You are on page 1of 63

 

 
MicroRNAs as Important Regulators of Exercise Adaptation

Gustavo J.J. Silva, Anja Bye, Hamid el Azzouzi, Ulrik Wisløff

PII: S0033-0620(17)30090-7
DOI: doi: 10.1016/j.pcad.2017.06.003
Reference: YPCAD 821

To appear in: Progress in Cardiovascular Diseases

Received date: 25 June 2017


Accepted date: 25 June 2017

Please cite this article as: Silva Gustavo J.J., Bye Anja, el Azzouzi Hamid, Wisløff Ulrik,
MicroRNAs as Important Regulators of Exercise Adaptation, Progress in Cardiovascular
Diseases (2017), doi: 10.1016/j.pcad.2017.06.003

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
ACCEPTED MANUSCRIPT

MicroRNAs as Important Regulators of Exercise Adaptation

PT
RI
Gustavo J. J. Silvaa, Anja Byea, Hamid el Azzouzib, Ulrik Wisløffa,c*

SC
aK.G. Jebsen Center of Exercise in Medicine at the Department of Circulation and

NU
Medical Imaging, Norwegian University of Science and Technology (NTNU),
Trondheim, Norway.
bDepartment of Cardiology, University Medical Center Utrecht-UMCU, Utrecht,
MA
The Netherlands.
cSchool of Human Movement & Nutrition Sciences, University of Queensland,

Australia.
ED

Short Title: MicroRNA and Exercise


Disclosures/COI: None
PT
CE
AC

* Address reprint requests and correspondence to: Ulrik Wisløff, K. G. Jebsen


Center of Exercise in Medicine, Norwegian University of Science and Technology
(NTNU). Prinsesse Kristinas gt. 3, 3rd floor, 7006 Trondheim, Norway.
E-mail address: ulrik.wisloff@ntnu.no (U. Wisløff)
ACCEPTED MANUSCRIPT
2

Abstract

A significant body of evidence supports the protective role of exercise training

PT
(ET) in cardiovascular diseases, skeletal muscle dystrophies, several types of

RI
cancer, Alzheimer disease or even in the recovery of spinal cord injury. In spite

of this, the molecular mechanisms underlying the beneficial effects of exercise

SC
training are not well understood and remain elusive. Several mechanisms have

NU
been proposed in the past, but more recently microRNAs (miRNAs), small non-

coding RNA molecules involved in a variety of basic biological processes that


MA
negatively modulate gene expression, recognized as important regulatory

molecules. In this review, we highlight recent advances on the miRNA


ED

involvement in the benefits of ET. Here, we assess the role of microRNAs

expressed in the heart, in the skeletal muscle, detected in the circulation (serum
PT

and plasma), and in other conditions (e.g., spinal cord injury). Additionally, the
CE

long-term effects of diverse ET modalities (e.g., running, cycling, resistance

training) in the cardiac miRNA profile are properly addressed.


AC

Keywords: microRNA, exercise training, heart, skeletal muscle, spinal cord


injury, brain

Abbreviations and Acronyms

AUC = Area under the curve

CAD = Coronary artery disease

CRF = Cardiorespiratory fitness


ACCEPTED MANUSCRIPT
3

CV = Cardiovascular

CVD = Cardiovascular disease

ET = Exercise training

PT
HF = Heart failure

hsa = Homo sapiens (human)

RI
HUNT = The Nord-Trøndelag Health Study

SC
HF = Heart failure

KEGG = Kyoto Encyclopedia of Genes and Genomes

dKO = Double knockout


NU
MA
LNA = Locked nucleic acid

LV = Left ventricular
ED

LVH = Left ventricular hypertrophy

mmu = Mus musculus (mouse)


PT

miRNA = MicroRNA
CE

myomiRNA = Skeletal muscle microRNA

ncRNA = noncoding RNA


AC

NK = Natural killer cells

pre-miRNA = Precursor-miRNA

pri-miRNA = Primary microRNA transcripts

PA = Physical activity

Pmax = maximum skeletal muscle force

RAAS = Renin-angiotensin-aldosterone system

RIPC = Remote ischemic preconditioning

rno = Rattus norvegicus (rat)

