC79
2+
Ca Akt
SUMO1 AAV-caPI3K
SUMO1

SMALL
Regulation of
MOLECULE
miRNA targets Proliferation of
N106
GENE THERAPY cardiac myocytes
AAV-SERCA2a

Reduced pathology
Improved/preserved heart function
FIGURE 11. Therapeutic strategies: targeting the protective actions of exercise. A schematic representing
gene-based (RNAi and viral vectors), natural products, ischemic conditioning, or small molecule-based thera-
pies that target the IGF1R-PI3K-Akt signaling axis or calcium handling.

454 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

PI3K(p110␣), and Akt genetic mouse models are pro- 1. Strategies targeting IGF1/IGF1R
tected in models of cardiac stress/disease including pres-
sure overload (266), dilated cardiomyopathy (266, 415, A) IGF1. Investigators have explored whether intrapericardial
437), diabetic cardiomyopathy (181, 349), atrial fibrilla- delivery of IGF1 or targeting IGF1 to injured heart tissue is
tion (335), and models of oxidative stress and ischemia able to mediate protection without adverse consequences.
(e.g., MI) (233, 236, 428). IGF1 and/or PI3K have also Some preliminary data show favorable outcomes. How-
been shown to regulate a number of processes/responses ever, animal numbers have been low or follow-up periods
that are also mediated by exercise including cardiac myo- have been short (TABLE 3). Thus further studies are
cyte growth, angiogenesis, electrical remodeling, and E-C warranted.
coupling; attenuation of fibrosis; promotion of cell sur-
B) IGF1R: BGP-15, SMALL MOLECULE WHICH INCREASES IGF1R PHOSPHORY-
vival; and regulation of mitochondrial function. Thus
LATION. BGP-15 is a hydroximic acid derivative that was
enhancing cardiac IGF1-PI3K(p110␣) signaling in pa-
known to act as a co-inducer of the stress-inducible form of
tients with heart failure may be beneficial. However, in
Hsp70 in some cell types (81), but whether it would induce
the majority of studies using the IGF1/IGF1R/PI3K/Akt
Hsp70 in the adult stressed heart in vivo was unknown. We
genetic mouse models, gene expression had been modi-
previously found that Hsp70 was elevated in the exercise-
fied before a cardiac insult/stress or simultaneously with
trained heart and caPI3K transgenic mouse heart, but not in
the stress. For an approach to enter the clinic, it is neces-
the dnPI3K exercise-trained heart which showed a blunted
sary to assess interventions at clinically relevant time physiological hypertrophic response (434). In addition,
points to develop and identify tools or agents with the Hsp70 was known to provide benefit in acute cardiac stress
potential to enter the clinic (i.e., interventions that can be settings (34). BGP-15 had also been tested in clinical trials
administered and provide benefit once cardiac pathology for other diseases (e.g., diabetes) and shown to be safe and
is present). Acute versus chronic activation of a pathway well tolerated, with no cardiac side effects being reported
or target also needs to be considered. Even though the (239). Thus it was of interest to examine whether BGP-15
majority of IGF1/IGF1R/PI3K/Akt transgenic models provided any therapeutic benefit in mouse models with car-
have shown protection in cardiac stress settings (265), diac pathology. Oral administration of BGP-15 in mice
there are also examples where this approach has not had with heart failure and atrial fibrillation for 4 wk was asso-
a favorable outcome. For instance, long-term transgenic ciated with reduced episodes of arrhythmia, improved car-
expression of the IGF1 ligand in the heart transitioned diac function, and decreased cardiac fibrosis (363). How-
from a physiological to pathological cardiac hypertro- ever, unexpectedly, the mechanism by which BGP-15 ex-
phic phenotype in mice (92). In addition, clinical trials in erted its protective effects appeared to be via increased
which IGF1 was chronically administered to humans phosphorylation of the IGF1R, rather than via the induc-
were disappointing (73, 344). One explanation for neg- tion of Hsp70 expression (363). BGP-15 provided benefit in
ative or adverse findings could be related to chronic IGF1 a heart failure model in which Hsp70 had been deleted, and
elevation activating fibroblasts and causing cardiac fibro- BGP-15 was shown to phosphorylate the IGF1R in neona-
sis. Over time, this would lead to pathological hypertro- tal rat ventricular myocytes (363). Daily administration of
phy and cardiac dysfunction. Subsequent clinical trials BGP-15 is considered to cause a short pulsatile increase in
used recombinant human growth hormone (rHGH) to IGF1R signaling because BGP-15 has a relatively short half-
increase circulating IGF1 levels. Preliminary studies re- life (81). Thus this may reduce the potential for chronic
ported favorable effects of rHGH administration on clin- long-term adverse effects observed with chronic IGF1 li-
ical symptoms and heart function (116, 132, 262). How- gand delivery.
ever, improvements in heart function were not recapitu-
2. Strategies targeting PI3K
lated in a large-scale clinical trial (317), possibly due to
acquired growth hormone resistance, a syndrome that
We developed an adeno-associated viral
A) PI3K GENE THERAPY.
affects 60 –70% of cachectic heart failure patients (10). (AAV) vector encoding the same caPI3K(p110␣) construct
For long-term treatments, cardiac selectivity or specific- to that used to generate the caPI3K transgenic mice (434).
ity is also of importance given that uncontrolled activa- Recombinant AAV serotype 6 (rAAV6) was combined with
tion of the IGF1-PI3K-Akt pathway can cause tumori- the cytomegalovirus (CMV) promoter to achieve robust
genesis in other organs and cell types (95, 127, 174). and selective expression in cardiac myocytes (140, 434).
Thus investigators have explored and developed new Adult mice administered rAAV6-CMV-caPI3K developed
strategies for activating the IGF1-PI3K-Akt pathway in physiological hypertrophy, reminiscent to that of exercised
the heart. These include different delivery systems, at- trained mice and caPI3K transgenic mice (434). Next, we
tempts to target damaged heart tissue, small molecules, investigated the therapeutic potential of rAAV6-CMV-
ischemic conditioning (pre-, post-, and remote-condi- caPI3K administration in mice with established LV remod-
tioning), and naturally occurring compounds, such as eling and cardiac dysfunction (4 wk of pressure overload
herbs (FIGURE 11 AND TABLE 3). induced by TAC) (434). Mice administered rAAV6-CMV-

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 455


BERNARDO ET AL.

Table 3. Interventions associated with protection and activation of the IGF1-PI3K-Akt pathway
Reference
Intervention/Drug Cardiac Stress Model Observation Nos.

IGF1 conjugated to Hoechst (histological Myocardial IR in mice Intravenous delivery of Hoechst-IGF1 (H-IGF1) 199
stain that binds to double-stranded selectively bound to exposed DNA and
DNA); tail vein injection targeted the injured heart. Twenty-eight
days post-MI, H-IGF1 was associated with
preserved cardiac function and less
fibrosis.
Intrapericardial delivery of IGF1 (150 MI in sheep (occlusion of LAD IGF1 was associated with increased ejection 261
␮g·kg-1·day-1) for 14 days with gel foam; procedure fraction which remained elevated 14 days
repeated until ejection after the last treatment (compared with
fraction was ⱕ40%) control saline); Note: only 3 sheep in each
group.
BGP-15 Mice with heart failure and Administration of BGP-15 by oral gavage was 363
atrial fibrillation associated with reduced episodes of
arrhythmia, improved cardiac function, and
decreased cardiac fibrosis.
AAV6-caPI3K Pressure overload in mice Improved cardiac function, increased 434
angiogenesis, and more favorable
expression of proteins involved in
cardiomyocyte contraction.
PI3K-regulated miRNAs: miR-34 family Study 1: pressure overload Study 1: treatment with antimiR-34 28
(transverse aortic improved cardiac function and attenuated
constriction, TAC) in mice lung congestion. This was associated with
reduced cardiac fibrosis, increased
angiogenesis, increased Akt activity,
decreased atrial natriuretic peptide gene
expression, and maintenance of
sarcoplasmic reticulum Ca2⫹-ATPase gene
expression.
Study 2: MI in mice Study 2: treatment with antimiR-34
attenuated cardiac remodeling and atrial
enlargement in the MI model.
In both studies, improved outcome in
antimiR-34-treated MI and TAC mice was
accompanied by upregulation of several
direct miR-34 targets, including vascular
endothelial growth factors, vinculin, protein
O-fucosyltranferase 1, Notch1, and
semaphorin 4B.
PI3K-regulated miRNAs: miR-34a Study 1: pressure overload in Study 1: inhibition of miR-34a in mice with 27, 32
mice moderate cardiac pathology attenuated
atrial enlargement and maintained cardiac
function, but had no significant effect on
fetal gene expression or cardiac fibrosis.
Inhibition of miR-34a in mice with severe
pathology provided no therapeutic benefit.
Study 2: dilated Study 2: disease prevention in LNA-antimiR-
cardiomyopathy (DCM) 34a-treated DCM female mice was
characterized by attenuated heart
enlargement and lung congestion, lower
expression of cardiac stress genes (B-type
natriuretic peptide, collagen gene
expression), less cardiac fibrosis, and
better cardiac function
Study 3: heart failure and Study 3: no evidence of significant protection
atrial fibrillation in the severe heart failure and atrial
fibrillation model in either sex.
PI3K-regulated miRNAs: miR-652 Pressure overload in mice Inhibition of miR-652 improved cardiac 31
function, attenuated cardiac hypertrophy,
reduced cardiac fibrosis, was associated
with less apoptosis, and preserved
angiogenesis. Jagged1 (a miR-652 target
and Notch1 ligand) protein and mRNA was
upregulated with antimiR-652 treatment.
Continued

456 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

Table 3.—Continued
Reference
Intervention/Drug Cardiac Stress Model Observation Nos.

PI3K-regulated miRNAs: miR-154 Pressure overload in mice Treatment with antimiR-154 improved 30
cardiac function and attenuated
pathological remodeling. Treatment was
associated with attenuation of heart and
cardiomyocyte size, less cardiac fibrosis,
lower expression of ANP and BNP,
attenuation of profibrotic markers, and
increased expression of p15 (a miR-154
target and cell cycle inhibitor).
Akt gene therapy Ischemia-reperfusion injury in Decreased apoptosis and/or infarct size and 128, 260
rodents improved cardiac function.
SC79-Akt activator Ischemia (35 min) and No protection against ischemic injury and no 290
reperfusion (60 min) in reduction in infarct size.
rats
Flavanol extraction from propolis Isoproterenol-induced cardiac Flavanol attenuated isoproterenol-induced 400
(traditional medicine) hypertrophy in mice (25 increases in ANP and ␤-MHC. The flavanol-
mg·kg-1·day-1 for 7 days) induced response was prevented with
wortmannin* (PI3K inhibitor). No cardiac
function measures with isoproterenol,
flavanol and with or without wortmannin.
Icaritin (traditional Chinese medicinal Myocardial IR in rats Icaritin reduced infarct size and was 464
herb) associated with increased Akt
phosphorylation, reduced markers of
inflammation and oxidative stress, and
better cardiac function. Wortmannin
inhibited some measures of icaritin-
induced protection (infarct size, markers of
inflammation, and oxidation).
Benfotiamine (Vitamin B1 analog which Study 1: type 1 diabetes Study 1: benfotiamine prevented systolic and 191
directs glucose to the pentose model (STZ) and type 2 diastolic dysfunction in vivo, and treatment
phosphate pathway) (db/db) was associated with increased Akt
signaling. Some beneficial responses of
benfotiamine in cardiac myocytes (e.g., cell
survival) were attributed to PI3K-Akt
signaling by using LY294002 and Ad.DN-
Akt. However, the contribution of the PI3K-
Akt pathway in vivo was not assessed.
Study 2: type 1 diabetic mice Study 2: benfotiamine treatment 190
(STZ) subjected to MI administered 4 wk before MI was
associated with better post-MI survival and
cardiac function as well as less apoptosis.
In vitro studies in cardiac myocytes with
Ad.DN-Akt implicated Akt signaling.
Sevoflurane (inhaled anesthetic)-induced Myocardial IR in rats SPC protects against IR injury (infarct size, 462
postconditioning (SPC) cardiac dysfunction, apoptosis). BEZ235
(PI3K/mTOR dual inhibitor) blunted SPC-
induced protection (infarct size, contractile
dysfunction, apoptosis).
Ischemic preconditioning (IP) MI in pigs (LAD ligation, IP was associated with smaller infarct size, 398
measured 7 days post-MI) fewer malignant ventricular arrhythmias,
less collagen content, better systolic
function, and vascular density.
LY294002* (PI3K inhibitor) prevented the
IP-induced protection.

*Wortmannin and LY294002 can regulate targets other than PI3K. DCM, dilated cardiomyopathy; IP, ischemic preconditioning; IR, ischemia-
reperfusion; LAD, left anterior descending artery; MI, myocardial infaraction; STZ, streptozotocin; TAC, transverse aortic constriction.

caPI3K displayed improvements in systolic function over ments in heart function were accompanied by increased
the 10 wk following rAAV6-CMV-caPI3K delivery. At the angiogenesis and more favorable expression of proteins in-
end of the study period, fractional shortening of treated volved in cardiomyocyte contraction (MHC isoforms and
mice had returned to pre-surgery values, whereas systolic SERCA2a). This study provides evidence that increasing
function of control mice remained poor (434). Improve- cardiac PI3K(p110␣) activity after the onset of cardiac dys-

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 457


BERNARDO ET AL.

function can improve outcome (434). The same PI3K gene B) AKT SMALL MOLECULE: SC79-AKT ACTIVATOR. Pharmacological ac-
therapy also provided protection in a mouse model of dia- tivation of Akt has also been considered an approach that
betic cardiomyopathy with established diastolic dysfunc- may be useful in promoting myocyte survival in the diseased
tion (334). With advances in vector technology, design, and heart. The small molecule SC79 was shown to activate Akt
delivery, gene therapy represents a promising therapeutic in the adult rat heart (290). However, acute pharmacolog-
strategy for the treatment of cardiac disease (146). ical activation of Akt by SC79 failed to reduce infarct size
and did not protect the rat heart against ischemic injury
B) PI3K-miRNA BASED DRUGS.Studies in our laboratory identified (290). Further studies examining dose and timing of admin-
miRNAs regulated by PI3K(p110␣) activity that were also istration may be warranted.
dysregulated in a cardiac disease setting (MI) (236). Manip-
ulation of miRNA expression (using miRNA inhibitors) has 4. Naturally occurring compounds acting via the
shown favorable therapeutic outcomes in patients with hep- PI3K-Akt pathway
atitis C virus (169) and is also being explored as a therapeu-
A number of naturally occurring compounds have been
tic approach for cancer (14). With optimization of cardiac
shown to possess cardioprotective properties, and many of
delivery systems, this approach may also represent a feasi-
these are reported to act, at least in part, via activation
ble avenue for the successful development of a miRNA-
of the PI3K-Akt pathway. The interpretation of a number of
based therapy for the treatment of heart failure. Inhibition
these studies has been based on experiments using the PI3K
of candidate PI3K-regulated miRNAs (miR-34, miR-34a,
inhibitors wortmannin or LY294002. Some caution needs
miR-652, or miR-154) using miRNA inhibitors was associ- to be taken when considering the conclusions of these re-
ated with reduced pathology and better cardiac function in ports because both inhibitors have the potential to inhibit/
mouse models of MI and/or pressure overload. Improved interact with other lipid kinases and proteins (17, 134).
outcomes were accompanied by the upregulation of target Naturally occurring compounds considered to act via PI3K-
genes implicated in regulating cardiac contractility, preserv- Akt signaling (e.g., flavonoids, icaritin, benfotiamine) are
ing angiogenesis and cell survival, and inhibiting fibrosis summarized in TABLE 3.
and inflammatory responses (27, 28, 30 –32) (FIGURE 10).
Additional reports have shown that miR-34a is also ele- 5. Ischemic conditioning and protection via
vated in the aged heart and other models of ischemia and PI3K-Akt signaling
that inhibition of miR-34a provides benefit via additional
target genes associated with protection against DNA dam- Ischemic conditioning (pre-, post-, remote-) has provided
age and telomere shortening as well as myocyte prolifera- some degree of protection in preclinical settings, and one of
tion and regeneration (43, 458). When inhibiting specific the key mechanisms implicated is PI3K-Akt signaling.
disease-regulated miRNAs, it is important to consider the However, activation of PI3K-Akt may occur downstream
potential regulation of additional miRNAs. Inhibition of of receptors other than IGF1R; mechanisms involving
miR-34 was shown to target secondary miRNAs (miR-31, eNOS and S6K1 have also been implicated. However, de-
let-7e) via transcription factor-miRNA regulatory net- spite positive results in rodent models, findings in the clinic
works, i.e., some miRNAs can regulate the expression of have been quite variable (154). There is also some evidence
transcription factors that regulate the expression of other that limb ischemic preconditioning may enhance athletic
miRNAs (316). performance, though further studies are required and the
mechanisms are unclear (379).
3. Strategies targeting Akt 6. Exercise-regulated miRNAs with proliferative
capacity
A) AKT GENE THERAPY. Akt1 is required for exercise-induced
physiological cardiac hypertrophy in mice (91) and is rec- Exercise training in mice has been shown to stimulate car-
ognized to play a central role in mediating cardioprotection diomyocyte proliferation (44). The injured adult heart has a
(155). Thus several groups have investigated the effects of limited capacity to regenerate, and the loss of cardiomyo-
Akt gene therapy in the adult rodent heart. Akt adenoviral cytes in the adult heart is a key component of most heart
gene transfer in the rat heart increased the contractility of failure pathologies. Thus therapeutics that can promote
isolated hearts (10 days post-delivery before significant car- cardiomyocyte proliferation may have a beneficial effect on
diac growth), most likely due to more systolic Ca2⫹ avail- cardiac repair and regeneration. A number of miRNAs have
ability (71). In addition, Akt gene therapy was also protec- been reported to promote cardiomyocyte proliferation in
tive in rodent models of ischemia. Akt gene therapy admin- the mouse heart, and some appear to be regulated in the
istered to mice 20 h before ischemia-reperfusion injury heart or circulation by exercise including miR-222 and
protected myocytes from apoptosis (128), and in vivo gene members of the miR-17–92 cluster (15, 243, 341, 380).
transfer of Akt in a rat model of cardiac ischemia-reperfu-
sion decreased the number of apoptotic cardiac myocytes The expression of miR-222 was shown to be increased in
and infarct size and improved cardiac function (260). the hearts of mice subjected to voluntary wheel running or

458 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

swim training (243). In addition, in humans miR-222 in- SERCA2a was delivered to the myocardium of patients
creased in the plasma of athletes after both acute and with heart failure using an AAV vector approach. Results
chronic exercise (15), and in heart failure patients following from phase 1 and 2 clinical trials of AAV-mediated gene
cardiopulmonary exercise testing (243). Targets of miR- transfer of SERCA2a in patients with heart failure demon-
222 including p27 and HIPK1 were shown to contribute to strated clinical benefits in patients receiving AAV-
the pro-proliferative effects of miR-222. While miR-222 SERCA2a (improvements in LV function/remodeling and 6
was shown to be required for exercise-induced cardiac min walking time) (170, 173, 470). However, results from a
growth, and transgenic overexpression of miR-222 pro- larger international clinical trial with AAV-SERCA2a in
tected the heart against cardiac dysfunction after ischemic patients with moderate-severe heart failure were not asso-
injury, an approach to pharmacologically increase miR-222 ciated with any benefits (139). Potential reasons for the
is yet to be described (243). negative findings were identified as suboptimal vector dose,
reduced transduction efficiency, and delivery method (139,
Two members of the miR-17–92 cluster (miR-19b and 342). However, despite the disappointing results from the
miR-20a) were shown to be elevated with exercise. miR- recent trial, the impetus for continuing to pursue gene ther-
19b expression was elevated in the hearts from rats sub- apy for heart failure remains strong (342). A number of
jected to swim training for 8 wk based on small RNA se- research groups and companies worldwide are designing
quencing (similar trend by qPCR) (341). Circulating levels second- or third-generation AAV vectors (e.g., chemical
of miR-20a were increased in healthy competitive athletes modifications such as PEGylation to make AAVs immuno-
after 90 days of sustained aerobic rowing exercise (15). logically inert), as well as investigating other delivery sys-
Cardiac-specific overexpression of the miR-17–92 cluster in tems (280, 342). Furthermore, promising results were iden-
mice induced cardiomyocyte proliferation in embryonic, tified in a small phase 2 clinical trial in heart failure patients
neonatal, and adult mouse hearts (66). Furthermore, over- with intracoronary gene transfer of adenylyl cyclase 6
expression of this cluster protected the mouse heart from (AC6; acts via regulating cAMP and SERCA2a). Improve-
MI-induced injury through increased cardiomyocyte prolif- ments were observed in ejection fraction at 4 wk post-de-
eration, decreased infarct size, and improved cardiac func- livery, and there was a decline in heart failure symptoms by
tion (FIGURE 11) (66). Another member of the miR-17–92 12 wk (150).
cluster, miR-17–3p, which contributes to exercise-induced
cardiac growth and regulates markers of myocyte prolifer- SERCA2a activity has also been targeted by the modifica-
ation, was also shown to have protective properties. In a tion of SUMO1 (required for preserving SERCA2a function
setting of ischemia-reperfusion injury, mice treated with a by SUMOylation) by either viral vector gene transfer (200,
miR-17–3p agomiR (which increased miR-17–3p expres- 413) or administration of a novel small molecule, N106
sion ~6-fold) displayed preserved cardiac function, attenu- (FIGURE 11) (201). Both approaches were able to improve
ation of cardiac apoptosis and fibrosis, and increased mark- cardiac function and increase SUMOlyation of SERCA2a in
ers of cardiomycoyte proliferation (380). animal models of heart failure.