ROC = Receiver Operating Characteristic


ACCEPTED MANUSCRIPT
4

RT = Resistance training

SHR = Spontaneously hypertensive rats

SNP = Single nucleotide polymorphism

PT
UTR = 3' untranslated region

VO2max = Maximal oxygen uptake

RI
SC
NU
MA
ED
PT
CE
AC
ACCEPTED MANUSCRIPT
5

Exercise Training

The lack of physical activity (PA) contributes to the worldwide rise in life-style

related diseases, such as cardiovascular (CV) disease (CVD) and metabolic

PT
diseases, and diabetes1. In line with this observation, large-scale epidemiological

studies performed in healthy subjects and in patients with CVD demonstrate that

RI
low aerobic exercise capacity is a stronger predictor of mortality than other

SC
established risk factors2-4. Conversely, high PA levels and cardiorespiratory

fitness (CRF) are associated with low risk of CVD and mortality3, 5, 6. It has also

NU
been demonstrated that CRF, measured as maximal oxygen uptake (VO2max), is a
MA
good indicator of CV health, and a strong predictor of CVD mortality in healthy

individuals and in patients with CVD3, 7-11. Levels of PA that increases CRF may
ED

therefore be one of the most important modifiable life-style interventions

indicated for preventing or controlling CV, metabolic or skeletal muscle diseases,


PT

and has been strongly recommended for both healthy individuals and for
CE

patients with CVD to improve CV health and reduce risk of premature

mortality12-14.
AC

Exercise training (ET) results in a large number of effects that benefit the subject

in both health and disease. Adaptations to endurance training include increased

VO2max, improved lipid profile, improved endothelial function, and increased

abundance of capillaries, mitochondria and metabolic enzymes in different

organs and systems15-18. However, a great majority of the studies in the exercise

field have focused on CV and skeletal muscle adaptations8, 11, 19-32. In the heart, a

central feature of regular ET is improved systolic and diastolic function and

larger cardiac output33-36. The long-term effects of ET include cardiomyocyte


ACCEPTED MANUSCRIPT
6

shortening and rates of contraction-relaxation improvements, providing a

cellular basis for a better organ function37. In contrast, CVD in general induce

cardiac dysfunction with reduced ejection fraction and lower cardiac output,

PT
which is also associated with impaired cardiomyocyte contractile function38-41.

Experimental studies of cellular effects of regular ET appear therefore to

RI
translate to humans, also when exercise training is applied therapeutically in

SC
CVD. Such studies suggest restored contractility and attenuated pathological

growth as important cellular mechanisms for the beneficial effects of PA in heart

failure (HF)37, 42-46.


NU
MA

However, it has been shown that aerobic ET not only improves cardiac function
ED

in patients with CVD, but also promotes a broad range of benefits in the skeletal

muscle. In fact, skeletal muscle abnormities induced by CVD has been extensively
PT

described, including rarefaction, type I to type II fibers switch, impaired


CE

metabolism, and atrophy47-51. Mainly, ET can prevent skeletal muscle atrophy,

increase in type I fiber distribution, and improve metabolic status47, 48, 51-55. At
AC

the molecular level, over the past few years, we have come to better understand

the intracellular signaling pathways mediating physiological adaptations induced

by both acute and chronic ET, and its benefits against pathological conditions.

Accordingly, recent efforts have identified intracellular mechanisms underlying

both cardiac and skeletal muscle abnormalities in CVD, and promising targets

such as metabolic enzymes and calcium handling-related proteins37, 42, 43, 52-54, 56-

61.
ACCEPTED MANUSCRIPT
7

MicroRNAs

In the early 1990s, a novel class of genes, known as microRNAs (miRNAs), have

emerged as powerful agents that control the expression of a vast gene pool in a

PT
sequence–specific manner and lead to post-transcriptional regulation62-65. The

laboratories of Dr. Victor Ambros and Dr. Gary Ruvkun discovered

RI
simultaneously the first miRNA, lin-4, in 1993 while studying post-embryonic

SC
developmental events in C. elegans66, 67. Actually, the lin-4 gene itself was initially