Collectively, these studies suggest that targeting miRNAs


that are regulated in settings of protection (i.e., mimicking C. Gene Therapy Approaches Targeting the
exercise) represents a promising approach for the develop- Protective Actions of Exercise:
ment of a novel miRNA-based therapy for the failing heart. Challenges and Potential for Turning
Experimental Laboratory Findings Into
Clinical Therapeutics
B. Therapies Targeting Calcium Handling
A significant attraction of using gene therapy for the heart
As described in section IID1B, exercise is known to have to mimic the protective actions of exercise (e.g., by targeting
beneficial effects on calcium handling in the heart by in- PI3K-Akt signaling or calcium handling as described above)
creasing SERCA2a uptake/activity/protein (195, 197, 198, is the potential to more specifically or selectively target the
447). For example, exercise training in rats was shown to heart. However, although many preclinical studies have
improve contractility, increase calcium sensitivity, and nor- shown benefit of gene transfer in animal models of heart
malize SERCA2a protein levels after MI (446). Investiga- failure, clinical trials have generally failed to meet primary
tors have developed therapies that target SERCA2a directly efficacy end points (70, 139, 150). A key contributing factor
or target regulators/modifiers of SERCA2a such as small is the difficulty of transducing the human heart efficiently
ubiquitin-like modifier-1 (SUMO1) (FIGURE 11). Preclinical (147). Despite this, the recent favorable safety profile of
studies in small and large heart failure animal models have gene delivery vectors targeting the heart in patients allows
demonstrated improvement in cardiac function and remod- further progress in this field (324). Hajjar and Ishikawa
eling as a consequence of overexpression of SERCA2a using (147) have recently outlined three approaches to improve
adenoviral vectors (57, 193, 281). Following these success- cardiac gene delivery. One option is to increase gene dose of
ful preclinical studies, clinical trials were conducted and the vector; however, patients would have to be monitored

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 459


BERNARDO ET AL.

closely for an increased immune response and arrhythmias. anesthesia, more strict regulatory requirements, an in-
A second strategy being actively pursued is to develop a creased aversion to risk from pharmaceutical companies,
more efficient gene delivery method. However, this is usu- increasing clinical trial costs, inappropriate patient popula-
ally associated with a more invasive method of delivery tions selected (including sex differences), and unexpected
(e.g., retrograde coronary sinus delivery). Thus this ap- toxic effects (77, 207, 425, 432). An additional challenge
proach may be better suited to patients requiring a surgical facing the cardiovascular research community was recently
procedure, such as a valve replacement. Finally, the third highlighted, i.e., inadequate assessment of cardiovascular
approach is to use vectors in the clinic with a higher trans- functional measurements as laboratories with physiology
duction efficacy (e.g., AAV9), or a reengineered vector with expertise dwindle in favor of molecular-based laboratories
higher cardiac tropism. Both of these strategies have been (425).
used successfully in preclinical large animal models (168,
329). It is expected that some of these approaches will be Preclinical studies in which key regulators of exercise-in-
incorporated into upcoming clinical trials (147). duced protection have been targeted have been promising.
However, innovative strategies for specifically targeting the
V. OBSTACLES AND CHALLENGES heart or diseased myocardium may be required. Future
studies in large-animal models and clinical trials will be
required to assess how these approaches compare with cur-
A. Comprehensively Capturing the rent drugs prescribed as standard of care. In considering the
Mechanisms Responsible for Exercise- multitude of effects exercise has on numerous cell types and
Induced Cardiac Protection processes within the heart and other organs, the discovery
of a single target or “exercise pill” with the ability to reca-
Defining the full complement of molecular and cellular pitulate all the salutary effects of exercise is probably un-
changes (acute and chronic) associated with exercise is chal- likely. Given exercise represents an effective nonpharmaco-
lenging given exercise represents a dynamic stimulus affect- logical approach, the strategy of treating patients so they
ing numerous organs and systems in the body; during the are able to undertake some form of exercise, and subse-
exercise bout but also during the recovery phase. It is also quently prescribe pharmacological agents together with ex-
unclear whether mechanisms identified in the most com- ercise is worthy of further study in animal studies and hu-
mon exercise training models in animals (i.e., moderate mans. The combination of a ␤-blocker together with exer-
swim, treadmill, and voluntary free wheel protocols) reflect cise training in heart failure mice provided benefits above
mechanisms that may occur with other common forms of either intervention alone based on measures including sur-
exercise in humans (e.g., cycling, weight training). With vival, exercise tolerance, and cardiac contractility (424).
“lack of time” being a common reason provided for not However, in prescribing exercise as a therapeutic interven-
undertaking regular exercise (418), there has been increased tion, it is also important to consider the physical and psy-
interest and research into assessing the potential benefits of chological road blocks that deter people from undertaking
intermittent bursts of relatively intense exercise [e.g., high- exercise on a regular basis. This is particularly challenging
intensity training, sprint interval training (SIT)] (135). SIT for the aged. It remains unknown why some individuals
was associated with improved measures of cardiometabolic enjoy exercise and for others it is a chore. Further research
health (e.g., peak oxygen uptake and insulin sensitivity) should be undertaken to understand the effects of exercise
despite the lower exercise volume and reduced exercise time on the brain and behavioral patterns.
(135). Furthermore, positive short-term effects of interrupt-
ing prolonged periods of sitting with light-intensity physical 1. Species differences: mouse versus human
activity (e.g., standing, stepping) on cardiometabolic health
have been reported (111, 157). To date, the effects of acute
Genetic mouse models have proven a valuable tool for un-
versus chronic and low/moderate- versus high-intensity ex-
derstanding the mechanisms underlying exercise-induced
ercise on the cellular and molecular mechanisms in the heart
have not been studied in detail. cardiac protection. However, there are key similarities and
differences between species that must be considered when
assessing the potential of translating a finding from mice to
B. Few Drugs Translating From the humans. Mice and humans have a genome size that is sim-
Research Laboratory to the Clinic ilar (mice: 2,600 Mb, human: 3,000 Mb), a comparable
number of genes (~30, 000 in both), and many proteins
For most human diseases, the number of drugs being suc- show conserved function (229, 319). The mouse and hu-
cessfully translated to the clinic has been disappointing, and man heart are both four-chambered organs, and electrical
lower than anticipated in the last decade. There are many signals are propagated in a similar way. However, there are
reasons that have been attributed to this failure including also key differences including the heart size, heart rate, vas-
lack of reproducibility of basic science studies (e.g., lack of culature, and differences in the contribution of some con-
rigor, nonspecific antibodies), species differences, effects of tractile proteins (e.g., ␤-MHC predominates in the human

460 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

heart and ␣-MHC predominates in the mouse heart; sex ning behavior is different (105, 311). Following 8 wk of
difference is also apparent) (40, 229). exercise-induced physiological hypertrophy (voluntary
wheel running), female ER␣ KO mice ran less and did not
2. Sex differences in settings of cardiac health develop cardiac hypertrophy compared with wild-type lit-
and disease termates, whereas ER␣ KO male mice performed similar to
their wild-type littermates (105). In contrast, neither male
Cardiovascular research in both humans and animals has nor female ER␤ KO mice developed cardiac hypertrophy
been carried out predominately in males (1, 186, 228); how- compared with sedentary controls, despite similar running
ever, it is now well recognized that there are distinct differ- behavior (measured by distance ran) compared with wild-
ences between the male and female heart (23, 343). Sex type littermates. The authors concluded that exercise-in-
differences have been identified in cardiac size, cardiac func- duced cardiac hypertrophy depends on ER␤ in both sexes
tion, E-C coupling (AP, myocyte contraction), cardiac gene (105). It is thought that ER␤ mediates exercise-induced car-
expression, energy metabolism (glucose uptake, fatty acid diac hypertrophy in female mice by modulating Akt and
utilization), and the activation of signaling pathways (40, MAPK phosphorylation, but the underlying mechanism re-
89, 138, 303, 417). This is particularly true in settings of mains to be elucidated in male mice (105).
cardiac disease, and males and females respond differently
to cardiovascular drugs (35, 186, 245, 333). There is also A number of signaling mechanisms associated with exer-
increasing evidence to show exercise-induced cardiac re- cise-induced cardiac protection are likely to be different in
modeling differs in males and females in animals and hu- males and females. The IGF1-PI3K-Akt pathway that plays
mans, although larger studies with more power will be re- a key role for exercise-induced hypertrophy and protection
quired in humans (343). Early studies performed in rats has been shown to be higher in cardiac tissue from females
demonstrated that female rats displayed a greater cardiac compared with males (40, 60). Other studies have identified
hypertrophic response (increased heart/body weight ratio alterations in signaling in female and male hearts in re-
of 31%) compared with male rats (increased heart/body sponse to exercise including Akt, Hsp70, CaMK, p38-
weight ratio of 17%) following 8 wk of swim training (5 MAPK, ERK1, and S6 phosphorylation (105, 210, 277,
days/wk) (366, 367). In another study, treadmill running (5 320, 410). On the basis of animal and human studies, car-
days/wk for 8 wk) resulted in increased cardiac hypertrophy diac substrate availability and utilization also seem to differ
in both male and female rats, but only isolated hearts from in males and females in response to exercise. However, data
trained male rats showed enhanced cardiac performance are currently limited, and findings are quite variable. This is
(365). In mice, females developed more cardiac hypertro- particularly true in human studies, potentially explained by
phy in response to voluntary running and treadmill running differences in age and the type and duration of exercise
compared with males (87, 105, 124, 209, 210), but this activity performed (chronic/acute, aerobic/anaerobic) (124,
difference was eliminated when mice were placed on a ca- 162, 303, 323, 326, 392, 394, 399, 440). Collectively, these
sein-based soy-free diet (209). In addition, female mice ex- differences have implications for therapeutic approaches.
hibited enhanced wheel running performance (running fur- For instance, since components of the IGF1-PI3K-Akt path-
ther and for longer; independent of strain and age) (105, way are enhanced in the heart from females (40, 60), ap-
210), and female mice performed better than male mice in proaches that target this pathway are likely to have differ-
endurance and stress treadmill tests (209). ential therapeutic effects between sexes. As an example, we
recently demonstrated that a miRNA-based therapy that
The underlying mechanisms responsible for the sex-medi- targeted a PI3K-regulated miRNA (miRNA-34a) had dis-
ated differences in exercise-induced cardiac remodeling are tinct effects on global miRNA expression in male and fe-
not well understood, but contributing factors include sex male hearts, and this was associated with more efficacy in
hormones and the interaction of these hormones with mul- the female disease model (dilated cardiomyopathy) than the
tiple signaling pathways (e.g., IGF1-PI3K-Akt), metabo- male model (32).
lism, and miRNAs (40, 411). The effect of estrogen in the
sexually dimorphic cardiac response to exercise has been
investigated in genetic mouse models with chronic deletion VI. PERSPECTIVES AND CONCLUSIONS
of all estrogens [aromatase (Ar) KO mice which produce no
estrogens], and in mice lacking estrogen receptor ␣ (ER␣ Exercise has well-recognized beneficial effects on the heart
KO) or ER␤ (ER␤ KO). Following voluntary wheel running in settings of health and disease and represents one of the
(7 days and 21 days), Ar KO female mice ran significantly few interventions known to reverse cardiac remodeling and
less than wild-type females but developed equivalent car- improve heart function in heart failure patients. This has led
diac hypertrophy, suggesting that estrogens influence run- to intense interest in identifying the key mechanisms re-
ning distance but not cardiac growth in response to exercise sponsible for exercise-induced cardiac protection. Studies
(145). No differences were observed between male Ar KO examining the effects of perturbing individual genes glob-
and wild-type mice (145). Studies in ER KO models have ally or specifically in cardiac myocytes in mouse models
been varied and complicated in global KOs in which run- have provided significant insight into understanding the

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 461


BERNARDO ET AL.

critical molecular mechanisms responsible for exercise-in- 6. Ahmetov II, Egorova ES, Gabdrakhmanova LJ, Fedotovskaya ON. Genes and Athletic
Performance: An Update. Med Sport Sci 61: 41–54, 2016. doi:10.1159/000445240.
duced cardiac hypertrophy. However, our understanding of
the effects of exercise and genes regulated by exercise on cell 7. Alleman RJ, Katunga LA, Nelson MA, Brown DA, Anderson EJ. The “Goldilocks Zone”
types other than myocytes, organelles, and processes [e.g., from a redox perspective–adaptive vs. deleterious responses to oxidative stress in
striated muscle. Front Physiol 5: 358, 2014. doi:10.3389/fphys.2014.00358.
E-C coupling, mitochondrial adaptations, cellular stress re-
sponses (ER stress response, autophagy, proteasome-ubiq- 8. Alleman RJ, Tsang AM, Ryan TE, Patteson DJ, McClung JM, Spangenburg EE, Shaikh
SR, Neufer PD, Brown DA. Exercise-induced protection against reperfusion arrhyth-
uitin system, DNA damage response), and extracellular ma- mia involves stabilization of mitochondrial energetics. Am J Physiol Heart Circ Physiol
trix] is relatively limited. Research within these understud- 310: H1360 –H1370, 2016. doi:10.1152/ajpheart.00858.2015.
ied areas, together with the ability to profile the genome,
9. Allen NE, Davey GK, Key TJ, Zhang S, Narod SA. Serum insulin-like growth factor I
transcriptome, proteome, and metabolome in far greater (IGF-I) concentration in men is not associated with the cytosine-adenosine repeat
depth than ever before, opens up new opportunities to more polymorphism of the IGF-I gene. Cancer Epidemiol Biomarkers Prev 11: 319 –320, 2002.
comprehensively define the signaling and regulatory aspects 10. Anker SD, Volterrani M, Pflaum CD, Strasburger CJ, Osterziel KJ, Doehner W, Ranke
of cell/organelle functions that underpin the protective MB, Poole-Wilson PA, Giustina A, Dietz R, Coats AJ. Acquired growth hormone
properties of exercise. An increased knowledge of exercise- resistance in patients with chronic heart failure: implications for therapy with growth
induced changes to the circulation will also be essential for hormone. J Am Coll Cardiol 38: 443– 452, 2001. doi:10.1016/S0735-1097(01)01385-7.

understanding how the heart interacts with others organs 11. Ascensão A, Lumini-Oliveira J, Machado NG, Ferreira RM, Gonçalves IO, Moreira AC,
and systems in response to exercise. Collectively, this has Marques F, Sardão VA, Oliveira PJ, Magalhães J. Acute exercise protects against
calcium-induced cardiac mitochondrial permeability transition pore opening in doxo-
the potential to lead to the discovery of new drug targets for rubicin-treated rats. Clin Sci (Lond) 120: 37– 49, 2011. doi:10.1042/CS20100254.
heart disease.
12. Ascensão A, Lumini-Oliveira J, Oliveira PJ, Magalhães J. Mitochondria as a target for
exercise-induced cardioprotection. Curr Drug Targets 12: 860 – 871, 2011. doi:10.
ACKNOWLEDGMENTS 2174/138945011795529001.

13. Backs J, Worst BC, Lehmann LH, Patrick DM, Jebessa Z, Kreusser MM, Sun Q, Chen
Address for reprint requests and other correspondence: J. Mc- L, Heft C, Katus HA, Olson EN. Selective repression of MEF2 activity by PKA-
Mullen, Baker Heart and Diabetes Institute, PO Box 6492, Mel- dependent proteolysis of HDAC4. J Cell Biol 195: 403– 415, 2011. doi:10.1083/jcb.
bourne 3004, Australia (e-mail: julie.mcmullen@baker.edu.au). 201105063.

14. Bader AG. miR-34 –a microRNA replacement therapy is headed to the clinic. Front
GRANTS Genet 3: 120, 2012. doi:10.3389/fgene.2012.00120.

15. Baggish AL, Hale A, Weiner RB, Lewis GD, Systrom D, Wang F, Wang TJ, Chan SY.
J. R. McMullen is a National Health and Medical Research Dynamic regulation of circulating microRNA during acute exhaustive exercise and
sustained aerobic exercise training. J Physiol 589: 3983–3994, 2011. doi:10.1113/
Council Senior Research Fellow (1078985). B. C. Bernardo jphysiol.2011.213363.
is supported by an Alice Baker and Eleanor Shaw Fellow-
ship (Baker Foundation, Melbourne, Australia), and K. L. 16. Baggish AL, Park J, Min PK, Isaacs S, Parker BA, Thompson PD, Troyanos C,
D’Hemecourt P, Dyer S, Thiel M, Hale A, Chan SY. Rapid upregulation and clearance
Weeks is supported by a National Heart Foundation of of distinct circulating microRNAs after prolonged aerobic exercise. J Appl Physiol
Australia Postdoctoral Fellowship (O12M6802). (1985) 116: 522–531, 2014. doi:10.1152/japplphysiol.01141.2013.

17. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, Klevernic I, Arthur JS,
Alessi DR, Cohen P. The selectivity of protein kinase inhibitors: a further update.
DISCLOSURES Biochem J 408: 297–315, 2007. doi:10.1042/BJ20070797.

18. Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term
No conflicts of interest, financial or otherwise, are declared regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev
by the authors. 89: 481–534, 2009. doi:10.1152/physrev.00042.2007.

19. Bei Y, Fu S, Chen X, Chen M, Zhou Q, Yu P, Yao J, Wang H, Che L, Xu J, Xiao J.


Cardiac cell proliferation is not necessary for exercise-induced cardiac growth but
REFERENCES required for its protection against ischaemia/reperfusion injury. J Cell Mol Med 21:
1648 –1655, 2017. doi:10.1111/jcmm.13078.
1. Editorial: Putting gender on the agenda. Nature 465: 665, 2010. doi:10.1038/465665a.
20. Bei Y, Xu T, Lv D, Yu P, Xu J, Che L, Das A, Tigges J, Toxavidis V, Ghiran I, Shah R, Li
2. Abel ED, Doenst T. Mitochondrial adaptations to physiological vs. pathological cardiac Y, Zhang Y, Das S, Xiao J. Exercise-induced circulating extracellular vesicles protect
hypertrophy. Cardiovasc Res 90: 234 –242, 2011. doi:10.1093/cvr/cvr015. against cardiac ischemia-reperfusion injury. Basic Res Cardiol 112: 38, 2017. doi:10.
1007/s00395-017-0628-z.
3. Adamopoulos S, Parissis J, Karatzas D, Kroupis C, Georgiadis M, Karavolias G, Para-
skevaidis J, Koniavitou K, Coats AJ, Kremastinos DT. Physical training modulates 21. Bekhite MM, Finkensieper A, Binas S, Müller J, Wetzker R, Figulla HR, Sauer H,
proinflammatory cytokines and the soluble Fas/soluble Fas ligand system in patients Wartenberg M. VEGF-mediated PI3K class IA and PKC signaling in cardiomyogenesis
with chronic heart failure. J Am Coll Cardiol 39: 653– 663, 2002. doi:10.1016/S0735- and vasculogenesis of mouse embryonic stem cells. J Cell Sci 124: 1819 –1830, 2011.
1097(01)01795-8. doi:10.1242/jcs.077594.

4. Adams V, Doring C, Schuler G. Impact of physical exercise on alterations in the 22. Belke DD. Swim-exercised mice show a decreased level of protein O-GlcNAcylation
skeletal muscle in patients with chronic heart failure. Front Biosci 13: 302–311, 2008. and expression of O-GlcNAc transferase in heart. J Appl Physiol (1985) 111: 157–162,
doi:10.2741/2680. 2011. doi:10.1152/japplphysiol.00147.2011.

5. Adams V, Linke A, Gielen S, Erbs S, Hambrecht R, Schuler G. Modulation of Murf-1 23. Bell JR, Bernasochi GB, Varma U, Raaijmakers AJ, Delbridge LM. Sex and sex hor-
and MAFbx expression in the myocardium by physical exercise training. Eur J Cardio- mones in cardiac stress–mechanistic insights. J Steroid Biochem Mol Biol 137: 124 –135,
vasc Prev Rehabil 15: 293–299, 2008. doi:10.1097/HJR.0b013e3282f3ec43. 2013. doi:10.1016/j.jsbmb.2013.05.015.

462 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

24. Bennett CE, Johnsen VL, Shearer J, Belke DD. Exercise training mitigates aberrant 42. Boluyt MO, Brevick JL, Rogers DS, Randall MJ, Scalia AF, Li ZB. Changes in the rat
cardiac protein O-GlcNAcylation in streptozotocin-induced diabetic mice. Life Sci 92: heart proteome induced by exercise training: Increased abundance of heat shock
657– 663, 2013. doi:10.1016/j.lfs.2012.09.007. protein hsp20. Proteomics 6: 3154 –3169, 2006. doi:10.1002/pmic.200401356.

25. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, 43. Boon RA, Iekushi K, Lechner S, Seeger T, Fischer A, Heydt S, Kaluza D, Tréguer K,
Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisén J. Evidence for cardio- Carmona G, Bonauer A, Horrevoets AJ, Didier N, Girmatsion Z, Biliczki P, Ehrlich JR,
myocyte renewal in humans. Science 324: 98 –102, 2009. doi:10.1126/science. Katus HA, Müller OJ, Potente M, Zeiher AM, Hermeking H, Dimmeler S. MicroRNA-
1164680. 34a regulates cardiac ageing and function. Nature 495: 107–110, 2013. doi:10.1038/
nature11919.
26. Bernardo BC, Charchar FJ, Lin RC, McMullen JR. A microRNA guide for clinicians and
basic scientists: background and experimental techniques. Heart Lung Circ 21: 131– 44. Boström P, Mann N, Wu J, Quintero PA, Plovie ER, Panáková D, Gupta RK, Xiao C,
142, 2012. doi:10.1016/j.hlc.2011.11.002. MacRae CA, Rosenzweig A, Spiegelman BM. C/EBP␤ controls exercise-induced car-
diac growth and protects against pathological cardiac remodeling. Cell 143: 1072–
27. Bernardo BC, Gao XM, Tham YK, Kiriazis H, Winbanks CE, Ooi JY, Boey EJ, Obad S, 1083, 2010. doi:10.1016/j.cell.2010.11.036.
Kauppinen S, Gregorevic P, Du XJ, Lin RC, McMullen JR. Silencing of miR-34a atten-
uates cardiac dysfunction in a setting of moderate, but not severe, hypertrophic 45. Bourdier G, Flore P, Sanchez H, Pepin JL, Belaidi E, Arnaud C. High-intensity training
cardiomyopathy. PLoS One 9: e90337, 2014. doi:10.1371/journal.pone.0090337. reduces intermittent hypoxia-induced ER stress and myocardial infarct size. Am J
Physiol Heart Circ Physiol 310: H279 –H289, 2016. doi:10.1152/ajpheart.00448.2015.
28. Bernardo BC, Gao XM, Winbanks CE, Boey EJ, Tham YK, Kiriazis H, Gregorevic P,
Obad S, Kauppinen S, Du XJ, Lin RC, McMullen JR. Therapeutic inhibition of the 46. Bozi LH, Jannig PR, Rolim N, Voltarelli VA, Dourado PM, Wisløff U, Brum PC. Aerobic
miR-34 family attenuates pathological cardiac remodeling and improves heart func- exercise training rescues cardiac protein quality control and blunts endoplasmic re-
tion. Proc Natl Acad Sci USA 109: 17615–17620, 2012. doi:10.1073/pnas.1206432109. ticulum stress in heart failure rats. J Cell Mol Med 20: 2208 –2212, 2016. doi:10.1111/
jcmm.12894.
29. Bernardo BC, McMullen JR. Molecular Aspects of Exercise-induced Cardiac Remod-
eling. Cardiol Clin 34: 515–530, 2016. doi:10.1016/j.ccl.2016.06.002. 47. Brehme M, Voisine C. Model systems of protein-misfolding diseases reveal chaperone
modifiers of proteotoxicity. Dis Model Mech 9: 823– 838, 2016. doi:10.1242/dmm.
30. Bernardo BC, Nguyen SS, Gao XM, Tham YK, Ooi JY, Patterson NL, Kiriazis H, Su Y, 024703.
Thomas CJ, Lin RC, Du XJ, McMullen JR. Inhibition of miR-154 Protects Against
Cardiac Dysfunction and Fibrosis in a Mouse Model of Pressure Overload. Sci Rep 6: 48. Brenner IK, Natale VM, Vasiliou P, Moldoveanu AI, Shek PN, Shephard RJ. Impact of
22442, 2016. doi:10.1038/srep22442. three different types of exercise on components of the inflammatory response. Eur J
Appl Physiol Occup Physiol 80: 452– 460, 1999. doi:10.1007/s004210050617.
31. Bernardo BC, Nguyen SS, Winbanks CE, Gao XM, Boey EJ, Tham YK, Kiriazis H, Ooi
JY, Porrello ER, Igoor S, Thomas CJ, Gregorevic P, Lin RC, Du XJ, McMullen JR. 49. Brouillette J, Clark RB, Giles WR, Fiset C. Functional properties of K⫹ currents in adult
Therapeutic silencing of miR-652 restores heart function and attenuates adverse mouse ventricular myocytes. J Physiol 559: 777–798, 2004. doi:10.1113/jphysiol.2004.
remodeling in a setting of established pathological hypertrophy. FASEB J 28: 5097– 063446.
5110, 2014. doi:10.1096/fj.14-253856.
50. Brown MD. Exercise and coronary vascular remodelling in the healthy heart. Exp
32. Bernardo BC, Ooi JYY, Matsumoto A, Tham YK, Singla S, Kiriazis H, Patterson NL, Physiol 88: 645– 658, 2003. doi:10.1113/eph8802618.
Sadoshima J, Obad S, Lin RCY, McMullen JR. Sex differences in response to miRNA-
34a therapy in mouse models of cardiac disease: identification of sex-, disease- and 51. Brown WM, Davison GW, McClean CM, Murphy MH. A Systematic Review of the
treatment-regulated miRNAs. J Physiol 594: 5959 –5974, 2016. doi:10.1113/ Acute Effects of Exercise on Immune and Inflammatory Indices in Untrained Adults.
JP272512. Sports Med Open 1: 35, 2015. doi:10.1186/s40798-015-0032-x.

33. Bernardo BC, Ooi JYY, McMullen JR. The yin and yang of adaptive and maladaptive 52. Brutsaert DL. Cardiac endothelial-myocardial signaling: its role in cardiac growth,
processes in heart failure. Drug Discov Today Ther Strateg 9: e163– e172, 2012. doi:10. contractile performance, and rhythmicity. Physiol Rev 83: 59 –115, 2003. doi:10.1152/
1016/j.ddstr.2013.10.001. physrev.00017.2002.

34. Bernardo BC, Weeks KL, Patterson NL, McMullen JR. HSP70: therapeutic potential in 53. Burelle Y, Wambolt RB, Grist M, Parsons HL, Chow JC, Antler C, Bonen A, Keller A,
acute and chronic cardiac disease settings. Future Med Chem 8: 2177–2183, 2016. Dunaway GA, Popov KM, Hochachka PW, Allard MF. Regular exercise is associated
doi:10.4155/fmc-2016-0192. with a protective metabolic phenotype in the rat heart. Am J Physiol Heart Circ Physiol
287: H1055–H1063, 2004. doi:10.1152/ajpheart.00925.2003.
35. Bernardo BC, Weeks KL, Pretorius L, McMullen JR. Molecular distinction between
physiological and pathological cardiac hypertrophy: experimental findings and thera- 54. Burgess ML, Buggy J, Price RL, Abel FL, Terracio L, Samarel AM, Borg TK. Exercise-
peutic strategies. Pharmacol Ther 128: 191–227, 2010. doi:10.1016/j.pharmthera. and hypertension-induced collagen changes are related to left ventricular function in
2010.04.005. rat hearts. Am J Physiol Heart Circ Physiol 270: H151–H159, 1996.

36. Bers DM. Cardiac excitation-contraction coupling. Nature 415: 198 –205, 2002. doi: 55. Burniston JG. Adaptation of the rat cardiac proteome in response to intensity-con-
10.1038/415198a. trolled endurance exercise. Proteomics 9: 106 –115, 2009. doi:10.1002/pmic.
200800268.
37. Bezzerides VJ, Platt C, Lerchenmüller C, Paruchuri K, Oh NL, Xiao C, Cao Y, Mann
N, Spiegelman BM, Rosenzweig A. CITED4 induces physiologic hypertrophy and 56. Bye A, Langaas M, Høydal MA, Kemi OJ, Heinrich G, Koch LG, Britton SL, Najjar SM,
promotes functional recovery after ischemic injury. JCI Insight 1: e85904, 2016. doi: Ellingsen Ø, Wisløff U. Aerobic capacity-dependent differences in cardiac gene ex-
10.1172/jci.insight.85904. pression. Physiol Genomics 33: 100 –109, 2008. doi:10.1152/physiolgenomics.00269.
2007.
38. Bing RJ. Myocardial metabolism. Circulation 12: 635– 647, 1955. doi:10.1161/01.CIR.
12.4.635. 57. Byrne MJ, Power JM, Preovolos A, Mariani JA, Hajjar RJ, Kaye DM. Recirculating
cardiac delivery of AAV2/1SERCA2a improves myocardial function in an experimental
39. Blair SN, Kohl HW III, Paffenbarger RS, Jr, Clark DG, Cooper KH, Gibbons LW. model of heart failure in large animals. Gene Ther 15: 1550 –1557, 2008. doi:10.1038/
Physical fitness and all-cause mortality. A prospective study of healthy men and gt.2008.120.
women. JAMA 262: 2395–2401, 1989. doi:10.1001/jama.1989.03430170057028.
58. Calvert JW, Condit ME, Aragón JP, Nicholson CK, Moody BF, Hood RL, Sindler AL,
40. Blenck CL, Harvey PA, Reckelhoff JF, Leinwand LA. The Importance of Biological Sex Gundewar S, Seals DR, Barouch LA, Lefer DJ. Exercise protects against myocardial
and Estrogen in Rodent Models of Cardiovascular Health and Disease. Circ Res 118: ischemia-reperfusion injury via stimulation of ␤(3)-adrenergic receptors and in-
1294 –1312, 2016. doi:10.1161/CIRCRESAHA.116.307509. creased nitric oxide signaling: role of nitrite and nitrosothiols. Circ Res 108: 1448 –
1458, 2011. doi:10.1161/CIRCRESAHA.111.241117.
41. Bleumink GS, Schut AF, Sturkenboom MC, Janssen JA, Witteman JC, van Duijn CM,
Hofman A, Stricker BH. A promoter polymorphism of the insulin-like growth factor-I 59. Calvert JW, Lefer DJ. Role of ␤-adrenergic receptors and nitric oxide signaling in
gene is associated with left ventricular hypertrophy. Heart 91: 239 –240, 2005. doi: exercise-mediated cardioprotection. Physiology (Bethesda) 28: 216 –224, 2013. doi:
10.1136/hrt.2003.019778. 10.1152/physiol.00011.2013.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 463


BERNARDO ET AL.

60. Camper-Kirby D, Welch S, Walker A, Shiraishi I, Setchell KD, Schaefer E, Kajstura J, levels in patients with chronic heart failure and coronary artery disease. Eur Heart J 23:
Anversa P, Sussman MA. Myocardial Akt activation and gender: increased nuclear 1854 –1860, 2002. doi:10.1053/euhj.2002.3239.
activity in females versus males. Circ Res 88: 1020 –1027, 2001. doi:10.1161/hh1001.
090858. 77. Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, Pangalos MN.
Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional
61. Carè A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang ML, Segnalini P, Gu framework. Nat Rev Drug Discov 13: 419 – 431, 2014. doi:10.1038/nrd4309.
Y, Dalton ND, Elia L, Latronico MV, Høydal M, Autore C, Russo MA, Dorn GW II,
Ellingsen O, Ruiz-Lozano P, Peterson KL, Croce CM, Peschle C, Condorelli G. Mi- 78. Couchonnal LF, Anderson ME. The role of calmodulin kinase II in myocardial physi-
croRNA-133 controls cardiac hypertrophy. Nat Med 13: 613– 618, 2007. doi:10. ology and disease. Physiology (Bethesda) 23: 151–159, 2008. doi:10.1152/physiol.
1038/nm1582. 00043.2007.

62. Carneiro-Júnior MA, Prímola-Gomes TN, Quintão-Júnior JF, Drummond LR, 79. Cox EJ, Marsh SA. Exercise and diabetes have opposite effects on the assembly and
Lavorato VN, Drummond FR, Felix LB, Oliveira EM, Cruz JS, Natali AJ, Mill JG. O-GlcNAc modification of the mSin3A/HDAC1/2 complex in the heart. Cardiovasc
Regional effects of low-intensity endurance training on structural and mechanical Diabetol 12: 101, 2013. doi:10.1186/1475-2840-12-101.
properties of rat ventricular myocytes. J Appl Physiol (1985) 115: 107–115, 2013.
doi:10.1152/japplphysiol.00041.2013. 80. Creemers EE, Tijsen AJ, Pinto YM. Circulating microRNAs: novel biomarkers and
extracellular communicators in cardiovascular disease? Circ Res 110: 483– 495, 2012.
63. Carneiro-Júnior MA, Quintão-Júnior JF, Drummond LR, Lavorato VN, Drummond doi:10.1161/CIRCRESAHA.111.247452.
FR, da Cunha DN, Amadeu MA, Felix LB, de Oliveira EM, Cruz JS, Prímola-Gomes
TN, Mill JG, Natali AJ. The benefits of endurance training in cardiomyocyte function in 81. Crul T, Toth N, Piotto S, Literati-Nagy P, Tory K, Haldimann P, Kalmar B, Greensmith
hypertensive rats are reversed within four weeks of detraining. J Mol Cell Cardiol 57: L, Torok Z, Balogh G, Gombos I, Campana F, Concilio S, Gallyas F, Nagy G, Berente
119 –128, 2013. doi:10.1016/j.yjmcc.2013.01.013. Z, Gungor B, Peter M, Glatz A, Hunya A, Literati-Nagy Z, Vigh L, Jr, Hoogstra-
Berends F, Heeres A, Kuipers I, Loen L, Seerden JP, Zhang D, Meijering RA, Henning
64. Chaturvedi P, Kalani A, Medina I, Familtseva A, Tyagi SC. Cardiosome mediated RH, Brundel BJ, Kampinga HH, Koranyi L, Szilvassy Z, Mandl J, Sumegi B, Febbraio
regulation of MMP9 in diabetic heart: role of mir29b and mir455 in exercise. J Cell Mol MA, Horvath I, Hooper PL, Vigh L. Hydroximic acid derivatives: pleiotropic HSP
Med 19: 2153–2161, 2015. doi:10.1111/jcmm.12589. co-inducers restoring homeostasis and robustness. Curr Pharm Des 19: 309 –346,
2013. doi:10.2174/138161213804143716.
65. Chen CY, Hsu HC, Lee BC, Lin HJ, Chen YH, Huang HC, Ho YL, Chen MF. Exercise
training improves cardiac function in infarcted rabbits: involvement of autophagic 82. Cutler MJ, Jeyaraj D, Rosenbaum DS. Cardiac electrical remodeling in health and
function and fatty acid utilization. Eur J Heart Fail 12: 323–330, 2010. doi:10.1093/ disease. Trends Pharmacol Sci 32: 174 –180, 2011. doi:10.1016/j.tips.2010.12.001.
eurjhf/hfq028.
83. Czarkowska-Paczek B, Bartlomiejczyk I, Przybylski J. The serum levels of growth
66. Chen J, Huang ZP, Seok HY, Ding J, Kataoka M, Zhang Z, Hu X, Wang G, Lin Z, Wang factors: PDGF, TGF-beta and VEGF are increased after strenuous physical exercise. J
S, Pu WT, Liao R, Wang DZ. mir-17–92 cluster is required for and sufficient to induce Physiol Pharmacol 57: 189 –197, 2006.
cardiomyocyte proliferation in postnatal and adult hearts. Circ Res 112: 1557–1566,
2013. doi:10.1161/CIRCRESAHA.112.300658. 84. Da Silva ND, Jr, Fernandes T, Soci UP, Monteiro AW, Phillips MI, DE Oliveira EM.
Swimming training in rats increases cardiac MicroRNA-126 expression and angiogen-
67. Chicco AJ, Schneider CM, Hayward R. Voluntary exercise protects against acute esis. Med Sci Sports Exerc 44: 1453–1462, 2012. doi:10.1249/MSS.
doxorubicin cardiotoxicity in the isolated perfused rat heart. Am J Physiol Regul Integr
0b013e31824e8a36.
Comp Physiol 289: R424 –R431, 2005. doi:10.1152/ajpregu.00636.2004.
85. Davis RT III, Simon JN, Utter M, Mungai P, Alvarez MG, Chowdhury SA, Heydemann
68. Chomistek AK, Henschel B, Eliassen AH, Mukamal KJ, Rimm EB. Frequency, Type,
A, Ke Y, Wolska BM, Solaro RJ. Knockout of p21-activated kinase-1 attenuates exer-
and Volume of Leisure-Time Physical Activity and Risk of Coronary Heart Disease in
cise-induced cardiac remodelling through altered calcineurin signalling. Cardiovasc Res
Young Women. Circulation 134: 290 –299, 2016. doi:10.1161/CIRCULATIONAHA.
108: 335–347, 2015. doi:10.1093/cvr/cvv234.
116.021516.
86. Day SM. The ubiquitin proteasome system in human cardiomyopathies and heart
69. Chung E, Heimiller J, Leinwand LA. Distinct cardiac transcriptional profiles defining
failure. Am J Physiol Heart Circ Physiol 304: H1283–H1293, 2013. doi:10.1152/
pregnancy and exercise. PLoS One 7: e42297, 2012. doi:10.1371/journal.pone.
ajpheart.00249.2012.
0042297.
87. De Bono JP, Adlam D, Paterson DJ, Channon KM. Novel quantitative phenotypes of
70. Chung ES, Miller L, Patel AN, Anderson RD, Mendelsohn FO, Traverse J, Silver KH,
exercise training in mouse models. Am J Physiol Regul Integr Comp Physiol 290: R926 –
Shin J, Ewald G, Farr MJ, Anwaruddin S, Plat F, Fisher SJ, AuWerter AT, Pastore JM,
R934, 2006. doi:10.1152/ajpregu.00694.2005.
Aras R, Penn MS. Changes in ventricular remodelling and clinical status during the year
following a single administration of stromal cell-derived factor-1 non-viral gene ther-
88. De Maeyer C, Beckers P, Vrints CJ, Conraads VM. Exercise training in chronic heart
apy in chronic ischaemic heart failure patients: the STOP-HF randomized Phase II
failure. Ther Adv Chronic Dis 4: 105–117, 2013. doi:10.1177/2040622313480382.
trial. Eur Heart J 36: 2228 –2238, 2015. doi:10.1093/eurheartj/ehv254.
89. De Simone G, Devereux RB, Daniels SR, Meyer RA. Gender differences in left ven-
71. Cittadini A, Monti MG, Iaccarino G, Di Rella F, Tsichlis PN, Di Gianni A, Strömer H,
tricular growth. Hypertension 26: 979 –983, 1995. doi:10.1161/01.HYP.26.6.979.
Sorriento D, Peschle C, Trimarco B, Saccà L, Condorelli G. Adenoviral gene transfer
of Akt enhances myocardial contractility and intracellular calcium handling. Gene Ther 90. De Waard MC, van Haperen R, Soullié T, Tempel D, de Crom R, Duncker DJ.
13: 8 –19, 2006. doi:10.1038/sj.gt.3302589. Beneficial effects of exercise training after myocardial infarction require full eNOS
expression. J Mol Cell Cardiol 48: 1041–1049, 2010. doi:10.1016/j.yjmcc.2010.02.
72. Coats AJ. Exercise training in heart failure. Curr Control Trials Cardiovasc Med 1: 155–
005.
160, 2000. doi:10.1186/CVM-1-3-155.
91. DeBosch B, Treskov I, Lupu TS, Weinheimer C, Kovacs A, Courtois M, Muslin AJ.
73. Colao A, Marzullo P, Di Somma C, Lombardi G. Growth hormone and the heart. Clin
Akt1 is required for physiological cardiac growth. Circulation 113: 2097–2104, 2006.
Endocrinol (Oxf) 54: 137–154, 2001. doi:10.1046/j.1365-2265.2001.01218.x.
doi:10.1161/CIRCULATIONAHA.105.595231.
74. Cole CR, Blackstone EH, Pashkow FJ, Snader CE, Lauer MS. Heart-rate recovery
92. Delaughter MC, Taffet GE, Fiorotto ML, Entman ML, Schwartz RJ. Local insulin-like
immediately after exercise as a predictor of mortality. N Engl J Med 341: 1351–1357,
1999. doi:10.1056/NEJM199910283411804. growth factor I expression induces physiologic, then pathologic, cardiac hypertrophy
in transgenic mice. FASEB J 13: 1923–1929, 1999.
75. Cole CR, Foody JM, Blackstone EH, Lauer MS. Heart rate recovery after submaximal
exercise testing as a predictor of mortality in a cardiovascularly healthy cohort. Ann 93. DeMarco VG, Johnson MS, Ma L, Pulakat L, Mugerfeld I, Hayden MR, Garro M, Knight
Intern Med 132: 552–555, 2000. doi:10.7326/0003-4819-132-7-200004040-00007. W, Britton SL, Koch LG, Sowers JR. Overweight female rats selectively breed for low
aerobic capacity exhibit increased myocardial fibrosis and diastolic dysfunction. Am J
76. Conraads VM, Beckers P, Bosmans J, De Clerck LS, Stevens WJ, Vrints CJ, Brutsaert Physiol Heart Circ Physiol 302: H1667–H1682, 2012. doi:10.1152/ajpheart.01027.
DL. Combined endurance/resistance training reduces plasma TNF-alpha receptor 2011.

464 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

94. Demirel HA, Powers SK, Zergeroglu MA, Shanely RA, Hamilton K, Coombes J, Naito 114. Evans M, Cogan KE, Egan B. Metabolism of ketone bodies during exercise and train-
H. Short-term exercise improves myocardial tolerance to in vivo ischemia-reperfu- ing: physiological basis for exogenous supplementation. J Physiol 595: 2857–2871,
sion in the rat. J Appl Physiol (1985) 91: 2205–2212, 2001. 2017. doi:10.1113/JP273185.

95. Denduluri SK, Idowu O, Wang Z, Liao Z, Yan Z, Mohammed MK, Ye J, Wei Q, Wang 115. Fan G-C, Ren X, Qian J, Yuan Q, Nicolaou P, Wang Y, Jones WK, Chu G, Kranias EG.
J, Zhao L, Luu HH. Insulin-like growth factor (IGF) signaling in tumorigenesis and the Novel cardioprotective role of a small heat-shock protein, Hsp20, against ischemia/
development of cancer drug resistance. Genes Dis 2: 13–25, 2015. doi:10.1016/j. reperfusion injury. Circulation 111: 1792–1799, 2005. doi:10.1161/01.CIR.
gendis.2014.10.004. 0000160851.41872.C6.