NU
identified in 1981 in a Cell paper by Marty Chalfie, Bob Horvitz, and John

Sulston68. At that time, these authors described the detailed cell lineage analysis,
MA
but not the molecular identity, of two genes that affect C. elegans developmental

timing, lin-4 and unc-86. Later, what both Ambros’s and Ruvkun’s groups have
ED

done was describe that lin-4 transcripts (of approximately 22 and 61 nt)

containing sequences complementary to a repeated sequence element in the 3'


PT

untranslated region (UTR) of lin-14 mRNA, suggesting that lin-4 regulates lin-14
CE

translation via an antisense RNA-RNA interaction. Since the first description of

their solid regulatory functions, miRNAs have been detected from plants to
AC

humans, and have revealed a formidable mediator of a broad range of cellular

adaptive processes such as differentiation, proliferation, apoptosis and

metabolism62, 65, 69.

MicroRNA Biogenesis

MiRNA biogenesis is a very complex process and involves multiple steps as

miRNAs can be transcribed as from non-coding regions (intergenic) or within the

protein coding genes (intragenic)62. Initiated by the RNA polymerase II62, their
ACCEPTED MANUSCRIPT
8

transcription results in the generation of primary transcripts (pri-miRNA), which

can encode individual miRNAs (monocistronic) or multiple miRNAs

(polycistronic)70. These pri-miRNAs are then processed into 60~70 nucleotides

PT
precursor-miRNAs (pre-miRNAs) by the RNase III endonuclease Drosha and its

cofactor Pasha62. As Pasha recognizes RNA stem loop structures and the

RI
neighboring single stranded sequences, this results in recruitment and cleavage

SC
by Drosha62. The pre-miRNAs are then exported to the cytosol by the Exportin-5

(XPO5) and processed into a ~22 nucleotide complex by DICER, whereby one of

NU
the duplex strands is degraded and the other becomes the mature miRNA. This
MA
mature miRNA is incorporated into the RNA-induced silencing complex (RISC)

allowing this complex to identify and bind to the 3’ or 5’UTR of the target mRNAs,
ED

resulting in its degradation or repression of protein translation62, 65.


PT

In contrast to miRNAs, little is known about ET-induced modulation on miRNA


CE

biogenesis. Recently, a study of an acute bout of endurance exercise in untrained

men showed increased gene expression of components of the miRNA biogenesis


AC

machinery. Acute endurance exercise increased mRNA of members of the

endonuclease III enzymes Drosha and DICER as well as the miRNA export

protein, Exportin 571.

Circulating MicroRNAs as New Biomarkers for Healthy and Disease

Conditions
ACCEPTED MANUSCRIPT
9

In 2008, independent groups72-74 demonstrated at the same time that miRNAs

could be detected in the circulation and are remarkably stable in biofluids,

followed by demonstrations in other body fluids like urine, cerebrospinal fluid,

PT
and saliva75. Several studies have since then shown that miRNAs can be secreted

into the bloodstream by different organs like the heart, vascular endothelial cells,

RI
skeletal muscle, liver and the brain74, 76-90. Although not yet fully understood, the

SC
mechanisms by which the miRNAs are detectable in biofluids are commencing to

be deciphered. Evidence has suggested that miRNAs are packaged in

NU
microparticles (exosomes, microvesicles, and apoptotic bodies)91-93 or associated
MA
with RNA-binding proteins (e.g., Argonaute 2)94 or lipoprotein complexes (high-

density lipoprotein)95. Some circulating miRNAs might just be waist products of


ED

ongoing physiological or pathological processes in tissue. However there is

evidence indicating that certain circulating miRNAs may have important


PT

functions as intercellular communicators77, 95-97. For instance, miRNA


CE

communication has been demonstrated between endothelial cells and

endothelial apoptotic bodies, smooth muscle cells and cardiomyocytes93, 98, 99.
AC

Interestingly, several studies have reported the association of changes in

circulating miRNAs levels with altered physiological states such as with aging, PA,

and pregnancy77, 78, 87, 89, 100-103. Due to their stability in blood, miRNAs have

emerged as promising clinical biomarkers of disease73, 74, 80, 82, 83, 89, 104-108. Our

group performed a prospective nested case-control study, with 10-year

observation period, and identified several circulating miRNAs (let-7d-5p, let-7g-