96. Denham J, Marques FZ, O’Brien BJ, Charchar FJ. Exercise: putting action into our 116. Fazio S, Sabatini D, Capaldo B, Vigorito C, Giordano A, Guida R, Pardo F, Biondi B,
epigenome. Sports Med 44: 189 –209, 2014. doi:10.1007/s40279-013-0114-1. Saccà L. A preliminary study of growth hormone in the treatment of dilated cardio-
myopathy. N Engl J Med 334: 809 – 814, 1996. doi:10.1056/NEJM199603283341301.
97. Deten A, Volz HC, Briest W, Zimmer HG. Cardiac cytokine expression is upregulated
in the acute phase after myocardial infarction. Experimental studies in rats. Cardiovasc 117. Fernandes T, Baraúna VG, Negrão CE, Phillips MI, Oliveira EM. Aerobic exercise
Res 55: 329 –340, 2002. doi:10.1016/S0008-6363(02)00413-3. training promotes physiological cardiac remodeling involving a set of microRNAs.
Am J Physiol Heart Circ Physiol 309: H543–H552, 2015. doi:10.1152/ajpheart.
98. Di Bello V, Lattanzi F, Picano E, Talarico L, Caputo MT, Di Muro C, Santoro G, 00899.2014.
Lunardi M, Distante A, Giusti C. Left ventricular performance and ultrasonic myocar-
dial quantitative reflectivity in endurance senior athletes: an echocardiographic study. 118. Fernandes T, Hashimoto NY, Magalhães FC, Fernandes FB, Casarini DE, Carmona
Eur Heart J 14: 358 –363, 1993. doi:10.1093/eurheartj/14.3.358. AK, Krieger JE, Phillips MI, Oliveira EM. Aerobic exercise training-induced left
ventricular hypertrophy involves regulatory MicroRNAs, decreased angiotensin-
99. Dick SA, Epelman S. Chronic Heart Failure and Inflammation: What Do We Really converting enzyme-angiotensin II, and synergistic regulation of angiotensin-con-
Know? Circ Res 119: 159 –176, 2016. doi:10.1161/CIRCRESAHA.116.308030. verting enzyme 2-angiotensin (1–7). Hypertension 58: 182–189, 2011. doi:10.
1161/HYPERTENSIONAHA.110.168252.
100. Disatnik MH, Hwang S, Ferreira JC, Mochly-Rosen D. New therapeutics to modulate
mitochondrial dynamics and mitophagy in cardiac diseases. J Mol Med (Berl) 93: 279 – 119. Ferreira R, Moreira-Gonçalves D, Azevedo AL, Duarte JA, Amado F, Vitorino R.
287, 2015. doi:10.1007/s00109-015-1256-4. Unraveling the exercise-related proteome signature in heart. Basic Res Cardiol 110:
454, 2015. doi:10.1007/s00395-014-0454-5.
101. Dolinsky VW, Dyck JR. Role of AMP-activated protein kinase in healthy and diseased
hearts. Am J Physiol Heart Circ Physiol 291: H2557–H2569, 2006. doi:10.1152/ 120. Ferreira R, Vitorino R, Padrão AI, Espadas G, Mancuso FM, Moreira-Gonçalves D,
ajpheart.00329.2006. Castro-Sousa G, Henriques-Coelho T, Oliveira PA, Barros AS, Duarte JA, Sabidó E,
Amado F. Lifelong exercise training modulates cardiac mitochondrial phosphopro-
102. Doroudgar S, Glembotski CC. The cardiokine story unfolds: ischemic stress-induced teome in rats. J Proteome Res 13: 2045–2055, 2014. doi:10.1021/pr4011926.
protein secretion in the heart. Trends Mol Med 17: 207–214, 2011. doi:10.1016/j.
121. Fillmore N, Mori J, Lopaschuk GD. Mitochondrial fatty acid oxidation alterations in
molmed.2010.12.003.
heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br J Pharmacol
103. Drozdov I, Tsoka S, Ouzounis CA, Shah AM. Genome-wide expression patterns in 171: 2080 –2090, 2014. doi:10.1111/bph.12475.
physiological cardiac hypertrophy. BMC Genomics 11: 557, 2010. doi:10.1186/1471-
122. Fischer CP. Interleukin-6 in acute exercise and training: what is the biological rele-
2164-11-557.
vance? Exerc Immunol Rev 12: 6 –33, 2006.
104. Duncker DJ, Bache RJ. Regulation of coronary blood flow during exercise. Physiol Rev
123. Fiuza-Luces C, Garatachea N, Berger NA, Lucia A. Exercise is the real polypill. Phys-
88: 1009 –1086, 2008. doi:10.1152/physrev.00045.2006.
iology (Bethesda) 28: 330 –358, 2013. doi:10.1152/physiol.00019.2013.
105. Dworatzek E, Mahmoodzadeh S, Schubert C, Westphal C, Leber J, Kusch A, Kararigas
124. Foryst-Ludwig A, Kreissl MC, Sprang C, Thalke B, Böhm C, Benz V, Gürgen D,
G, Fliegner D, Moulin M, Ventura-Clapier R, Gustafsson JA, Davidson MM, Dragun D,
Dragun D, Schubert C, Mai K, Stawowy P, Spranger J, Regitz-Zagrosek V, Unger T,
Regitz-Zagrosek V. Sex differences in exercise-induced physiological myocardial hy-
Kintscher U. Sex differences in physiological cardiac hypertrophy are associated with
pertrophy are modulated by oestrogen receptor beta. Cardiovasc Res 102: 418 – 428, exercise-mediated changes in energy substrate availability. Am J Physiol Heart Circ
2014. doi:10.1093/cvr/cvu065. Physiol 301: H115–H122, 2011. doi:10.1152/ajpheart.01222.2010.
106. Echouffo-Tcheugui JB, Butler J, Yancy CW, Fonarow GC. Association of Physical 125. Frayling TM, Hattersley AT, McCarthy A, Holly J, Mitchell SM, Gloyn AL, Owen K,
Activity or Fitness With Incident Heart Failure: A Systematic Review and Meta-Anal- Davies D, Smith GD, Ben-Shlomo Y. A putative functional polymorphism in the IGF-I
ysis. Circ Heart Fail 8: 853– 861, 2015. doi:10.1161/CIRCHEARTFAILURE.115. gene: association studies with type 2 diabetes, adult height, glucose tolerance, and
002070. fetal growth in U.K. populations. Diabetes 51: 2313–2316, 2002. doi:10.2337/
diabetes.51.7.2313.
108. Eghbali M. Cardiac fibroblasts: function, regulation of gene expression, and pheno-
typic modulation. Basic Res Cardiol 87, Suppl 2: 183–189, 1992. 126. Frühbeis C, Helmig S, Tug S, Simon P, Krämer-Albers EM. Physical exercise induces
rapid release of small extracellular vesicles into the circulation. J Extracell Vesicles 4:
109. Eijsvogels TM, Fernandez AB, Thompson PD. Are There Deleterious Cardiac Effects
28239, 2015. doi:10.3402/jev.v4.28239.
of Acute and Chronic Endurance Exercise? Physiol Rev 96: 99 –125, 2016. doi:10.1152/
physrev.00029.2014. 127. Fruman DA, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nat
Rev Drug Discov 13: 140 –156, 2014. doi:10.1038/nrd4204.
110. Eisner DA, Dibb KM, Trafford AW. The mechanism and significance of the slow
changes of ventricular action potential duration following a change of heart rate. Exp 128. Fujio Y, Nguyen T, Wencker D, Kitsis RN, Walsh K. Akt promotes survival of cardi-
Physiol 94: 520 –528, 2009. doi:10.1113/expphysiol.2008.044008. omyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart.
Circulation 101: 660 – 667, 2000. doi:10.1161/01.CIR.101.6.660.
111. Ekelund U, Steene-Johannessen J, Brown WJ, Fagerland MW, Owen N, Powell KE,
Bauman A, Lee IM; Lancet Physical Activity Series 2 Executive Committe; Lancet 129. Galindo CL, Skinner MA, Errami M, Olson LD, Watson DA, Li J, McCormick JF,
Sedentary Behaviour Working Group. Does physical activity attenuate, or even elim- McIver LJ, Kumar NM, Pham TQ, Garner HR. Transcriptional profile of isoprot-
inate, the detrimental association of sitting time with mortality? A harmonised meta- erenol-induced cardiomyopathy and comparison to exercise-induced cardiac hy-
analysis of data from more than 1 million men and women. Lancet 388: 1302–1310, pertrophy and human cardiac failure. BMC Physiol 9: 23, 2009. doi:10.1186/1472-
2016. doi:10.1016/S0140-6736(16)30370-1. 6793-9-23.

112. Erickson JR, He BJ, Grumbach IM, Anderson ME. CaMKII in the cardiovascular system: 130. Gaur V, Connor T, Sanigorski A, Martin SD, Bruce CR, Henstridge DC, Bond ST,
sensing redox states. Physiol Rev 91: 889 –915, 2011. doi:10.1152/physrev.00018.2010. McEwen KA, Kerr-Bayles L, Ashton TD, Fleming C, Wu M, Pike Winer LS, Chen D,
Hudson GM, Schwabe JWR, Baar K, Febbraio MA, Gregorevic P, Pfeffer FM, Walder
113. Ertek S, Cicero A. Impact of physical activity on inflammation: effects on cardiovas- KR, Hargreaves M, McGee SL. Disruption of the Class IIa HDAC Corepressor Com-
cular disease risk and other inflammatory conditions. Arch Med Sci 8: 794 – 804, 2012. plex Increases Energy Expenditure and Lipid Oxidation. Cell Reports 16: 2802–2810,
doi:10.5114/aoms.2012.31614. 2016. doi:10.1016/j.celrep.2016.08.005.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 465


BERNARDO ET AL.

131. Geffken DF, Cushman M, Burke GL, Polak JF, Sakkinen PA, Tracy RP. Association 150. Hammond HK, Penny WF, Traverse JH, Henry TD, Watkins MW, Yancy CW, Sweis
between physical activity and markers of inflammation in a healthy elderly population. RN, Adler ED, Patel AN, Murray DR, Ross RS, Bhargava V, Maisel A, Barnard DD, Lai
Am J Epidemiol 153: 242–250, 2001. doi:10.1093/aje/153.3.242. NC, Dalton ND, Lee ML, Narayan SM, Blanchard DG, Gao MH. Intracoronary gene
transfer of adenylyl cyclase 6 in patients with heart failure: a randomized clinical trial.
132. Genth-Zotz S, Zotz R, Geil S, Voigtländer T, Meyer J, Darius H. Recombinant growth JAMA Cardiol 1: 163–171, 2016. doi:10.1001/jamacardio.2016.0008.
hormone therapy in patients with ischemic cardiomyopathy : effects on hemodynam-
ics, left ventricular function, and cardiopulmonary exercise capacity. Circulation 99: 151. Haram PM, Kemi OJ, Wisloff U. Adaptation of endothelium to exercise training:
18 –21, 1999. doi:10.1161/01.CIR.99.1.18. insights from experimental studies. Front Biosci 13: 336 –346, 2008. doi:10.2741/
2683.
133. Gertz EW, Wisneski JA, Stanley WC, Neese RA. Myocardial substrate utilization
during exercise in humans. Dual carbon-labeled carbohydrate isotope experiments. J 152. Harris MB, Starnes JW. Effects of body temperature during exercise training on
Clin Invest 82: 2017–2025, 1988. doi:10.1172/JCI113822. myocardial adaptations. Am J Physiol Heart Circ Physiol 280: H2271–H2280, 2001.

134. Gharbi SI, Zvelebil MJ, Shuttleworth SJ, Hancox T, Saghir N, Timms JF, Waterfield 153. Harrison BC, Roberts CR, Hood DB, Sweeney M, Gould JM, Bush EW, McKinsey TA.
MD. Exploring the specificity of the PI3K family inhibitor LY294002. Biochem J 404: The CRM1 nuclear export receptor controls pathological cardiac gene expression.
15–21, 2007. doi:10.1042/BJ20061489. Mol Cell Biol 24: 10636 –10649, 2004. doi:10.1128/MCB.24.24.10636-10649.2004.

135. Gillen JB, Martin BJ, MacInnis MJ, Skelly LE, Tarnopolsky MA, Gibala MJ. Twelve 154. Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, Engstrom
Weeks of Sprint Interval Training Improves Indices of Cardiometabolic Health Similar T, Ferdinandy P, Carbrera-Fuentes HA, Heusch G, Ibanez B, Iliodromitis EK, Inserte
to Traditional Endurance Training despite a Five-Fold Lower Exercise Volume and J, Jennings R, Kalia N, Kharbanda R, Lecour S, Marber M, Miura T, Ovize M, Perez-
Time Commitment. PLoS One 11: e0154075, 2016. doi:10.1371/journal.pone. Pinzon MA, Piper HM, Przyklenk K, Schmidt MR, Redington A, Ruiz-Meana M, Vilahur
0154075. G, Vinten-Johansen J, Yellon DM, Garcia-Dorado D. Ischaemic conditioning and tar-
geting reperfusion injury: a 30 year voyage of discovery. Basic Res Cardiol 111: 70,
136. Giusti B, Marini M, Rossi L, Lapini I, Magi A, Capalbo A, Lapalombella R, di Tullio S, 2016. doi:10.1007/s00395-016-0588-8.
Samaja M, Esposito F, Margonato V, Boddi M, Abbate R, Veicsteinas A. Gene expres-
sion profile of rat left ventricles reveals persisting changes following chronic mild 155. Hausenloy DJ, Yellon DM. Survival kinases in ischemic preconditioning and postcon-
exercise protocol: implications for cardioprotection. BMC Genomics 10: 342, 2009. ditioning. Cardiovasc Res 70: 240 –253, 2006. doi:10.1016/j.cardiores.2006.01.017.
doi:10.1186/1471-2164-10-342.
156. He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q,
137. Gordan R, Gwathmey JK, Xie LH. Autonomic and endocrine control of cardiovascular Korsmeyer S, Packer M, May HI, Hill JA, Virgin HW, Gilpin C, Xiao G, Bassel-Duby R,
function. World J Cardiol 7: 204 –214, 2015. doi:10.4330/wjc.v7.i4.204. Scherer PE, Levine B. Exercise-induced BCL2-regulated autophagy is required for
muscle glucose homeostasis. Nature 481: 511–515, 2012. doi:10.1038/nature10758.
138. Grant AO. Cardiac ion channels. Circ Arrhythm Electrophysiol 2: 185–194, 2009. doi:
10.1161/CIRCEP.108.789081. 157. Healy GN, Winkler EA, Owen N, Anuradha S, Dunstan DW. Replacing sitting time
with standing or stepping: associations with cardio-metabolic risk biomarkers. Eur
139. Greenberg B, Butler J, Felker GM, Ponikowski P, Voors AA, Desai AS, Barnard D, Heart J 36: 2643–2649, 2015. doi:10.1093/eurheartj/ehv308.
Bouchard A, Jaski B, Lyon AR, Pogoda JM, Rudy JJ, Zsebo KM. Calcium upregulation by
percutaneous administration of gene therapy in patients with cardiac disease (CUPID 158. Heinonen I, Kudomi N, Kemppainen J, Kiviniemi A, Noponen T, Luotolahti M, Luoto
2): a randomised, multinational, double-blind, placebo-controlled, phase 2b trial. Lan- P, Oikonen V, Sipilä HT, Kopra J, Mononen I, Duncker DJ, Knuuti J, Kalliokoski KK.
cet 387: 1178 –1186, 2016. doi:10.1016/S0140-6736(16)00082-9. Myocardial blood flow and its transit time, oxygen utilization, and efficiency of highly
endurance-trained human heart. Basic Res Cardiol 109: 413, 2014. doi:10.1007/
140. Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, Russell s00395-014-0413-1.
DW, Chamberlain JS. Systemic delivery of genes to striated muscles using adeno-
associated viral vectors. Nat Med 10: 828 – 834, 2004. doi:10.1038/nm1085. 159. Heinonen I, Sorop O, de Beer VJ, Duncker DJ, Merkus D. What can we learn about
treating heart failure from the heart’s response to acute exercise? Focus on the
141. Grimm M, Brown JH. Beta-adrenergic receptor signaling in the heart: role of CaMKII. coronary microcirculation. J Appl Physiol (1985) 119: 934 –943, 2015. doi:10.1152/
J Mol Cell Cardiol 48: 322–330, 2010. doi:10.1016/j.yjmcc.2009.10.016. japplphysiol.00053.2015.

142. Groenendyk J, Agellon LB, Michalak M. Coping with endoplasmic reticulum stress in 160. Henstridge DC, Febbraio MA, Hargreaves M. Heat shock proteins and exercise
the cardiovascular system. Annu Rev Physiol 75: 49 – 67, 2013. doi:10.1146/annurev- adaptations. Our knowledge thus far and the road still ahead. J Appl Physiol (1985) 120:
physiol-030212-183707. 683– 691, 2016. doi:10.1152/japplphysiol.00811.2015.

143. Groenendyk J, Sreenivasaiah PK, Kim DH, Agellon LB, Michalak M. Biology of endo- 161. Hewitt G, Jurk D, Marques FD, Correia-Melo C, Hardy T, Gackowska A, Anderson R,
plasmic reticulum stress in the heart. Circ Res 107: 1185–1197, 2010. doi:10.1161/ Taschuk M, Mann J, Passos JF. Telomeres are favoured targets of a persistent DNA
CIRCRESAHA.110.227033. damage response in ageing and stress-induced senescence. Nat Commun 3: 708, 2012.
doi:10.1038/ncomms1708.
144. Gustafsson AB, Gottlieb RA. Heart mitochondria: gates of life and death. Cardiovasc
Res 77: 334 –343, 2008. doi:10.1093/cvr/cvm005. 162. Higginbotham MB, Morris KG, Coleman RE, Cobb FR. Sex-related differences in the
normal cardiac response to upright exercise. Circulation 70: 357–366, 1984. doi:10.
145. Haines CD, Harvey PA, Leinwand LA. Estrogens mediate cardiac hypertrophy in a 1161/01.CIR.70.3.357.
stimulus-dependent manner. Endocrinology 153: 4480 – 4490, 2012. doi:10.1210/en.
2012-1353. 163. Hill JA. Electrical remodeling in cardiac hypertrophy. Trends Cardiovasc Med 13: 316 –
322, 2003. doi:10.1016/j.tcm.2003.08.002.
146. Hajjar RJ. Potential of gene therapy as a treatment for heart failure. J Clin Invest 123:
53– 61, 2013. doi:10.1172/JCI62837. 164. Hohl M, Wagner M, Reil JC, Müller SA, Tauchnitz M, Zimmer AM, Lehmann LH, Thiel
G, Böhm M, Backs J, Maack C. HDAC4 controls histone methylation in response to
147. Hajjar RJ, Ishikawa K. Introducing Genes to the Heart: All About Delivery. Circ Res elevated cardiac load. J Clin Invest 123: 1359 –1370, 2013. doi:10.1172/JCI61084.
120: 33–35, 2017. doi:10.1161/CIRCRESAHA.116.310039.
165. Høydal MA, Kaurstad G, Rolim NP, Johnsen AB, Alves M, Koch LG, Britton SL, Stølen
148. Hamilton KL, Powers SK, Sugiura T, Kim S, Lennon S, Tumer N, Mehta JL. Short-term TO, Smith GL, Wisløff U. High inborn aerobic capacity does not protect the heart
exercise training can improve myocardial tolerance to I/R without elevation in heat following myocardial infarction. J Appl Physiol (1985) 115: 1788 –1795, 2013. doi:10.
shock proteins. Am J Physiol Heart Circ Physiol 281: H1346 –H1352, 2001. 1152/japplphysiol.00312.2013.

149. Hamm NC, Stammers AN, Susser SE, Hlynsky MW, Kimber DE, Kehler S, Duhamel 166. Huffman KM, Slentz CA, Bateman LA, Thompson D, Muehlbauer MJ, Bain JR, Stevens
TA. Regulation of Cardiac Sarco(endo)plasmic Reticulum Calcium-ATPases RD, Wenner BR, Kraus VB, Newgard CB, Kraus WE. Exercise-induced changes in
(SERCA2a) in Response to Exercise. In: Regulation of Ca2⫹-ATPases, V-ATPases and metabolic intermediates, hormones, and inflammatory markers associated with im-
F-ATPases, edited by Chakraborti S, Dhalla NS. Switzerland: Springer International, provements in insulin sensitivity. Diabetes Care 34: 174 –176, 2011. doi:10.2337/dc10-
2016, p. 187–206. doi:10.1007/978-3-319-24780-9_11. 0709.

466 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

167. Ihara Y, Morishima-Kawashima M, Nixon R. The ubiquitin-proteasome system and benefit of exercise training, resulting in cardiac deficits. Diabetes 53, Suppl 3: S169 –
the autophagic-lysosomal system in Alzheimer disease. Cold Spring Harb Perspect Med S175, 2004. doi:10.2337/diabetes.53.suppl_3.S169.
2: a006361, 2012. doi:10.1101/cshperspect.a006361.
184. Kane GC, Liu XK, Yamada S, Olson TM, Terzic A. Cardiac KATP channels in health and
168. Ishikawa K, Fish KM, Tilemann L, Rapti K, Aguero J, Santos-Gallego CG, Lee A, disease. J Mol Cell Cardiol 38: 937–943, 2005. doi:10.1016/j.yjmcc.2005.02.026.
Karakikes I, Xie C, Akar FG, Shimada YJ, Gwathmey JK, Asokan A, McPhee S, Samulski
J, Samulski RJ, Sigg DC, Weber T, Kranias EG, Hajjar RJ. Cardiac I-1c overexpression 185. Kang M, Chung KY, Walker JW. G-protein coupled receptor signaling in myocardium:
with reengineered AAV improves cardiac function in swine ischemic heart failure. Mol not for the faint of heart. Physiology (Bethesda) 22: 174 –184, 2007. doi:10.1152/
Ther 22: 2038 –2045, 2014. doi:10.1038/mt.2014.127. physiol.00051.2006.

169. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der 186. Kararigas G, Seeland U, Barcena de Arellano ML, Dworatzek E, Regitz-Zagrosek V.
Meer AJ, Patick AK, Chen A, Zhou Y, Persson R, King BD, Kauppinen S, Levin AA, Why the study of the effects of biological sex is important. Commentary. Ann Ist Super
Hodges MR. Treatment of HCV infection by targeting microRNA. N Engl J Med 368: Sanita 52: 149 –150, 2016. doi:10.4415/ANN_16_02_03.
1685–1694, 2013. doi:10.1056/NEJMoa1209026.
187. Karlowatz RJ, Scharhag J, Rahnenführer J, Schneider U, Jakob E, Kindermann W, Zang
170. Jaski BE, Jessup ML, Mancini DM, Cappola TP, Pauly DF, Greenberg B, Borow K, KD. Polymorphisms in the IGF1 signalling pathway including the myostatin gene are
Dittrich H, Zsebo KM, Hajjar RJ; Calcium Up-Regulation by Percutaneous Adminis- associated with left ventricular mass in male athletes. Br J Sports Med 45: 36 – 41, 2011.
tration of Gene Therapy In Cardiac Disease (CUPID) Trial Investigators. Calcium doi:10.1136/bjsm.2008.050567.
upregulation by percutaneous administration of gene therapy in cardiac disease (CU-
188. Karsenty G, Olson EN. Bone and Muscle Endocrine Functions: Unexpected Para-
PID Trial), a first-in-human phase 1/2 clinical trial. J Card Fail 15: 171–181, 2009.
digms of Inter-organ Communication. Cell 164: 1248 –1256, 2016. doi:10.1016/j.cell.
doi:10.1016/j.cardfail.2009.01.013.
2016.02.043.
171. Jaswal JS, Keung W, Wang W, Ussher JR, Lopaschuk GD. Targeting fatty acid and
189. Kasapis C, Thompson PD. The effects of physical activity on serum C-reactive protein
carbohydrate oxidation–a novel therapeutic intervention in the ischemic and failing
and inflammatory markers: a systematic review. J Am Coll Cardiol 45: 1563–1569,
heart. Biochim Biophys Acta 1813: 1333–1350, 2011. doi:10.1016/j.bbamcr.2011.01.
2005. doi:10.1016/j.jacc.2004.12.077.
015.
190. Katare R, Caporali A, Emanueli C, Madeddu P. Benfotiamine improves functional
172. Jennings G, Nelson L, Nestel P, Esler M, Korner P, Burton D, Bazelmans J. The effects
recovery of the infarcted heart via activation of pro-survival G6PD/Akt signaling path-
of changes in physical activity on major cardiovascular risk factors, hemodynamics,
way and modulation of neurohormonal response. J Mol Cell Cardiol 49: 625– 638,
sympathetic function, and glucose utilization in man: a controlled study of four levels
2010. doi:10.1016/j.yjmcc.2010.05.014.
of activity. Circulation 73: 30 – 40, 1986. doi:10.1161/01.CIR.73.1.30.
191. Katare RG, Caporali A, Oikawa A, Meloni M, Emanueli C, Madeddu P. Vitamin B1
173. Jessup M, Greenberg B, Mancini D, Cappola T, Pauly DF, Jaski B, Yaroshinsky A,
Zsebo KM, Dittrich H, Hajjar RJ; Calcium Upregulation by Percutaneous Administra- analog benfotiamine prevents diabetes-induced diastolic dysfunction and heart failure
tion of Gene Therapy in Cardiac Disease (CUPID) Investigators. Calcium Upregula- through Akt/Pim-1-mediated survival pathway. Circ Heart Fail 3: 294 –305, 2010.
tion by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID): a doi:10.1161/CIRCHEARTFAILURE.109.903450.
phase 2 trial of intracoronary gene therapy of sarcoplasmic reticulum Ca2⫹-ATPase in
192. Kaumann A, Bartel S, Molenaar P, Sanders L, Burrell K, Vetter D, Hempel P, Karcze-
patients with advanced heart failure. Circulation 124: 304 –313, 2011. doi:10.1161/
wski P, Krause EG. Activation of beta2-adrenergic receptors hastens relaxation and
CIRCULATIONAHA.111.022889.
mediates phosphorylation of phospholamban, troponin I, and C-protein in ventricular
174. Jiang BH, Liu LZ. PI3K/PTEN signaling in angiogenesis and tumorigenesis. Adv Cancer myocardium from patients with terminal heart failure. Circulation 99: 65–72, 1999.
Res 102: 19 – 65, 2009. doi:10.1016/S0065-230X(09)02002-8. doi:10.1161/01.CIR.99.1.65.

175. Jiang HK, Wang YH, Sun L, He X, Zhao M, Feng ZH, Yu XJ, Zang WJ. Aerobic interval 193. Kawase Y, Ly HQ, Prunier F, Lebeche D, Shi Y, Jin H, Hadri L, Yoneyama R, Hoshino
training attenuates mitochondrial dysfunction in rats post-myocardial infarction: roles K, Takewa Y, Sakata S, Peluso R, Zsebo K, Gwathmey JK, Tardif JC, Tanguay JF, Hajjar
of mitochondrial network dynamics. Int J Mol Sci 15: 5304 –5322, 2014. doi:10.3390/ RJ. Reversal of cardiac dysfunction after long-term expression of SERCA2a by gene
ijms15045304. transfer in a pre-clinical model of heart failure. J Am Coll Cardiol 51: 1112–1119, 2008.
doi:10.1016/j.jacc.2007.12.014.
176. Jin H, Yang R, Li W, Lu H, Ryan AM, Ogasawara AK, Van Peborgh J, Paoni NF. Effects
of exercise training on cardiac function, gene expression, and apoptosis in rats. Am J 194. Kee HJ, Eom GH, Joung H, Shin S, Kim JR, Cho YK, Choe N, Sim BW, Jo D, Jeong MH,
Physiol Heart Circ Physiol 279: H2994 –H3002, 2000. Kim KK, Seo JS, Kook H. Activation of histone deacetylase 2 by inducible heat shock
protein 70 in cardiac hypertrophy. Circ Res 103: 1259 –1269, 2008. doi:10.1161/01.
177. Johnsen AB, Høydal M, Røsbjørgen R, Stølen T, Wisløff U. Aerobic interval training RES.0000338570.27156.84.
partly reverse contractile dysfunction and impaired Ca2⫹ handling in atrial myocytes
from rats with post infarction heart failure. PLoS One 8: e66288, 2013. doi:10.1371/ 195. Kemi OJ, Ceci M, Condorelli G, Smith GL, Wisloff U. Myocardial sarcoplasmic retic-
journal.pone.0066288. ulum Ca2⫹ ATPase function is increased by aerobic interval training. Eur J Cardiovasc
Prev Rehabil 15: 145–148, 2008. doi:10.1097/HJR.0b013e3282efd4e0.
178. Jost N, Papp JG, Varró A. Slow delayed rectifier potassium current (IKs) and the
repolarization reserve. Ann Noninvasive Electrocardiol 12: 64 –78, 2007. doi:10.1111/ 196. Kemi OJ, Ellingsen O, Ceci M, Grimaldi S, Smith GL, Condorelli G, Wisløff U. Aerobic
j.1542-474X.2007.00140.x. interval training enhances cardiomyocyte contractility and Ca2⫹ cycling by phosphor-
ylation of CaMKII and Thr-17 of phospholamban. J Mol Cell Cardiol 43: 354 –361, 2007.
179. Jouven X, Empana JP, Schwartz PJ, Desnos M, Courbon D, Ducimetière P. Heart-rate doi:10.1016/j.yjmcc.2007.06.013.
profile during exercise as a predictor of sudden death. N Engl J Med 352: 1951–1958,
2005. doi:10.1056/NEJMoa043012. 197. Kemi OJ, Ellingsen O, Smith GL, Wisloff U. Exercise-induced changes in calcium
handling in left ventricular cardiomyocytes. Front Biosci 13: 356 –368, 2008. doi:10.
180. Kaese S, Verheule S. Cardiac electrophysiology in mice: a matter of size. Front Physiol 2741/2685.
3: 345, 2012. doi:10.3389/fphys.2012.00345.
198. Kemi OJ, Wisløff U. Mechanisms of exercise-induced improvements in the contractile
181. Kajstura J, Fiordaliso F, Andreoli AM, Li B, Chimenti S, Medow MS, Limana F, Nadal- apparatus of the mammalian myocardium. Acta Physiol (Oxf) 199: 425– 439, 2010.
Ginard B, Leri A, Anversa P. IGF-1 overexpression inhibits the development of dia- doi:10.1111/j.1748-1716.2010.02132.x.
betic cardiomyopathy and angiotensin II-mediated oxidative stress. Diabetes 50:
1414 –1424, 2001. doi:10.2337/diabetes.50.6.1414. 199. Khan RS, Martinez MD, Sy JC, Pendergrass KD, Che PL, Brown ME, Cabigas EB,
Dasari M, Murthy N, Davis ME. Targeting extracellular DNA to deliver IGF-1 to the
182. Kamo T, Akazawa H, Komuro I. Cardiac nonmyocytes in the hub of cardiac hyper- injured heart. Sci Rep 4: 4257, 2014. doi:10.1038/srep04257.
trophy. Circ Res 117: 89 –98, 2015. doi:10.1161/CIRCRESAHA.117.305349.
200. Kho C, Lee A, Jeong D, Oh JG, Chaanine AH, Kizana E, Park WJ, Hajjar RJ. SUMO1-
183. Kane GC, Behfar A, Yamada S, Perez-Terzic C, O’Cochlain F, Reyes S, Dzeja PP, Miki dependent modulation of SERCA2a in heart failure. Nature 477: 601– 605, 2011.
T, Seino S, Terzic A. ATP-sensitive K⫹ channel knockout compromises the metabolic doi:10.1038/nature10407.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 467


BERNARDO ET AL.

201. Kho C, Lee A, Jeong D, Oh JG, Gorski PA, Fish K, Sanchez R, DeVita RJ, Christensen 219. Lai CH, Ho TJ, Kuo WW, Day CH, Pai PY, Chung LC, Liao PH, Lin FH, Wu ET, Huang
G, Dahl R, Hajjar RJ. Small-molecule activation of SERCA2a SUMOylation for the CY. Exercise training enhanced SIRT1 longevity signaling replaces the IGF1 survival
treatment of heart failure. Nat Commun 6: 7229, 2015. doi:10.1038/ncomms8229. pathway to attenuate aging-induced rat heart apoptosis. Age (Dordr) 36: 9706, 2014.
doi:10.1007/s11357-014-9706-4.
202. Kiang JG, Tsokos GC. Heat shock protein 70 kDa: molecular biology, biochemistry,
and physiology. Pharmacol Ther 80: 183–201, 1998. doi:10.1016/S0163- 220. Lai L, Leone TC, Keller MP, Martin OJ, Broman AT, Nigro J, Kapoor K, Koves TR,
7258(98)00028-X. Stevens R, Ilkayeva OR, Vega RB, Attie AD, Muoio DM, Kelly DP. Energy metabolic
reprogramming in the hypertrophied and early stage failing heart: a multisystems
203. Kim J, Wende AR, Sena S, Theobald HA, Soto J, Sloan C, Wayment BE, Litwin SE, approach. Circ Heart Fail 7: 1022–1031, 2014. doi:10.1161/CIRCHEARTFAILURE.
Holzenberger M, LeRoith D, Abel ED. Insulin-like growth factor I receptor signaling is 114.001469.
required for exercise-induced cardiac hypertrophy. Mol Endocrinol 22: 2531–2543,
2008. doi:10.1210/me.2008-0265. 221. Lattanzi F, Di Bello V, Picano E, Caputo MT, Talarico L, Di Muro C, Landini L, Santoro
G, Giusti C, Distante A. Normal ultrasonic myocardial reflectivity in athletes with
204. Koch LG, Kemi OJ, Qi N, Leng SX, Bijma P, Gilligan LJ, Wilkinson JE, Wisløff H, Høydal increased left ventricular mass. A tissue characterization study. Circulation 85: 1828 –
MA, Rolim N, Abadir PM, van Grevenhof EM, Smith GL, Burant CF, Ellingsen O, 1834, 1992. doi:10.1161/01.CIR.85.5.1828.
Britton SL, Wisløff U. Intrinsic aerobic capacity sets a divide for aging and longevity.
222. Laughlin MH, Bowles DK, Duncker DJ. The coronary circulation in exercise training.
Circ Res 109: 1162–1172, 2011. doi:10.1161/CIRCRESAHA.111.253807.
Am J Physiol Heart Circ Physiol 302: H10 –H23, 2012. doi:10.1152/ajpheart.00574.
205. Kodama S, Saito K, Tanaka S, Maki M, Yachi Y, Asumi M, Sugawara A, Totsuka K, 2011.
Shimano H, Ohashi Y, Yamada N, Sone H. Cardiorespiratory fitness as a quantitative
223. Leask A. Potential therapeutic targets for cardiac fibrosis: TGFbeta, angiotensin, en-
predictor of all-cause mortality and cardiovascular events in healthy men and women:
dothelin, CCN2, and PDGF, partners in fibroblast activation. Circ Res 106: 1675–
a meta-analysis. JAMA 301: 2024 –2035, 2009. doi:10.1001/jama.2009.681.
1680, 2010. doi:10.1161/CIRCRESAHA.110.217737.
206. Kojda G, Hambrecht R. Molecular mechanisms of vascular adaptations to exercise.
224. Lee DC, Sui X, Artero EG, Lee IM, Church TS, McAuley PA, Stanford FC, Kohl
Physical activity as an effective antioxidant therapy? Cardiovasc Res 67: 187–197, 2005. HW III, Blair SN. Long-term effects of changes in cardiorespiratory fitness and
doi:10.1016/j.cardiores.2005.04.032. body mass index on all-cause and cardiovascular disease mortality in men: the
Aerobics Center Longitudinal Study. Circulation 124: 2483–2490, 2011. doi:10.
207. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug
1161/CIRCULATIONAHA.111.038422.
Discov 3: 711–715, 2004. doi:10.1038/nrd1470.
225. Lee IM, Sesso HD, Oguma Y, Paffenbarger RS Jr. Relative intensity of physical activity
208. Kong SW, Bodyak N, Yue P, Liu Z, Brown J, Izumo S, Kang PM. Genetic expression
and risk of coronary heart disease. Circulation 107: 1110 –1116, 2003. doi:10.1161/
profiles during physiological and pathological cardiac hypertrophy and heart failure in
01.CIR.0000052626.63602.58.
rats. Physiol Genomics 21: 34 – 42, 2005. doi:10.1152/physiolgenomics.00226.2004.
226. Lee SH, Kim J, Ryu JY, Lee S, Yang DK, Jeong D, Kim J, Lee S-H, Kim JM, Hajjar RJ, Park
209. Konhilas JP, Chen H, Luczak E, McKee LA, Regan J, Watson PA, Stauffer BL, Khalpey WJ. Transcription coactivator Eya2 is a critical regulator of physiological hypertrophy.
ZI, Mckinsey TA, Horn T, LaFleur B, Leinwand LA. Diet and sex modify exercise and J Mol Cell Cardiol 52: 718 –726, 2012. doi:10.1016/j.yjmcc.2011.12.002.
cardiac adaptation in the mouse. Am J Physiol Heart Circ Physiol 308: H135–H145,
2015. doi:10.1152/ajpheart.00532.2014. 227. Lee SH, Yang DK, Choi BY, Lee YH, Kim SY, Jeong D, Hajjar RJ, Park WJ. The
transcription factor Eya2 prevents pressure overload-induced adverse cardiac re-
210. Konhilas JP, Maass AH, Luckey SW, Stauffer BL, Olson EN, Leinwand LA. Sex modifies modeling. J Mol Cell Cardiol 46: 596 – 605, 2009. doi:10.1016/j.yjmcc.2008.12.021.
exercise and cardiac adaptation in mice. Am J Physiol Heart Circ Physiol 287: H2768 –
H2776, 2004. doi:10.1152/ajpheart.00292.2004. 228. Legato MJ. Gender-specific medicine in the genomic era. Clin Sci (Lond) 130: 1–7,
2016. doi:10.1042/CS20150551.
211. Kourtis N, Tavernarakis N. Cellular stress response pathways and ageing: intricate
molecular relationships. EMBO J 30: 2520 –2531, 2011. doi:10.1038/emboj.2011.162. 229. Leong IU, Skinner JR, Shelling AN, Love DR. Zebrafish as a model for long QT
syndrome: the evidence and the means of manipulating zebrafish gene expression.
212. Koziris LP, Hickson RC, Chatterton RT, Jr, Groseth RT, Christie JM, Goldflies DG, Acta Physiol (Oxf) 199: 257–276, 2010. doi:10.1111/j.1748-1716.2010.02111.x.
Unterman TG. Serum levels of total and free IGF-I and IGFBP-3 are increased and
maintained in long-term training. J Appl Physiol (1985) 86: 1436 –1442, 1999. 230. Lewis GD, Farrell L, Wood MJ, Martinovic M, Arany Z, Rowe GC, Souza A, Cheng S,
McCabe EL, Yang E, Shi X, Deo R, Roth FP, Asnani A, Rhee EP, Systrom DM,
213. Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of Semigran MJ, Vasan RS, Carr SA, Wang TJ, Sabatine MS, Clish CB, Gerszten RE.
obesity-associated diabetes. Nat Rev Drug Discov 15: 639 – 660, 2016. doi:10.1038/ Metabolic signatures of exercise in human plasma. Sci Transl Med 2: 33ra37, 2010.
nrd.2016.75. doi:10.1126/scitranslmed.3001006.

214. Kuster DW, Merkus D, Blonden LA, Kremer A, van IJcken WF, Verhoeven AJ, 231. Li HH, Willis MS, Lockyer P, Miller N, McDonough H, Glass DJ, Patterson C.
Duncker DJ. Gene reprogramming in exercise-induced cardiac hypertrophy in swine: Atrogin-1 inhibits Akt-dependent cardiac hypertrophy in mice via ubiquitin-depen-
a transcriptional genomics approach. J Mol Cell Cardiol 77: 168 –174, 2014. doi:10. dent coactivation of Forkhead proteins. J Clin Invest 117: 3211–3223, 2007. doi:10.
1016/j.yjmcc.2014.10.006. 1172/JCI31757.

215. Kwak HB. Aging, exercise, and extracellular matrix in the heart. J Exerc Rehabil 9: 232. Li M, Gao P, Zhang J. Crosstalk between Autophagy and Apoptosis: Potential and
338 –347, 2013. doi:10.12965/jer.130049. Emerging Therapeutic Targets for Cardiac Diseases. Int J Mol Sci 17: 332, 2016.
doi:10.3390/ijms17030332.
216. Kwak HB, Kim JH, Joshi K, Yeh A, Martinez DA, Lawler JM. Exercise training reduces
233. Li Q, Li B, Wang X, Leri A, Jana KP, Liu Y, Kajstura J, Baserga R, Anversa P. Overex-
fibrosis and matrix metalloproteinase dysregulation in the aging rat heart. FASEB J 25:
pression of insulin-like growth factor-1 in mice protects from myocyte death after
1106 –1117, 2011. doi:10.1096/fj.10-172924.
infarction, attenuating ventricular dilation, wall stress, and cardiac hypertrophy. J Clin
217. Kwong JQ, Lu X, Correll RN, Schwanekamp JA, Vagnozzi RJ, Sargent MA, York AJ, Invest 100: 1991–1999, 1997. doi:10.1172/JCI119730.
Zhang J, Bers DM, Molkentin JD. The Mitochondrial Calcium Uniporter Selectively
234. Liddy KA, White MY, Cordwell SJ. Functional decorations: post-translational modifi-
Matches Metabolic Output to Acute Contractile Stress in the Heart. Cell Reports 12:
cations and heart disease delineated by targeted proteomics. Genome Med 5: 20,
15–22, 2015. doi:10.1016/j.celrep.2015.06.002.
2013. doi:10.1186/gm424.
218. Kyu HH, Bachman VF, Alexander LT, Mumford JE, Afshin A, Estep K, Veerman JL,
235. Lin JH, Walter P, Yen TS. Endoplasmic reticulum stress in disease pathogenesis. Annu
Delwiche K, Iannarone ML, Moyer ML, Cercy K, Vos T, Murray CJ, Forouzanfar MH. Rev Pathol 3: 399 – 425, 2008. doi:10.1146/annurev.pathmechdis.3.121806.151434.
Physical activity and risk of breast cancer, colon cancer, diabetes, ischemic heart
disease, and ischemic stroke events: systematic review and dose-response meta- 236. Lin RC, Weeks KL, Gao XM, Williams RB, Bernardo BC, Kiriazis H, Matthews VB,
analysis for the Global Burden of Disease Study 2013. BMJ 354: i3857, 2016. doi:10. Woodcock EA, Bouwman RD, Mollica JP, Speirs HJ, Dawes IW, Daly RJ, Shioi T,
1136/bmj.i3857. Izumo S, Febbraio MA, Du XJ, McMullen JR. PI3K(p110 alpha) protects against myo-

468 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

cardial infarction-induced heart failure: identification of PI3K-regulated miRNA and 254. Marques FZ, Booth SA, Prestes PR, Curl CL, Delbridge LM, Lewandowski P, Harrap
mRNA. Arterioscler Thromb Vasc Biol 30: 724 –732, 2010. doi:10.1161/ATVBAHA. SB, Charchar FJ. Telomere dynamics during aging in polygenic left ventricular hyper-
109.201988. trophy. Physiol Genomics 48: 42– 49, 2016. doi:10.1152/physiolgenomics.00083.2015.

237. Ling C, Rönn T. Epigenetic adaptation to regular exercise in humans. Drug Discov 255. Marsh SA, Collins HE, Chatham JC. Protein O-GlcNAcylation and cardiovascular
Today 19: 1015–1018, 2014. doi:10.1016/j.drudis.2014.03.006. (patho)physiology. J Biol Chem 289: 34449 –34456, 2014. doi:10.1074/jbc.R114.
585984.
238. Lira VA, Kronemberger A, Call JA, Caster HM, Pereira RO, Laker RC, Zhang M, Yan
Z. Defective autophagy causes a maladpative cardiac phenotype to exercise that leads 256. Martin ML, Blaxall BC. Cardiac intercellular communication: are myocytes and fibro-
to premature death and FGF21-mediated protection against obesity and insulin resis- blasts fair-weather friends? J Cardiovasc Transl Res 5: 768 –782, 2012. doi:10.1007/
tance. FASEB J 30: 1014, 2016. doi:10.1038/s41598-017-08480-2. s12265-012-9404-5.

239. Literáti-Nagy B, Kulcsár E, Literáti-Nagy Z, Buday B, Péterfai E, Horváth T, Tory K, 257. Martinelli NC, Cohen CR, Santos KG, Castro MA, Biolo A, Frick L, Silvello D, Lopes
Kolonics A, Fleming A, Mandl J, Korányi L. Improvement of insulin sensitivity by a A, Schneider S, Andrades ME, Clausell N, Matte U, Rohde LE. An analysis of the global
novel drug, BGP-15, in insulin-resistant patients: a proof of concept randomized expression of microRNAs in an experimental model of physiological left ventricular
hypertrophy. PLoS One 9: e93271, 2014. doi:10.1371/journal.pone.0093271.
double-blind clinical trial. Horm Metab Res 41: 374 –380, 2009. doi:10.1055/s-0028-
1128142. 258. Masoud WG, Ussher JR, Wang W, Jaswal JS, Wagg CS, Dyck JR, Lygate CA, Neubauer
S, Clanachan AS, Lopaschuk GD. Failing mouse hearts utilize energy inefficiently and
240. Liu LX, Arany Z. Maternal cardiac metabolism in pregnancy. Cardiovasc Res 101:
benefit from improved coupling of glycolysis and glucose oxidation. Cardiovasc Res
545–553, 2014. doi:10.1093/cvr/cvu009.
101: 30 –38, 2014. doi:10.1093/cvr/cvt216.
241. Liu M, Dudley SC Jr. Role for the Unfolded Protein Response in Heart Disease and 259. Massion PB, Feron O, Dessy C, Balligand JL. Nitric oxide and cardiac function: ten
Cardiac Arrhythmias. Int J Mol Sci 17: E52, 2015. doi:10.3390/ijms17010052. years after, and continuing. Circ Res 93: 388 –398, 2003. doi:10.1161/01.RES.
0000088351.58510.21.
242. Liu W, Zhou X, Yu F, Hu J, Hu W. Arg972 Insulin receptor substrate-1 is associated
with decreased serum angiotensin-converting enzyme 2 levels in acute myocardial 260. Matsui T, Tao J, del Monte F, Lee KH, Li L, Picard M, Force TL, Franke TF, Hajjar RJ,
infarction patients: in vivo and in vitro evidence. Cardiovasc Diabetol 12: 151, 2013. Rosenzweig A. Akt activation preserves cardiac function and prevents injury after
doi:10.1186/1475-2840-12-151. transient cardiac ischemia in vivo. Circulation 104: 330 –335, 2001. doi:10.1161/01.
CIR.104.3.330.
243. Liu X, Xiao J, Zhu H, Wei X, Platt C, Damilano F, Xiao C, Bezzerides V, Boström P,
Che L, Zhang C, Spiegelman BM, Rosenzweig A. miR-222 is necessary for exercise- 261. Matthews KG, Devlin GP, Stuart SP, Jensen JA, Doughty RN, Conaglen JV, Bass JJ.
induced cardiac growth and protects against pathological cardiac remodeling. Cell Intrapericardial IGF-I improves cardiac function in an ovine model of chronic heart
Metab 21: 584 –595, 2015. doi:10.1016/j.cmet.2015.02.014. failure. Heart Lung Circ 14: 98 –103, 2005. doi:10.1016/j.hlc.2005.02.002.