5p, miR-26a-5p, miR-29c-3p, miR-103a-3p, miR-106a-5p, miR-148b-3p, miR-

151a-5p, miR-424-5p, miR-660-5p) that predict future fatal acute myocardial

infarction in healthy participants from the Nord-Trøndelag Health (HUNT)


ACCEPTED MANUSCRIPT
10

Study109. Additionally, a miRNA model added to the Framingham Risk Score for

global CVD risk prediction revealed that 5 (let-7g-5p, miR-106a-5p, miR-144-3p,

miR-424-5p and miR-660-5p) out of the 10 miRNAs listed above significantly

PT
increased the area under the curve (AUC) obtained from the Receiver Operating

Characteristic (ROC)-curves analysis, from 0.72 (95% CI: [0.61, 0.82]) to 0.91

RI
(95% CI: [0.85, 0.96])109. Therefore, assessment of CVD risk supported by novel

SC
circulating biomarkers, such as miRNAs, is important to stratifying the

individuals at high-risk, to optimizing treatment strategies, and to enhancing our

understanding of the underlying biology.


NU
MA
ED

Role of MicroRNAs in Exercise

MiRNAs have been also found essential as mediators of processes associated


PT

with ET adaptation including cardiac110-113 and skeletal muscle114, 115

hypertrophies, angiogenesis116, 117, atherosclerosis118, neuron regeneration119,


CE

metabolism/CRF85, 120, 121. Additionally, miRNAs seem to play important roles in


AC

the acute71, 77, 88, 122 and chronic76, 86, 123 endurance exercise, resistance (RT)

exercise90, 124, in athletes (i.e., versus marathon runners78, 85), performed on a

treadmill125 or using a swimming110, 112, 113, 116, 117 protocol; in animal models (i.e.,

mouse, rats, etc)112, 117, 120, in individuals/patient cohorts76, 78, 86, 123, as well as, in

the general population121, 126. Here, we review emerging features of miRNA-

target interactions involved in the benefits of ET in the cardiac and skeletal

muscle tissues, as well as, in the spinal cord injury and in the immune system

(Fig 1). Furthermore, we will discuss the changes in the circulating microRNA
ACCEPTED MANUSCRIPT
11

levels induced by exercise-trained, and genetic variants influencing mechanisms

of post-transcriptional regulation by miRNAs.

PT
The current review efforts are therefore aimed at elucidating the role of non-

coding RNAs, such as miRNAs, with respect to beneficial effects of ET (acute and

RI
chronic) on different systems (i.e., heart, skeletal muscle, etc.), and in

SC
physiological and pathological (i.e., hypertension, HF, spinal cord injury, etc.)

conditions.

NU
MA
The Influence of Exercise on Cardiac MicroRNAs

In the last years, the use of different large-scale screening approaches provided
ED

the tools to uncover the entire cardiac transcriptome landscape, including

mRNAs, miRNAs, long noncoding RNAs and other RNA molecules, suggesting
PT

that changes in the transcriptome homeostasis are critically associated with


CE

different processes in the heart127. With regard to the role of miRNAs in the

embryonic heart development, several candidates have been implicated in the


AC

past years128-132. The role of miRNAs in myocardial development was first

assessed in the fruit fly D. melanogaster. It was demonstrated that the

overexpression of the orthologue of miR-1 in D. melanogaster (dmiR-1) in cardiac

mesoderm results in fewer cardiac cells. Conversely, loss-of-function approach

for miR-1 revealed to be uniformly lethal, showing that dmiR-1 is involved in

maintaining muscle gene expression and determination of specific cardiac cell

types from pluripotent progenitors130, 131. Additionally, the targeted deletion of

the Dicer gene, which is critical for processing of pre-miRNAs into their mature
ACCEPTED MANUSCRIPT
12

form in the heart, provokes spontaneous cardiac remodeling, dilated

cardiomyopathy and HF, reflecting the apparent role of miRNAs to modulate

multiple targets and interfere in the embryonic heart development128, 129. In the