244. Lopaschuk GD, Ussher JR. Evolving Concepts of Myocardial Energy Metabolism: 262. McElhinney DB, Colan SD, Moran AM, Wypij D, Lin M, Majzoub JA, Crawford EC,
More Than Just Fats and Carbohydrates. Circ Res 119: 1173–1176, 2016. doi:10.1161/ Bartlett JM, McGrath EA, Newburger JW. Recombinant human growth hormone
CIRCRESAHA.116.310078. treatment for dilated cardiomyopathy in children. Pediatrics 114: e452– e458, 2004.
doi:10.1542/peds.2004-0072.
245. Luczak ED, Leinwand LA. Sex-based cardiac physiology. Annu Rev Physiol 71: 1–18,
2009. doi:10.1146/annurev.physiol.010908.163156. 263. McFarlin BK, Flynn MG, Campbell WW, Stewart LK, Timmerman KL. TLR4 is lower
in resistance-trained older women and related to inflammatory cytokines. Med Sci
246. Ludlow AT, Ludlow LW, Roth SM. Do telomeres adapt to physiological stress? Ex- Sports Exerc 36: 1876 –1883, 2004. doi:10.1249/01.MSS.0000145465.71269.10.
ploring the effect of exercise on telomere length and telomere-related proteins.
BioMed Res Int 2013: 601368, 2013. doi:10.1155/2013/601368. 264. McGee SL, Fairlie E, Garnham AP, Hargreaves M. Exercise-induced histone modifi-
cations in human skeletal muscle. J Physiol 587: 5951–5958, 2009. doi:10.1113/
247. Luo J, McMullen JR, Sobkiw CL, Zhang L, Dorfman AL, Sherwood MC, Logsdon MN, jphysiol.2009.181065.
Horner JW, DePinho RA, Izumo S, Cantley LC. Class IA phosphoinositide 3-kinase
265. McMullen JR. Role of insulin-like growth factor 1 and phosphoinositide 3-kinase in a
regulates heart size and physiological cardiac hypertrophy. Mol Cell Biol 25: 9491–
setting of heart disease. Clin Exp Pharmacol Physiol 35: 349 –354, 2008. doi:10.1111/
9502, 2005. doi:10.1128/MCB.25.21.9491-9502.2005.
j.1440-1681.2007.04873.x.
248. Ma X, Fu Y, Xiao H, Song Y, Chen R, Shen J, An X, Shen Q, Li Z, Zhang Y. Cardiac 266. McMullen JR, Amirahmadi F, Woodcock EA, Schinke-Braun M, Bouwman RD, Hewitt
Fibrosis Alleviated by Exercise Training Is AMPK-Dependent. PLoS One 10: e0129971, KA, Mollica JP, Zhang L, Zhang Y, Shioi T, Buerger A, Izumo S, Jay PY, Jennings GL.
2015. doi:10.1371/journal.pone.0129971. Protective effects of exercise and phosphoinositide 3-kinase(p110alpha) signaling in
dilated and hypertrophic cardiomyopathy. Proc Natl Acad Sci USA 104: 612– 617,
249. Ma Z, Qi J, Meng S, Wen B, Zhang J. Swimming exercise training-induced left ventric-
2007. doi:10.1073/pnas.0606663104.
ular hypertrophy involves microRNAs and synergistic regulation of the PI3K/AKT/
mTOR signaling pathway. Eur J Appl Physiol 113: 2473–2486, 2013. doi:10.1007/ 267. McMullen JR, Shioi T, Huang W-Y, Zhang L, Tarnavski O, Bisping E, Schinke M, Kong
s00421-013-2685-9. S, Sherwood MC, Brown J, Riggi L, Kang PM, Izumo S. The insulin-like growth factor
1 receptor induces physiological heart growth via the phosphoinositide
250. Magalhães J, Gonçalves IO, Lumini-Oliveira J, Marques-Aleixo I, Passos E, Rocha- 3-kinase(p110alpha) pathway. J Biol Chem 279: 4782– 4793, 2004. doi:10.1074/jbc.
Rodrigues S, Machado NG, Moreira AC, Rizo D, Viscor G, Oliveira PJ, Torrella JR, M310405200.
Ascensão A. Modulation of cardiac mitochondrial permeability transition and ap-
optotic signaling by endurance training and intermittent hypobaric hypoxia. Int J 268. McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Dorfman AL, Longnus S,
Cardiol 173: 40 – 45, 2014. doi:10.1016/j.ijcard.2014.02.011. Pende M, Martin KA, Blenis J, Thomas G, Izumo S. Deletion of ribosomal S6 kinases
does not attenuate pathological, physiological, or insulin-like growth factor 1 recep-
251. Manoury B, Montiel V, Balligand JL. Nitric oxide synthase in post-ischaemic remod- tor-phosphoinositide 3-kinase-induced cardiac hypertrophy. Mol Cell Biol 24: 6231–
elling: new pathways and mechanisms. Cardiovasc Res 94: 304 –315, 2012. doi:10. 6240, 2004. doi:10.1128/MCB.24.14.6231-6240.2004.
1093/cvr/cvr360.
269. McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Kang PM, Izumo S.
252. Mao K, Kobayashi S, Jaffer ZM, Huang Y, Volden P, Chernoff J, Liang Q. Regulation of Phosphoinositide 3-kinase(p110alpha) plays a critical role for the induction of physi-
Akt/PKB activity by P21-activated kinase in cardiomyocytes. J Mol Cell Cardiol 44: ological, but not pathological, cardiac hypertrophy. Proc Natl Acad Sci USA 100:
429 – 434, 2008. doi:10.1016/j.yjmcc.2007.10.016. 12355–12360, 2003. doi:10.1073/pnas.1934654100.

253. Marioni JC, Mason CE, Mane SM, Stephens M, Gilad Y. RNA-seq: an assessment of 270. Mearini G, Schlossarek S, Willis MS, Carrier L. The ubiquitin-proteasome system in
technical reproducibility and comparison with gene expression arrays. Genome Res cardiac dysfunction. Biochim Biophys Acta 1782: 749 –763, 2008. doi:10.1016/j.bbadis.
18: 1509 –1517, 2008. doi:10.1101/gr.079558.108. 2008.06.009.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 469


BERNARDO ET AL.

271. Medford HM, Porter K, Marsh SA. Immediate effects of a single exercise bout on hypertrophy by novel irreversible inhibitors of CRM1. Biochim Biophys Acta 1789:
protein O-GlcNAcylation and chromatin regulation of cardiac hypertrophy. Am J 422– 431, 2009. doi:10.1016/j.bbagrm.2009.04.001.
Physiol Heart Circ Physiol 305: H114 –H123, 2013. doi:10.1152/ajpheart.00135.2013.
288. Mooren FC, Viereck J, Krüger K, Thum T. Circulating microRNAs as potential bio-
272. Melo SF, Barauna VG, Júnior MA, Bozi LH, Drummond LR, Natali AJ, de Oliveira EM. markers of aerobic exercise capacity. Am J Physiol Heart Circ Physiol 306: H557–H563,
Resistance training regulates cardiac function through modulation of miRNA-214. Int 2014. doi:10.1152/ajpheart.00711.2013.
J Mol Sci 16: 6855– 6867, 2015. doi:10.3390/ijms16046855.
289. Moreira-Gonçalves D, Henriques-Coelho T, Fonseca H, Ferreira R, Padrão AI, Santa
273. Melo SF, Barauna VG, Neves VJ, Fernandes T, Lara LS, Mazzotti DR, Oliveira EM. C, Vieira S, Silva AF, Amado F, Leite-Moreira A, Duarte JA. Intermittent cardiac
Exercise training restores the cardiac microRNA-1 and -214 levels regulating Ca2⫹ overload results in adaptive hypertrophy and provides protection against left ventric-
handling after myocardial infarction. BMC Cardiovasc Disord 15: 166, 2015. doi:10. ular acute pressure overload insult. J Physiol 593: 3885–3897, 2015. doi:10.1113/
1186/s12872-015-0156-4. JP270685.

274. Melo SF, Fernandes T, Baraúna VG, Matos KC, Santos AA, Tucci PJ, Oliveira EM. 290. Moreira JB, Wohlwend M, Alves MN, Wisløff U, Bye A. A small molecule activator of
Expression of MicroRNA-29 and Collagen in Cardiac Muscle after Swimming Training AKT does not reduce ischemic injury of the rat heart. J Transl Med 13: 76, 2015.
in Myocardial-Infarcted Rats. Cell Physiol Biochem 33: 657– 669, 2014. doi:10.1159/ doi:10.1186/s12967-015-0444-x.
000358642.
291. Morris JN, Everitt MG, Pollard R, Chave SP, Semmence AM. Vigorous exercise in
275. Mesirca P, Marger L, Toyoda F, Rizzetto R, Audoubert M, Dubel S, Torrente AG, leisure-time: protection against coronary heart disease. Lancet 2: 1207–1210, 1980.
Difrancesco ML, Muller JC, Leoni AL, Couette B, Nargeot J, Clapham DE, Wickman doi:10.1016/S0140-6736(80)92476-9.
K, Mangoni ME. The G-protein-gated K⫹ channel, IKACh, is required for regulation of
pacemaker activity and recovery of resting heart rate after sympathetic stimulation. J 292. Morris KV, Mattick JS. The rise of regulatory RNA. Nat Rev Genet 15: 423– 437, 2014.
Gen Physiol 142: 113–126, 2013. doi:10.1085/jgp.201310996. doi:10.1038/nrg3722.

276. Milne KJ, Noble EG. Exercise-induced elevation of HSP70 is intensity dependent. J 293. Murlasits Z, Lee Y, Powers SK. Short-term exercise does not increase ER stress
Appl Physiol (1985) 93: 561–568, 2002. doi:10.1152/japplphysiol.00528.2001. protein expression in cardiac muscle. Med Sci Sports Exerc 39: 1522–1528, 2007.
doi:10.1249/mss.0b013e3180cc25c7.
277. Milne KJ, Thorp DB, Melling CW, Noble EG. Castration inhibits exercise-induced
accumulation of Hsp70 in male rodent hearts. Am J Physiol Heart Circ Physiol 290: 294. Nah J, Fernández AF, Kitsis RN, Levine B, Sadoshima J. Does Autophagy Mediate
H1610 –H1616, 2006. doi:10.1152/ajpheart.01103.2004. Cardiac Myocyte Death During Stress? Circ Res 119: 893– 895, 2016. doi:10.1161/
CIRCRESAHA.116.309765.
278. Min JN, Whaley RA, Sharpless NE, Lockyer P, Portbury AL, Patterson C. CHIP
deficiency decreases longevity, with accelerated aging phenotypes accompanied by 295. Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I,
altered protein quality control. Mol Cell Biol 28: 4018 – 4025, 2008. doi:10.1128/MCB. Matsumura Y, Asahi M, Nishida K, Hori M, Mizushima N, Otsu K. The role of au-
00296-08. tophagy in cardiomyocytes in the basal state and in response to hemodynamic stress.
Nat Med 13: 619 – 624, 2007. doi:10.1038/nm1574.
279. Minamino T, Komuro I, Kitakaze M. Endoplasmic reticulum stress as a therapeutic
target in cardiovascular disease. Circ Res 107: 1071–1082, 2010. doi:10.1161/ 296. Natale VM, Brenner IK, Moldoveanu AI, Vasiliou P, Shek P, Shephard RJ. Effects of
CIRCRESAHA.110.227819. three different types of exercise on blood leukocyte count during and following
exercise. Sao Paulo Med J 121: 9 –14, 2003. doi:10.1590/S1516-31802003000100003.
280. Mingozzi F, High KA. Immune responses to AAV vectors: overcoming barriers to
successful gene therapy. Blood 122: 23–36, 2013. doi:10.1182/blood-2013-01- 296a.National Human Genome Research Institute. DNA Sequencing Costs: Data from the
306647. NHGRI Genome Sequencing Program (GSP). https://www.genome.gov/sequencing-
costsdata.
281. Miyamoto MI, del Monte F, Schmidt U, DiSalvo TS, Kang ZB, Matsui T, Guerrero JL,
Gwathmey JK, Rosenzweig A, Hajjar RJ. Adenoviral gene transfer of SERCA2a im- 297. Nattel S, Carlsson L. Innovative approaches to anti-arrhythmic drug therapy. Nat Rev
proves left-ventricular function in aortic-banded rats in transition to heart failure. Proc Drug Discov 5: 1034 –1049, 2006. doi:10.1038/nrd2112.
Natl Acad Sci USA 97: 793–798, 2000. doi:10.1073/pnas.97.2.793.
298. Nelson L, Jennings GL, Esler MD, Korner PI. Effect of changing levels of physical
282. Moc C, Taylor AE, Chesini GP, Zambrano CM, Barlow MS, Zhang X, Gustafsson AB, activity on blood-pressure and haemodynamics in essential hypertension. Lancet 2:
Purcell NH. Physiological activation of Akt by PHLPP1 deletion protects against path- 473– 476, 1986. doi:10.1016/S0140-6736(86)90354-5.
ological hypertrophy. Cardiovasc Res 105: 160 –170, 2015. doi:10.1093/cvr/cvu243.
299. Nerbonne JM, Kass RS. Molecular physiology of cardiac repolarization. Physiol Rev 85:
283. Molenaar P, Bartel S, Cochrane A, Vetter D, Jalali H, Pohlner P, Burrell K, Karczewski 1205–1253, 2005. doi:10.1152/physrev.00002.2005.
P, Krause EG, Kaumann A. Both beta(2)- and beta(1)-adrenergic receptors mediate
hastened relaxation and phosphorylation of phospholamban and troponin I in ventric- 300. Nerbonne JM, Nichols CG, Schwarz TL, Escande D. Genetic manipulation of cardiac
ular myocardium of Fallot infants, consistent with selective coupling of beta(2)-adren- K(⫹) channel function in mice: what have we learned, and where do we go from here?
ergic receptors to G(s)-protein. Circulation 102: 1814 –1821, 2000. doi:10.1161/01. Circ Res 89: 944 –956, 2001. doi:10.1161/hh2301.100349.
CIR.102.15.1814.
301. Neri Serneri GG, Boddi M, Modesti PA, Cecioni I, Coppo M, Padeletti L, Michelucci
284. Molenaar P, Savarimuthu SM, Sarsero D, Chen L, Semmler AB, Carle A, Yang I, Bartel A, Colella A, Galanti G. Increased cardiac sympathetic activity and insulin-like growth
S, Vetter D, Beyerdörfer I, Krause EG, Kaumann AJ. (-)-Adrenaline elicits positive factor-I formation are associated with physiological hypertrophy in athletes. Circ Res
inotropic, lusitropic, and biochemical effects through beta2-adrenoceptors in human 89: 977–982, 2001. doi:10.1161/hh2301.100982.
atrial myocardium from nonfailing and failing hearts, consistent with Gs coupling but
not with Gi coupling. Naunyn Schmiedebergs Arch Pharmacol 375: 11–28, 2007. doi:10. 302. Niizuma S, Inuzuka Y, Okuda J, Kato T, Kawashima T, Tamaki Y, Iwanaga Y, Yoshida
1007/s00210-007-0138-x. Y, Kosugi R, Watanabe-Maeda K, Machida Y, Tsuji S, Aburatani H, Izumi T, Kita T,
Kimura T, Shioi T. Effect of persistent activation of phosphoinositide 3-kinase on
285. Mollova MY, Katus HA, Backs J. Regulation of CaMKII signaling in cardiovascular heart. Life Sci 90: 619 – 628, 2012. doi:10.1016/j.lfs.2012.02.010.
disease. Front Pharmacol 6: 178, 2015. doi:10.3389/fphar.2015.00178.
303. Nio AQ, Stöhr EJ, Shave R. The female human heart at rest and during exercise: a
286. Monleon D, Garcia-Valles R, Morales JM, Brioche T, Olaso-Gonzalez G, Lopez- review. Eur J Sport Sci 15: 286 –295, 2015. doi:10.1080/17461391.2014.936323.
Grueso R, Gomez-Cabrera MC, Viña J. Metabolomic analysis of long-term spontane-
ous exercise in mice suggests increased lipolysis and altered glucose metabolism when 304. Nishida K, Taneike M, Otsu K. The role of autophagic degradation in the heart. J Mol
animals are at rest. J Appl Physiol (1985) 117: 1110 –1119, 2014. doi:10.1152/ Cell Cardiol 78: 73–79, 2015. doi:10.1016/j.yjmcc.2014.09.029.
japplphysiol.00585.2014.
305. Nissinen SI, Mäkikallio TH, Seppänen T, Tapanainen JM, Salo M, Tulppo MP, Huikuri
287. Monovich L, Koch KA, Burgis R, Osimboni E, Mann T, Wall D, Gao J, Feng Y, Vega RB, HV. Heart rate recovery after exercise as a predictor of mortality among survivors of
Turner BA, Hood DB, Law A, Papst PJ, Koditek D, Chapo JA, Reid BG, Melvin LS, acute myocardial infarction. Am J Cardiol 91: 711–714, 2003. doi:10.1016/S0002-
Pagratis NC, McKinsey TA. Suppression of HDAC nuclear export and cardiomyocyte 9149(02)03410-0.

470 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

306. Noble EG, Moraska A, Mazzeo RS, Roth DA, Olsson MC, Moore RL, Fleshner M. 324. Penny WF, Hammond HK. Randomized Clinical Trials of Gene Transfer for Heart
Differential expression of stress proteins in rat myocardium after free wheel or tread- Failure with Reduced Ejection Fraction. Hum Gene Ther 28: 378 –384, 2017. doi:10.
mill run training. J Appl Physiol (1985) 86: 1696 –1701, 1999. 1089/hum.2016.166.

307. Noh J, Wende AR, Olsen CD, Kim B, Bevins J, Zhu Y, Zhang QJ, Riehle C, Abel ED. 325. Petersen AM, Pedersen BK. The anti-inflammatory effect of exercise. J Appl Physiol
Phosphoinositide dependent protein kinase 1 is required for exercise-induced cardiac (1985) 98: 1154 –1162, 2005. doi:10.1152/japplphysiol.00164.2004.
hypertrophy but not the associated mitochondrial adaptations. J Mol Cell Cardiol 89, Pt
B: 297–305, 2015. doi:10.1016/j.yjmcc.2015.10.015. 326. Petersen SE, Hudsmith LE, Robson MD, Doll HA, Francis JM, Wiesmann F, Jung BA,
Hennig J, Watkins H, Neubauer S. Sex-specific characteristics of cardiac function,
308. Ntanasis-Stathopoulos J, Tzanninis JG, Philippou A, Koutsilieris M. Epigenetic regula- geometry, and mass in young adult elite athletes. J Magn Reson Imaging 24: 297–303,
tion on gene expression induced by physical exercise. J Musculoskelet Neuronal Inter- 2006. doi:10.1002/jmri.20633.
act 13: 133–146, 2013.
327. Piña IL, Apstein CS, Balady GJ, Belardinelli R, Chaitman BR, Duscha BD, Fletcher BJ,
309. Nunes RB, Alves JP, Kessler LP, Dal Lago P. Aerobic exercise improves the inflam- Fleg JL, Myers JN, Sullivan MJ; American Heart Association Committee on exercise,
matory profile correlated with cardiac remodeling and function in chronic heart failure rehabilitation, and prevention. Exercise and heart failure: a statement from the Amer-
rats. Clinics (Sao Paulo) 68: 876 – 882, 2013. doi:10.6061/clinics/2013(06)24. ican Heart Association Committee on exercise, rehabilitation, and prevention. Circu-
lation 107: 1210 –1225, 2003. doi:10.1161/01.CIR.0000055013.92097.40.
310. O’Neill BT, Kim J, Wende AR, Theobald HA, Tuinei J, Buchanan J, Guo A, Zaha VG,
Davis DK, Schell JC, Boudina S, Wayment B, Litwin SE, Shioi T, Izumo S, Birnbaum MJ, 328. Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’Antoni ML, Debuque R, Chandran A,
Abel ED. A conserved role for phosphatidylinositol 3-kinase but not Akt signaling Wang L, Arora K, Rosenthal NA, Tallquist MD. Revisiting Cardiac Cellular Composi-
in mitochondrial adaptations that accompany physiological cardiac hypertrophy. tion. Circ Res 118: 400 – 409, 2016. doi:10.1161/CIRCRESAHA.115.307778.
Cell Metab 6: 294 –306, 2007. doi:10.1016/j.cmet.2007.09.001.
329. Pleger ST, Shan C, Ksienzyk J, Bekeredjian R, Boekstegers P, Hinkel R, Schinkel S,
311. Ogawa S, Chan J, Gustafsson JA, Korach KS, Pfaff DW. Estrogen increases locomotor Leuchs B, Ludwig J, Qiu G, Weber C, Raake P, Koch WJ, Katus HA, Müller OJ, Most
P. Cardiac AAV9-S100A1 gene therapy rescues post-ischemic heart failure in a pre-
activity in mice through estrogen receptor alpha: specificity for the type of activity.
clinical large animal model. Sci Transl Med 3: 92ra64, 2011. doi:10.1126/scitranslmed.
Endocrinology 144: 230 –239, 2003. doi:10.1210/en.2002-220519.
3002097.
312. Ogura Y, Iemitsu M, Naito H, Kakigi R, Kakehashi C, Maeda S, Akema T. Single bout
330. Polakoviová M, Musil P, Laczo E, Hamar D, Kyselovi J. Circulating MicroRNAs as
of running exercise changes LC3-II expression in rat cardiac muscle. Biochem Biophys
Potential Biomarkers of Exercise Response. Int J Mol Sci 17: E1553, 2016. doi:10.
Res Commun 414: 756 –760, 2011. doi:10.1016/j.bbrc.2011.09.152.
3390/ijms17101553.
313. Oka T, Maillet M, Watt AJ, Schwartz RJ, Aronow BJ, Duncan SA, Molkentin JD.
331. Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, Sadek HA.
Cardiac-specific deletion of Gata4 reveals its requirement for hypertrophy, compen-
Transient regenerative potential of the neonatal mouse heart. Science 331: 1078 –
sation, and myocyte viability. Circ Res 98: 837– 845, 2006. doi:10.1161/01.RES.
1080, 2011. doi:10.1126/science.1200708.
0000215985.18538.c4.
332. Powers SK, Sollanek KJ, Wiggs MP, Demirel HA, Smuder AJ. Exercise-induced im-
314. Olfert IM, Howlett RA, Tang K, Dalton ND, Gu Y, Peterson KL, Wagner PD, Breen
provements in myocardial antioxidant capacity: the antioxidant players and cardio-
EC. Muscle-specific VEGF deficiency greatly reduces exercise endurance in mice. J
protection. Free Radic Res 48: 43–51, 2014. doi:10.3109/10715762.2013.825371.
Physiol 587: 1755–1767, 2009. doi:10.1113/jphysiol.2008.164384.
333. Prabhavathi K, Selvi KT, Poornima KN, Sarvanan A. Role of biological sex in normal
315. Ooi JYY, Bernardo BC, McMullen JR. The therapeutic potential of miRNAs regulated cardiac function and in its disease outcome - a review. J Clin Diagn Res 8: BE01–BE04,
in settings of physiological cardiac hypertrophy. Future Med Chem 6: 205–222, 2014. 2014.
doi:10.4155/fmc.13.196.
334. Prakoso D, De Blasio MJ, Qin C, Rosli S, Kiriazis H, Qian H, Du XJ, Weeks KL,
316. Ooi JYY, Bernardo BC, Singla S, Patterson NL, Lin RCY, McMullen JR. Identification of Gregorevic P, McMullen JR, Ritchie RH. Phosphoinositide 3-kinase (p110␣) gene
miR-34 regulatory networks in settings of disease and antimiR-therapy: implications delivery limits diabetes-induced cardiac NADPH oxidase and cardiomyopathy in a
for treating cardiac pathology and other diseases. RNA Biol 14: 500 –513, 2017. doi: mouse model with established diastolic dysfunction. Clin Sci (Lond) 131: 1345–1360,
10.1080/15476286.2016.1181251. 2017. doi:10.1042/CS20170063.

317. Osterziel KJ, Strohm O, Schuler J, Friedrich M, Hänlein D, Willenbrock R, Anker SD, 335. Pretorius L, Du XJ, Woodcock EA, Kiriazis H, Lin RC, Marasco S, Medcalf RL, Ming Z,
Poole-Wilson PA, Ranke MB, Dietz R. Randomised, double-blind, placebo-controlled Head GA, Tan JW, Cemerlang N, Sadoshima J, Shioi T, Izumo S, Lukoshkova EV, Dart
trial of human recombinant growth hormone in patients with chronic heart failure due AM, Jennings GL, McMullen JR. Reduced phosphoinositide 3-kinase (p110alpha) acti-
to dilated cardiomyopathy. Lancet 351: 1233–1237, 1998. doi:10.1016/S0140- vation increases the susceptibility to atrial fibrillation. Am J Pathol 175: 998 –1009,
6736(97)11329-0. 2009. doi:10.2353/ajpath.2009.090126.

318. Pagan J, Seto T, Pagano M, Cittadini A. Role of the ubiquitin proteasome system in the 336. Puglisi JL, Negroni JA, Chen-Izu Y, Bers DM. The force-frequency relationship: in-
heart. Circ Res 112: 1046 –1058, 2013. doi:10.1161/CIRCRESAHA.112.300521. sights from mathematical modeling. Adv Physiol Educ 37: 28 –34, 2013. doi:10.1152/
advan.00072.2011.
319. Paigen K. One hundred years of mouse genetics: an intellectual history. I. The classical
period (1902-1980). Genetics 163: 1–7, 2003. 337. Puhl SL, Müller A, Wagner M, Devaux Y, Böhm M, Wagner DR, Maack C. Exercise
attenuates inflammation and limits scar thinning after myocardial infarction in mice.
320. Paroo Z, Haist JV, Karmazyn M, Noble EG. Exercise improves postischemic cardiac Am J Physiol Heart Circ Physiol 309: H345–H359, 2015. doi:10.1152/ajpheart.00683.
function in males but not females: consequences of a novel sex-specific heat shock 2014.
protein 70 response. Circ Res 90: 911–917, 2002. doi:10.1161/01.RES.0000016963.
43856.B1. 338. Pullen PR, Nagamia SH, Mehta PK, Thompson WR, Benardot D, Hammoud R, Parrott
JM, Sola S, Khan BV. Effects of yoga on inflammation and exercise capacity in patients
321. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a with chronic heart failure. J Card Fail 14: 407– 413, 2008. doi:10.1016/j.cardfail.2007.
secretory organ. Nat Rev Endocrinol 8: 457– 465, 2012. doi:10.1038/nrendo. 12.007.
2012.49.
339. Pullen PR, Thompson WR, Benardot D, Brandon LJ, Mehta PK, Rifai L, Vadnais DS,
322. Pedersen BK, Saltin B. Evidence for prescribing exercise as therapy in chronic disease. Parrott JM, Khan BV. Benefits of yoga for African American heart failure patients. Med
Scand J Med Sci Sports 16, Suppl 1: 3– 63, 2006. doi:10.1111/j.1600-0838.2006. Sci Sports Exerc 42: 651– 657, 2010. doi:10.1249/MSS.0b013e3181bf24c4.
00520.x.
340. Quindry JC, Hamilton KL, French JP, Lee Y, Murlasits Z, Tumer N, Powers SK.
323. Pelliccia A, Maron BJ, Culasso F, Spataro A, Caselli G. Athlete’s heart in women. Exercise-induced HSP-72 elevation and cardioprotection against infarct and apopto-
Echocardiographic characterization of highly trained elite female athletes. JAMA 276: sis. J Appl Physiol (1985) 103: 1056 –1062, 2007. doi:10.1152/japplphysiol.00263.
211–215, 1996. doi:10.1001/jama.1996.03540030045030. 2007.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 471


BERNARDO ET AL.

341. Ramasamy S, Velmurugan G, Shanmugha Rajan K, Ramprasath T, Kalpana K. MiRNAs 359. Sakamoto M, Minamino T, Toko H, Kayama Y, Zou Y, Sano M, Takaki E, Aoyagi T,
with apoptosis regulating potential are differentially expressed in chronic exercise- Tojo K, Tajima N, Nakai A, Aburatani H, Komuro I. Upregulation of heat shock
induced physiologically hypertrophied hearts. PLoS One 10: e0121401, 2015. doi:10. transcription factor 1 plays a critical role in adaptive cardiac hypertrophy. Circ Res 99:
1371/journal.pone.0121401. 1411–1418, 2006. doi:10.1161/01.RES.0000252345.80198.97.

342. Ratner M. Heart failure gene therapy disappoints but experts keep the faith. Nat 360. Salama G, London B. Mouse models of long QT syndrome. J Physiol 578: 43–53, 2007.
Biotechnol 33: 573–574, 2015. doi:10.1038/nbt0615-573a. doi:10.1113/jphysiol.2006.118745.

343. Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardio- 361. Sand KL, Flatebo T, Andersen MB, Maghazachi AA. Effects of exercise on leukocytosis
vascular Disease. Physiol Rev 97: 1–37, 2017. doi:10.1152/physrev.00021.2015. and blood hemostasis in 800 healthy young females and males. World J Exp Med 3:
11–20, 2013. doi:10.5493/wjem.v3.i1.11.
344. Ren J, Samson WK, Sowers JR. Insulin-like growth factor I as a cardiac hormone:
physiological and pathophysiological implications in heart disease. J Mol Cell Cardiol 31: 362. Santos-Parker JR, LaRocca TJ, Seals DR. Aerobic exercise and other healthy lifestyle
2049 –2061, 1999. doi:10.1006/jmcc.1999.1036. factors that influence vascular aging. Adv Physiol Educ 38: 296 –307, 2014. doi:10.1152/
advan.00088.2014.
345. Reuben DB, Judd-Hamilton L, Harris TB, Seeman TE; MacArthur Studies of Successful
Aging. The associations between physical activity and inflammatory markers in high- 363. Sapra G, Tham YK, Cemerlang N, Matsumoto A, Kiriazis H, Bernardo BC, Henstridge
functioning older persons: MacArthur Studies of Successful Aging. J Am Geriatr Soc 51: DC, Ooi JY, Pretorius L, Boey EJ, Lim L, Sadoshima J, Meikle PJ, Mellet NA, Wood-
1125–1130, 2003. doi:10.1046/j.1532-5415.2003.51380.x. cock EA, Marasco S, Ueyama T, Du XJ, Febbraio MA, McMullen JR. The small-
molecule BGP-15 protects against heart failure and atrial fibrillation in mice. Nat
346. Rhee EP, Gerszten RE. Metabolomics and cardiovascular biomarker discovery. Clin Commun 5: 5705, 2014. doi:10.1038/ncomms6705.
Chem 58: 139 –147, 2012. doi:10.1373/clinchem.2011.169573.
364. Sawada S, Kon M, Wada S, Ushida T, Suzuki K, Akimoto T. Profiling of circulating
347. Riehle C, Wende AR, Zhu Y, Oliveira KJ, Pereira RO, Jaishy BP, Bevins J, Valdez S, Noh microRNAs after a bout of acute resistance exercise in humans. PLoS One 8: e70823,
J, Kim BJ, Moreira AB, Weatherford ET, Manivel R, Rawlings TA, Rech M, White MF, 2013. doi:10.1371/journal.pone.0070823.
Abel ED. Insulin receptor substrates are essential for the bioenergetic and hypertro-
phic response of the heart to exercise training. Mol Cell Biol 34: 3450 –3460, 2014. 365. Schaible TF, Penpargkul S, Scheuer J. Cardiac responses to exercise training in male
doi:10.1128/MCB.00426-14. and female rats. J Appl Physiol Respir Environ Exerc Physiol 50: 112–117, 1981.

348. Ritchie RH, Leo CH, Qin C, Stephenson EJ, Bowden MA, Buxton KD, Lessard SJ, Rivas 366. Schaible TF, Scheuer J. Cardiac function in hypertrophied hearts from chronically
DA, Koch LG, Britton SL, Hawley JA, Woodman OL. Low intrinsic exercise capacity exercised female rats. J Appl Physiol Respir Environ Exerc Physiol 50: 1140 –1145, 1981.
in rats predisposes to age-dependent cardiac remodeling independent of macrovas-
367. Schaible TF, Scheuer J. Effects of physical training by running or swimming on ventric-
cular function. Am J Physiol Heart Circ Physiol 304: H729 –H739, 2013. doi:10.1152/
ular performance of rat hearts. J Appl Physiol Respir Environ Exerc Physiol 46: 854 – 860,
ajpheart.00638.2012.
1979.
349. Ritchie RH, Love JE, Huynh K, Bernardo BC, Henstridge DC, Kiriazis H, Tham YK,
368. Schaper J, Meiser E, Stämmler G. Ultrastructural morphometric analysis of myocar-
Sapra G, Qin C, Cemerlang N, Boey EJ, Jandeleit-Dahm K, Du XJ, McMullen JR.
dium from dogs, rats, hamsters, mice, and from human hearts. Circ Res 56: 377–391,
Enhanced phosphoinositide 3-kinase(p110␣) activity prevents diabetes-induced car-
1985. doi:10.1161/01.RES.56.3.377.
diomyopathy and superoxide generation in a mouse model of diabetes. Diabetologia
55: 3369 –3381, 2012. doi:10.1007/s00125-012-2720-0. 369. Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr
Biol 24: R453–R462, 2014. doi:10.1016/j.cub.2014.03.034.
350. Rodriguez-Miguelez P, Fernandez-Gonzalo R, Almar M, Mejías Y, Rivas A, de Paz JA,
Cuevas MJ, González-Gallego J. Role of Toll-like receptor 2 and 4 signaling pathways 370. Schiekofer S, Shiojima I, Sato K, Galasso G, Oshima Y, Walsh K. Microarray analysis of
on the inflammatory response to resistance training in elderly subjects. Age (Dordr) 36: Akt1 activation in transgenic mouse hearts reveals transcript expression profiles as-
9734, 2014. doi:10.1007/s11357-014-9734-0. sociated with compensatory hypertrophy and failure. Physiol Genomics 27: 156 –170,
2006. doi:10.1152/physiolgenomics.00234.2005.
351. Rohrer DK, Schauble EH, Desai KH, Kobilka BK, Bernstein D. Alterations in dynamic
heart rate control in the beta 1-adrenergic receptor knockout mouse. Am J Physiol 371. Schuler G, Adams V, Goto Y. Role of exercise in the prevention of cardiovascular
Heart Circ Physiol 274: H1184 –H1193, 1998. disease: results, mechanisms, and new perspectives. Eur Heart J 34: 1790 –1799,
2013. doi:10.1093/eurheartj/eht111.
352. Rönn T, Volkov P, Davegårdh C, Dayeh T, Hall E, Olsson AH, Nilsson E, Tornberg A,
Dekker Nitert M, Eriksson KF, Jones HA, Groop L, Ling C. A six months exercise 372. Seddon M, Shah AM, Casadei B. Cardiomyocytes as effectors of nitric oxide signalling.
intervention influences the genome-wide DNA methylation pattern in human adipose Cardiovasc Res 75: 315–326, 2007. doi:10.1016/j.cardiores.2007.04.031.
tissue. PLoS Genet 9: e1003572, 2013. doi:10.1371/journal.pgen.1003572.
373. Senyo SE, Steinhauser ML, Pizzimenti CL, Yang VK, Cai L, Wang M, Wu TD, Guer-
353. Roof SR, Ho HT, Little SC, Ostler JE, Brundage EA, Periasamy M, Villamena FA, quin-Kern JL, Lechene CP, Lee RT. Mammalian heart renewal by pre-existing cardi-
Györke S, Biesiadecki BJ, Heymes C, Houser SR, Davis JP, Ziolo MT. Obligatory role omyocytes. Nature 493: 433– 436, 2013. doi:10.1038/nature11682.
of neuronal nitric oxide synthase in the heart’s antioxidant adaptation with exercise. J
Mol Cell Cardiol 81: 54 – 61, 2015. doi:10.1016/j.yjmcc.2015.01.003. 374. Sergeeva IA, Christoffels VM. Regulation of expression of atrial and brain natriuretic
peptide, biomarkers for heart development and disease. Biochim Biophys Acta 1832:
354. Roof SR, Tang L, Ostler JE, Periasamy M, Györke S, Billman GE, Ziolo MT. Neuronal 2403–2413, 2013. doi:10.1016/j.bbadis.2013.07.003.
nitric oxide synthase is indispensable for the cardiac adaptive effects of exercise. Basic
Res Cardiol 108: 332, 2013. doi:10.1007/s00395-013-0332-6. 375. Serra AJ, Santos MH, Bocalini DS, Antônio EL, Levy RF, Santos AA, Higuchi ML, Silva
JA, Jr, Magalhães FC, Baraúna VG, Krieger JE, Tucci PJ. Exercise training inhibits
355. Rosen CJ, Kurland ES, Vereault D, Adler RA, Rackoff PJ, Craig WY, Witte S, Rogers J, inflammatory cytokines and more than prevents myocardial dysfunction in rats with
Bilezikian JP. Association between serum insulin growth factor-I (IGF-I) and a simple sustained beta-adrenergic hyperactivity. J Physiol 588: 2431–2442, 2010. doi:10.1113/
sequence repeat in IGF-I gene: implications for genetic studies of bone mineral den- jphysiol.2010.187310.
sity. J Clin Endocrinol Metab 83: 2286 –2290, 1998. doi:10.1210/jcem.83.7.4964.
376. Shah SH, Kraus WE, Newgard CB. Metabolomic profiling for the identification of
356. Rosenbloom J, Mendoza FA, Jimenez SA. Strategies for anti-fibrotic therapies. Biochim novel biomarkers and mechanisms related to common cardiovascular diseases: form
Biophys Acta 1832: 1088 –1103, 2013. doi:10.1016/j.bbadis.2012.12.007. and function. Circulation 126: 1110 –1120, 2012. doi:10.1161/CIRCULATIONAHA.
111.060368.
357. Sadoshima J. Redox regulation of growth and death in cardiac myocytes. Antioxid
Redox Signal 8: 1621–1624, 2006. doi:10.1089/ars.2006.8.1621. 377. Shammas MA. Telomeres, lifestyle, cancer, and aging. Curr Opin Clin Nutr Metab Care
14: 28 –34, 2011. doi:10.1097/MCO.0b013e32834121b1.
358. Safdar A, Saleem A, Tarnopolsky MA. The potential of endurance exercise-derived
exosomes to treat metabolic diseases. Nat Rev Endocrinol 12: 504 –517, 2016. doi:10. 378. Shan J, Kushnir A, Betzenhauser MJ, Reiken S, Li J, Lehnart SE, Lindegger N, Mongillo
1038/nrendo.2016.76. M, Mohler PJ, Marks AR. Phosphorylation of the ryanodine receptor mediates the

472 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

cardiac fight or flight response in mice. J Clin Invest 120: 4388 – 4398, 2010. doi:10. 396. Strøm CC, Aplin M, Ploug T, Christoffersen TEH, Langfort J, Viese M, Galbo H,
1172/JCI32726. Haunsø S, Sheikh SP. Expression profiling reveals differences in metabolic gene ex-
pression between exercise-induced cardiac effects and maladaptive cardiac hypertro-
379. Sharma V, Marsh R, Cunniffe B, Cardinale M, Yellon DM, Davidson SM. From Pro- phy. FEBS J 272: 2684 –2695, 2005. doi:10.1111/j.1742-4658.2005.04684.x.
tecting the Heart to Improving Athletic Performance - the Benefits of Local and
Remote Ischaemic Preconditioning. Cardiovasc Drugs Ther 29: 573–588, 2015. doi:10. 397. Sturmberg JP, Bennett JM, Picard M, Seely AJ. The trajectory of life. Decreasing
1007/s10557-015-6621-6. physiological network complexity through changing fractal patterns. Front Physiol 6:
169, 2015. doi:10.3389/fphys.2015.00169.
380. Shi J, Bei Y, Kong X, Liu X, Lei Z, Xu T, Wang H, Xuan Q, Chen P, Xu J, Che L, Liu H,
Zhong J, Sluijter JP, Li X, Rosenzweig A, Xiao J. miR-17-3p Contributes to Exercise- 398. Su F, Zhao L, Zhang S, Wang J, Chen N, Gong Q, Tang J, Wang H, Yao J, Wang Q,
Induced Cardiac Growth and Protects against Myocardial Ischemia-Reperfusion In- Zhong M, Yan J. Cardioprotection by PI3K-mediated signaling is required for anti-
jury. Theranostics 7: 664 – 676, 2017. doi:10.7150/thno.15162. arrhythmia and myocardial repair in response to ischemic preconditioning in infarcted
pig hearts. Lab Invest 95: 860 – 871, 2015. doi:10.1038/labinvest.2015.64.
381. Shiber A, Ravid T. Chaperoning proteins for destruction: diverse roles of Hsp70
chaperones and their co-chaperones in targeting misfolded proteins to the protea- 399. Sullivan MJ, Cobb FR, Higginbotham MB. Stroke volume increases by similar mecha-
some. Biomolecules 4: 704 –724, 2014. doi:10.3390/biom4030704. nisms during upright exercise in normal men and women. Am J Cardiol 67: 1405–1412,
1991. doi:10.1016/0002-9149(91)90472-W.
382. Shioi T, Kang PM, Douglas PS, Hampe J, Yballe CM, Lawitts J, Cantley LC, Izumo S.
The conserved phosphoinositide 3-kinase pathway determines heart size in mice. 400. Sun GW, Qiu ZD, Wang WN, Sui X, Sui DJ. Flavonoids Extraction from Propolis
EMBO J 19: 2537–2548, 2000. doi:10.1093/emboj/19.11.2537. Attenuates Pathological Cardiac Hypertrophy through PI3K/AKT Signaling Pathway.
Evid Based Complement Alternat Med 2016: 6281376, 2016. doi:10.1155/2016/
383. Shiojima I, Sato K, Izumiya Y, Schiekofer S, Ito M, Liao R, Colucci WS, Walsh K. 6281376.
Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the
transition to heart failure. J Clin Invest 115: 2108 –2118, 2005. doi:10.1172/JCI24682. 401. Sun Z, Hamilton KL, Reardon KF. Phosphoproteomics and molecular cardiology:
techniques, applications and challenges. J Mol Cell Cardiol 53: 354 –368, 2012. doi:10.
384. Shiraishi I, Melendez J, Ahn Y, Skavdahl M, Murphy E, Welch S, Schaefer E, Walsh K, 1016/j.yjmcc.2012.06.001.
Rosenzweig A, Torella D, Nurzynska D, Kajstura J, Leri A, Anversa P, Sussman MA.
Nuclear targeting of Akt enhances kinase activity and survival of cardiomyocytes. Circ 402. Tam BT, Pei XM, Yung BY, Yip SP, Chan LW, Wong CS, Siu PM. Autophagic Adap-
Res 94: 884 – 891, 2004. doi:10.1161/01.RES.0000124394.01180.BE. tations to Long-term Habitual Exercise in Cardiac Muscle. Int J Sports Med 36: 526 –
534, 2015. doi:10.1055/s-0034-1398494.
385. Simpson RJ, Kunz H, Agha N, Graff R. Exercise and the Regulation of Immune Func-
403. Tanasescu M, Leitzmann MF, Rimm EB, Willett WC, Stampfer MJ, Hu FB. Exercise
tions. Prog Mol Biol Transl Sci 135: 355–380, 2015. doi:10.1016/bs.pmbts.2015.08.
type and intensity in relation to coronary heart disease in men. JAMA 288: 1994 –2000,
001.
2002. doi:10.1001/jama.288.16.1994.
386. Siu PM, Bryner RW, Martyn JK, Alway SE. Apoptotic adaptations from exercise train-
404. Taneike M, Yamaguchi O, Nakai A, Hikoso S, Takeda T, Mizote I, Oka T, Tamai T,
ing in skeletal and cardiac muscles. FASEB J 18: 1150 –1152, 2004. doi:10.1096/fj.03-
Oyabu J, Murakawa T, Nishida K, Shimizu T, Hori M, Komuro I, Takuji Shirasawa TS,
1291fje.
Mizushima N, Otsu K. Inhibition of autophagy in the heart induces age-related car-
387. Soci UP, Fernandes T, Barauna VG, Hashimoto NY, Mota GF, Rosa KT, Irigoyen MC, diomyopathy. Autophagy 6: 600 – 606, 2010. doi:10.4161/auto.6.5.11947.
Phillips MI, de Oliveira EM. Epigenetic control of exercise training-induced cardiac
405. Tao L, Bei Y, Lin S, Zhang H, Zhou Y, Jiang J, Chen P, Shen S, Xiao J, Li X. Exercise
hypertrophy by miR-208. Clin Sci (Lond) CS20160480, 2016. doi:10.1042/
Training Protects Against Acute Myocardial Infarction via Improving Myocardial En-
CS20160480.
ergy Metabolism and Mitochondrial Biogenesis. Cell Physiol Biochem 37: 162–175,
388. Soci UP, Fernandes T, Hashimoto NY, Mota GF, Amadeu MA, Rosa KT, Irigoyen MC, 2015. doi:10.1159/000430342.
Phillips MI, Oliveira EM. MicroRNAs 29 are involved in the improvement of ventric-
406. Taylor RP, Harris MB, Starnes JW. Acute exercise can improve cardioprotection
ular compliance promoted by aerobic exercise training in rats. Physiol Genomics 43:
without increasing heat shock protein content. Am J Physiol Heart Circ Physiol 276:
665– 673, 2011. doi:10.1152/physiolgenomics.00145.2010.
H1098 –H1102, 1999.
389. Song HK, Hong SE, Kim T, Kim DH. Deep RNA sequencing reveals novel cardiac
407. Thannickal VJ, Zhou Y, Gaggar A, Duncan SR. Fibrosis: ultimate and proximate causes.
transcriptomic signatures for physiological and pathological hypertrophy. PLoS One 7: J Clin Invest 124: 4673– 4677, 2014. doi:10.1172/JCI74368.
e35552, 2012. doi:10.1371/journal.pone.0035552.
408. Thomas DP, Cotter TA, Li X, McCormick RJ, Gosselin LE. Exercise training attenu-
390. Soonpaa MH, Field LJ. Survey of studies examining mammalian cardiomyocyte DNA ates aging-associated increases in collagen and collagen crosslinking of the left but not
synthesis. Circ Res 83: 15–26, 1998. doi:10.1161/01.RES.83.1.15. the right ventricle in the rat. Eur J Appl Physiol 85: 164 –169, 2001. doi:10.1007/
s004210100447.
391. Soonpaa MH, Kim KK, Pajak L, Franklin M, Field LJ. Cardiomyocyte DNA synthesis
and binucleation during murine development. Am J Physiol Heart Circ Physiol 271: 409. Thomas DP, Zimmerman SD, Hansen TR, Martin DT, McCormick RJ. Collagen gene
H2183–H2189, 1996. expression in rat left ventricle: interactive effect of age and exercise training. J Appl
Physiol (1985) 89: 1462–1468, 2000.
392. Soto PF, Herrero P, Schechtman KB, Waggoner AD, Baumstark JM, Ehsani AA,
Gropler RJ. Exercise training impacts the myocardial metabolism of older individuals in 410. Thorp DB, Haist JV, Leppard J, Milne KJ, Karmazyn M, Noble EG. Exercise training
a gender-specific manner. Am J Physiol Heart Circ Physiol 295: H842–H850, 2008. improves myocardial tolerance to ischemia in male but not in female rats. Am J
doi:10.1152/ajpheart.91426.2007. Physiol Regul Integr Comp Physiol 293: R363–R371, 2007. doi:10.1152/ajpregu.
00363.2006.
393. Souza RW, Fernandez GJ, Cunha JP, Piedade WP, Soares LC, Souza PA, de Campos
DH, Okoshi K, Cicogna AC, Dal-Pai-Silva M, Carvalho RF. Regulation of cardiac 411. Tian C, Zucker IH. Therapeutic microRNA-based strategies in cardiovascular disease
microRNAs induced by aerobic exercise training during heart failure. Am J Physiol discriminate sex and age difference. J Physiol 594: 5731–5732, 2016. doi:10.1113/
Heart Circ Physiol 309: H1629 –H1641, 2015. doi:10.1152/ajpheart.00941.2014. JP272892.

394. Spirito P, Pelliccia A, Proschan MA, Granata M, Spataro A, Bellone P, Caselli G, Biffi A, 412. Tibbits GF, Kashihara H, O’Reilly K. Na⫹-Ca2⫹ exchange in cardiac sarcolemma:
Vecchio C, Maron BJ. Morphology of the “athlete’s heart” assessed by echocardiog- modulation of Ca2⫹ affinity by exercise. Am J Physiol Cell Physiol 256: C638 –C643,
raphy in 947 elite athletes representing 27 sports. Am J Cardiol 74: 802– 806, 1994. 1989.
doi:10.1016/0002-9149(94)90439-1.
413. Tilemann L, Lee A, Ishikawa K, Aguero J, Rapti K, Santos-Gallego C, Kohlbrenner E,
395. Stanley WC, Chandler MP. Energy metabolism in the normal and failing heart: poten- Fish KM, Kho C, Hajjar RJ. SUMO-1 gene transfer improves cardiac function in a
tial for therapeutic interventions. Heart Fail Rev 7: 115–130, 2002. doi:10.1023/ large-animal model of heart failure. Sci Transl Med 5: 211ra159, 2013. doi:10.1126/
A:1015320423577. scitranslmed.3006487.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 473


BERNARDO ET AL.

414. Tinker A, Aziz Q, Thomas A. The role of ATP-sensitive potassium channels in cellular 432. Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, Pairaudeau
function and protection in the cardiovascular system. Br J Pharmacol 171: 12–23, 2014. G, Pennie WD, Pickett SD, Wang J, Wallace O, Weir A. An analysis of the attrition of
doi:10.1111/bph.12407. drug candidates from four major pharmaceutical companies. Nat Rev Drug Discov 14:
475– 486, 2015. doi:10.1038/nrd4609.
415. Touvron M, Escoubet B, Mericskay M, Angelini A, Lamotte L, Santini MP, Rosenthal
N, Daegelen D, Tuil D, Decaux JF. Locally expressed IGF1 propeptide improves 433. Weeks KL, Avkiran M. Roles and post-translational regulation of cardiac class IIa
mouse heart function in induced dilated cardiomyopathy by blocking myocardial fi- histone deacetylase isoforms. J Physiol 593: 1785–1797, 2015. doi:10.1113/jphysiol.
brosis and SRF-dependent CTGF induction. Dis Model Mech 5: 481– 491, 2012. doi: 2014.282442.
10.1242/dmm.009456.
434. Weeks KL, Gao X, Du XJ, Boey EJ, Matsumoto A, Bernardo BC, Kiriazis H, Cemer-
416. Travers JG, Kamal FA, Robbins J, Yutzey KE, Blaxall BC. Cardiac Fibrosis. Circ Res 118: lang N, Tan JW, Tham YK, Franke TF, Qian H, Bogoyevitch MA, Woodcock EA,
1021–1040, 2016. doi:10.1161/CIRCRESAHA.115.306565. Febbraio MA, Gregorevic P, McMullen JR. Phosphoinositide 3-kinase p110␣ is a
master regulator of exercise-induced cardioprotection and PI3K gene therapy res-
417. Trépanier-Boulay V, St-Michel C, Tremblay A, Fiset C. Gender-based differences in cues cardiac dysfunction. Circ Heart Fail 5: 523–534, 2012. doi:10.1161/
cardiac repolarization in mouse ventricle. Circ Res 89: 437– 444, 2001. doi:10.1161/ CIRCHEARTFAILURE.112.966622.
hh1701.095644.
435. Weeks KL, McMullen JR. The athlete’s heart vs. the failing heart: can signaling explain
418. Trost SG, Owen N, Bauman AE, Sallis JF, Brown W. Correlates of adults’ participation the two distinct outcomes? Physiology (Bethesda) 26: 97–105, 2011. doi:10.1152/
in physical activity: review and update. Med Sci Sports Exerc 34: 1996 –2001, 2002. physiol.00043.2010.
doi:10.1097/00005768-200212000-00020.
436. Wei X, Liu X, Rosenzweig A. What do we know about the cardiac benefits of exer-
419. Tsarouhas K, Tsitsimpikou C, Haliassos A, Georgoulias P, Koutsioras I, Kouretas D, cise? Trends Cardiovasc Med 25: 529 –536, 2015. doi:10.1016/j.tcm.2014.12.014.
Kogias J, Liosis I, Rentoukas E, Kyriakides Z. Study of insulin resistance, TNF-␣, total
antioxidant capacity and lipid profile in patients with chronic heart failure under 437. Welch S, Plank D, Witt S, Glascock B, Schaefer E, Chimenti S, Andreoli AM, Limana F,
exercise. In Vivo 25: 1031–1037, 2011. Leri A, Kajstura J, Anversa P, Sussman MA. Cardiac-specific IGF-1 expression atten-
uates dilated cardiomyopathy in tropomodulin-overexpressing transgenic mice. Circ
420. Usui S, Maejima Y, Pain J, Hong C, Cho J, Park JY, Zablocki D, Tian B, Glass DJ, Res 90: 641– 648, 2002. doi:10.1161/01.RES.0000013780.77774.75.
Sadoshima J. Endogenous muscle atrophy F-box mediates pressure overload-induced
cardiac hypertrophy through regulation of nuclear factor-kappaB. Circ Res 109: 161– 438. Werner C, Hanhoun M, Widmann T, Kazakov A, Semenov A, Pöss J, Bauersachs J,
Thum T, Pfreundschuh M, Müller P, Haendeler J, Böhm M, Laufs U. Effects of physical
171, 2011. doi:10.1161/CIRCRESAHA.110.238717.
exercise on myocardial telomere-regulating proteins, survival pathways, and apopto-
421. Vaessen N, Heutink P, Janssen JA, Witteman JC, Testers L, Hofman A, Lamberts SW, sis. J Am Coll Cardiol 52: 470 – 482, 2008. doi:10.1016/j.jacc.2008.04.034.
Oostra BA, Pols HA, van Duijn CM. A polymorphism in the gene for IGF-I: functional
439. Wettschureck N, Rütten H, Zywietz A, Gehring D, Wilkie TM, Chen J, Chien KR,
properties and risk for type 2 diabetes and myocardial infarction. Diabetes 50: 637–
Offermanns S. Absence of pressure overload induced myocardial hypertrophy after
642, 2001. doi:10.2337/diabetes.50.3.637.
conditional inactivation of Galphaq/Galpha11 in cardiomyocytes. Nat Med 7: 1236 –
422. Valiente-Alandi I, Schafer AE, Blaxall BC. Extracellular matrix-mediated cellular com- 1240, 2001. doi:10.1038/nm1101-1236.
munication in the heart. J Mol Cell Cardiol 91: 228 –237, 2016. doi:10.1016/j.yjmcc.
440. Wheatley CM, Snyder EM, Johnson BD, Olson TP. Sex differences in cardiovascular
2016.01.011.
function during submaximal exercise in humans. Springerplus 3: 445, 2014. doi:10.
423. Van Putten S, Shafieyan Y, Hinz B. Mechanical control of cardiac myofibroblasts. J Mol 1186/2193-1801-3-445.
Cell Cardiol 93: 133–142, 2016. doi:10.1016/j.yjmcc.2015.11.025.
441. Whelan RS, Konstantinidis K, Xiao RP, Kitsis RN. Cardiomyocyte life-death decisions
424. Vanzelli AS, Medeiros A, Rolim N, Bartholomeu JB, Cunha TF, Bechara LR, Gomes in response to chronic ␤-adrenergic signaling. Circ Res 112: 408 – 410, 2013. doi:10.
ER, Mattos KC, Sirvente R, Salemi VM, Mady C, Negrao CE, Guatimosim S, Brum PC. 1161/CIRCRESAHA.113.300805.
Integrative effect of carvedilol and aerobic exercise training therapies on improving
442. White FC, Bloor CM, McKirnan MD, Carroll SM. Exercise training in swine promotes
cardiac contractility and remodeling in heart failure mice. PLoS One 8: e62452, 2013.
growth of arteriolar bed and capillary angiogenesis in heart. J Appl Physiol (1985) 85:
doi:10.1371/journal.pone.0062452. 1160 –1168, 1998.
425. Vatner SF. Why So Few New Cardiovascular Drugs Translate to the Clinics. Circ Res 443. Willis MS, Min JN, Wang S, McDonough H, Lockyer P, Wadosky KM, Patterson C.
119: 714 –717, 2016. doi:10.1161/CIRCRESAHA.116.309512. Carboxyl terminus of Hsp70-interacting protein (CHIP) is required to modulate car-
diac hypertrophy and attenuate autophagy during exercise. Cell Biochem Funct 31:
426. Vega RB, Harrison BC, Meadows E, Roberts CR, Papst PJ, Olson EN, McKinsey TA.
724 –735, 2013. doi:10.1002/cbf.2962.
Protein kinases C and D mediate agonist-dependent cardiac hypertrophy through
nuclear export of histone deacetylase 5. Mol Cell Biol 24: 8374 – 8385, 2004. doi:10. 444. Willis MS, Patterson C. Hold me tight: role of the heat shock protein family of chap-
1128/MCB.24.19.8374-8385.2004. erones in cardiac disease. Circulation 122: 1740 –1751, 2010. doi:10.1161/
CIRCULATIONAHA.110.942250.
427. Ventura-Clapier R, Mettauer B, Bigard X. Beneficial effects of endurance training on
cardiac and skeletal muscle energy metabolism in heart failure. Cardiovasc Res 73: 445. Wilson MG, Ellison GM, Cable NT. Basic science behind the cardiovascular benefits of
10 –18, 2007. doi:10.1016/j.cardiores.2006.09.003. exercise. Heart 101: 758 –765, 2015. doi:10.1136/heartjnl-2014-306596.

428. Vinciguerra M, Santini MP, Martinez C, Pazienza V, Claycomb WC, Giuliani A, 446. Wisløff U, Loennechen JP, Currie S, Smith GL, Ellingsen Ø. Aerobic exercise reduces
Rosenthal N. mIGF-1/JNK1/SirT1 signaling confers protection against oxidative stress cardiomyocyte hypertrophy and increases contractility, Ca2⫹ sensitivity and SERCA-2
in the heart. Aging Cell 11: 139 –149, 2012. doi:10.1111/j.1474-9726.2011.00766.x. in rat after myocardial infarction. Cardiovasc Res 54: 162–174, 2002. doi:10.1016/
S0008-6363(01)00565-X.
429. Völkers M, Toko H, Doroudgar S, Din S, Quijada P, Joyo AY, Ornelas L, Joyo E,
Thuerauf DJ, Konstandin MH, Gude N, Glembotski CC, Sussman MA. Pathological 447. Wisløff U, Loennechen JP, Falck G, Beisvag V, Currie S, Smith G, Ellingsen O. In-
hypertrophy amelioration by PRAS40-mediated inhibition of mTORC1. Proc Natl creased contractility and calcium sensitivity in cardiac myocytes isolated from endur-
Acad Sci USA 110: 12661–12666, 2013. doi:10.1073/pnas.1301455110. ance trained rats. Cardiovasc Res 50: 495–508, 2001. doi:10.1016/S0008-
6363(01)00210-3.
430. Wang C, Wang X. The interplay between autophagy and the ubiquitin-proteasome
system in cardiac proteotoxicity. Biochim Biophys Acta 1852: 188 –194, 2015. doi:10. 448. Wisløff U, Najjar SM, Ellingsen O, Haram PM, Swoap S, Al-Share Q, Fernström M,
1016/j.bbadis.2014.07.028. Rezaei K, Lee SJ, Koch LG, Britton SL. Cardiovascular risk factors emerge after
artificial selection for low aerobic capacity. Science 307: 418 – 420, 2005. doi:10.1126/
431. Waring CD, Vicinanza C, Papalamprou A, Smith AJ, Purushothaman S, Goldspink DF, science.1108177.
Nadal-Ginard B, Torella D, Ellison GM. The adult heart responds to increased work-
load with physiologic hypertrophy, cardiac stem cell activation, and new myocyte 449. Wisløff U, Støylen A, Loennechen JP, Bruvold M, Rognmo Ø, Haram PM, Tjønna AE,
formation. Eur Heart J 35: 2722–2731, 2014. doi:10.1093/eurheartj/ehs338. Helgerud J, Slørdahl SA, Lee SJ, Videm V, Bye A, Smith GL, Najjar SM, Ellingsen Ø,

474 Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org


EXERCISE-INDUCED CARDIAC PROTECTION

Skjaerpe T. Superior cardiovascular effect of aerobic interval training versus moderate 460. Yeh JK, Aloia JF, Chen M, Ling N, Koo HC, Millard WJ. Effect of growth hormone
continuous training in heart failure patients: a randomized study. Circulation 115: administration and treadmill exercise on serum and skeletal IGF-I in rats. Am J Physiol
3086 –3094, 2007. doi:10.1161/CIRCULATIONAHA.106.675041. Endocrinol Metab 266: E129 –E135, 1994.

450. Wu J, Ruas JL, Estall JL, Rasbach KA, Choi JH, Ye L, Boström P, Tyra HM, Crawford 461. Zhang C, Xu Z, He XR, Michael LH, Patterson C. CHIP, a cochaperone/ubiquitin
RW, Campbell KP, Rutkowski DT, Kaufman RJ, Spiegelman BM. The unfolded protein ligase that regulates protein quality control, is required for maximal cardioprotection
response mediates adaptation to exercise in skeletal muscle through a PGC-1␣/ after myocardial infarction in mice. Am J Physiol Heart Circ Physiol 288: H2836 –H2842,
ATF6␣ complex. Cell Metab 13: 160 –169, 2011. doi:10.1016/j.cmet.2011.01.003. 2005. doi:10.1152/ajpheart.01122.2004.

451. Wu Y, Anderson ME. CaMKII in sinoatrial node physiology and dysfunction. Front 462. Zhang J, Wang C, Yu S, Luo Z, Chen Y, Liu Q, Hua F, Xu G, Yu P. Sevoflurane
Pharmacol 5: 48, 2014. doi:10.3389/fphar.2014.00048. postconditioning protects rat hearts against ischemia-reperfusion injury via the
activation of PI3K/AKT/mTOR signaling. Sci Rep 4: 7317, 2014. doi:10.1038/
452. Xiao J, Xu T, Li J, Lv D, Chen P, Zhou Q, Xu J. Exercise-induced physiological
srep07317.
hypertrophy initiates activation of cardiac progenitor cells. Int J Clin Exp Pathol 7:
663– 669, 2014. 463. Zhang M, Shah AM. ROS signalling between endothelial cells and cardiac cells. Car-
diovasc Res 102: 249 –257, 2014. doi:10.1093/cvr/cvu050.
453. Xie J, He G, Chen Q, Sun J, Dai Q, Lu J, Li G, Wu H, Li R, Chen J, Xu W, Xu B.
Syndecan 4 signaling is required for exercise-induced cardiac hypertrophy. Mol Med
464. Zhang W, Xing B, Yang L, Shi J, Zhou X. Icaritin Attenuates Myocardial Ischemia and
22: 1, 2016. doi:10.2119/molmed.2015.00026.
Reperfusion Injury Via Anti-Inflammatory and Anti-Oxidative Stress Effects in Rats. Am
454. Yang DK, Choi BY, Lee YH, Kim YG, Cho MC, Hong SE, Kim DH, Hajjar RJ, Park WJ. J Chin Med 43: 1083–1097, 2015. doi:10.1142/S0192415X15500627.
Gene profiling during regression of pressure overload-induced cardiac hypertrophy.
465. Zhou P, Pu WT. Recounting Cardiac Cellular Composition. Circ Res 118: 368 –370,
Physiol Genomics 30: 1–7, 2007. doi:10.1152/physiolgenomics.00246.2006.
2016. doi:10.1161/CIRCRESAHA.116.308139.
455. Yang KC, Foeger NC, Marionneau C, Jay PY, McMullen JR, Nerbonne JM. Homeo-
466. Ziaeian B, Fonarow GC. Epidemiology and aetiology of heart failure. Nat Rev Cardiol
static regulation of electrical excitability in physiological cardiac hypertrophy. J Physiol
13: 368 –378, 2016. doi:10.1038/nrcardio.2016.25.
588: 5015–5032, 2010. doi:10.1113/jphysiol.2010.197418.
467. Zimmer P, Bloch W. Physical exercise and epigenetic adaptations of the cardiovascu-
456. Yang KC, Jay PY, McMullen JR, Nerbonne JM. Enhanced cardiac PI3K␣ signalling
mitigates arrhythmogenic electrical remodelling in pathological hypertrophy and lar system. Herz 40: 353–360, 2015. doi:10.1007/s00059-015-4213-7.
heart failure. Cardiovasc Res 93: 252–262, 2012. doi:10.1093/cvr/cvr283.
468. Zingman LV, Hodgson DM, Bast PH, Kane GC, Perez-Terzic C, Gumina RJ, Pucar D,
457. Yang L, Li Y, Wang X, Mu X, Qin D, Huang W, Alshahrani S, Nieman M, Peng J, Bienengraeber M, Dzeja PP, Miki T, Seino S, Alekseev AE, Terzic A. Kir6.2 is required
Essandoh K, Peng T, Wang Y, Lorenz J, Soleimani M, Zhao ZQ, Fan GC. Overexpres- for adaptation to stress. Proc Natl Acad Sci USA 99: 13278 –13283, 2002. doi:10.1073/
sion of miR-223 Tips the Balance of Pro- and Anti-hypertrophic Signaling Cascades pnas.212315199.
toward Physiologic Cardiac Hypertrophy. J Biol Chem 291: 15700 –15713, 2016. doi:
469. Zingman LV, Zhu Z, Sierra A, Stepniak E, Burnett CM, Maksymov G, Anderson
10.1074/jbc.M116.715805.
ME, Coetzee WA, Hodgson-Zingman DM. Exercise-induced expression of car-
458. Yang Y, Cheng HW, Qiu Y, Dupee D, Noonan M, Lin YD, Fisch S, Unno K, Sereti KI, Liao diac ATP-sensitive potassium channels promotes action potential shortening and
R. MicroRNA-34a Plays a Key Role in Cardiac Repair and Regeneration Following Myocardial energy conservation. J Mol Cell Cardiol 51: 72– 81, 2011. doi:10.1016/j.yjmcc.
Infarction. Circ Res 117: 450–459, 2015. doi:10.1161/CIRCRESAHA.117.305962. 2011.03.010.

459. Yasuda S, Goto Y, Takaki H, Asaumi Y, Baba T, Miyazaki S, Nonogi H. Exercise- 470. Zsebo K, Yaroshinsky A, Rudy JJ, Wagner K, Greenberg B, Jessup M, Hajjar RJ. Long-
induced hepatocyte growth factor production in patients after acute myocardial in- term effects of AAV1/SERCA2a gene transfer in patients with severe heart failure:
farction: its relationship to exercise capacity and brain natriuretic peptide levels. Circ analysis of recurrent cardiovascular events and mortality. Circ Res 114: 101–108,
J 68: 304 –307, 2004. doi:10.1253/circj.68.304. 2014. doi:10.1161/CIRCRESAHA.113.302421.

Physiol Rev • VOL 98 • JANUARY 2018 • www.prv.org 475

You might also like