PT
adult myocardium, several studies have demonstrated the involvement of

miRNAs in a number of physiological110-113, 116, 117 and pathological125, 127, 133-140

RI
conditions. Changes in cardiac miRNA expression levels have been associated

SC
with cardiac stress and development of cardiac hypertrophy due to pressure

overload133-135, 141, myocardial infarction139, and also, in humans, end-stage HF127,

142, 143.
NU
MA

Aerobic ET leads to a physiological, nonpathological left ventricular (LV)


ED

hypertrophy (LVH). However, in order to study the beneficial effects of ET, a

number of experimental strategies have been established, such as treadmill37, 44,


PT

61, 144, 145 and voluntary wheel146, 147 running, swimming117, 148, 149 and RT150-152.
CE

These approaches have been applied to numerous animal models with various

backgrounds. However, important differences exist between these experimental


AC

approaches, which may affect the interpretation of the results.

Most of the studies performed in rats and mice have applied continuous

treadmill running, characterized by fixed or progressively increasing speed,

inclination, and duration during the session (Table 1 and 2). Different treadmill

running protocols have been developed, lasting from weeks to months, with

individual running session durations ranging from minutes to hours and running

speeds ranging 10-97 m/min, and with the treadmill inclinations ranging 0-25°

(0-47 %)45, 46, 153, 154. Care and colleagues125 assessed the role of cardiac miRNAs
ACCEPTED MANUSCRIPT
13

in the three murine models of cardiac hypertrophy, i.e. transverse aortic arch–

constricted mice, transgenic mice with selective cardiac overexpression of a

constitutively active mutant of the Akt kinase, and ET performed in rats.

PT
Interestingly, cardiac hypertrophy in all three models (pathological or

physiological cardiac hypertrophy) resulted in reduced expression of both hsa-

RI
miR-133 and hsa-miR-1.

SC
Conversely, swimming exercise protocols induce different responses in terms of

NU
acute and chronic hemodynamic changes compared to running protocols149.
MA
Additionally, swimming is recognized for its efficiency in inducing myocardial

hypertrophy and a significant increase in LV end-diastolic volume in rats148.


ED

Therefore, Ma and colleagues111 demonstrated that as swimming exercise-

induced physiological cardiac hypertrophy is correlated with the modulation of


PT

few miRNAs (rno-miR-21, rno-miR-124, rno-miR-144, and rno-miR-145) that


CE

has components of the PI3K/AKT/mTOR signaling pathway (Pik3a, Pten, and

Tsc2) as validated targets. Fernandes and colleagues155 studied, in females


AC

Wistar rats, the effects of two different swimming ET regiments, moderate (60

min/day, 5x/wks for 10 wks with 5% body overload) and high (60 min/day,

5x/wks for 10 wks with 5% body overload, 2-3 times a day) volumes, on

microRNA profile and their targets. Both swimming ET regiments altered the

expression of cardiac miRNAs targeting renin-angiotensin aldosterone system

(RAAS) components and were associated with the development of LVH.

Swimming exercise increased rno-miR-27a and rno-miR-27b, targeting

angiotensin converting enzyme (Ace) gene, and decreased rno-miR-143,

targeting Ace type 2 (Ace2), in the heart. da Silva Jr and colleagues116 showed
ACCEPTED MANUSCRIPT
14

that swimming ET also induced an increase in rno-miR-126 which target Spred-1

(associated with angiogenesis), which decreased after 10 weeks of ET. Although

Pi3kr2 is also a well-validated target for rno-miR-126, swimming exercised rats

PT
did not change its expression.

RI
The involvement of different members of the miR-29 family in the cardiac

SC
hypertrophy induced by swimming exercise training has been also assessed.

Rno-miR-29c expression increased in trained rats and was inversely correlated

NU
with ColIaI and ColIIIaI expression and OH-proline concentration, which is
MA
relevant to the improved left ventricle compliance observed113. Members of the

miR-29 family (miR-29a, miR-29b-1, miR-29b-2, miR-29c) seem to orchestrate a


ED

complex network of signaling pathways regulating deposition of extracellular

matrix proteins in different tissues and pathological conditions156. In fact, miR-


PT

29 family has been implicated in development of cardiac139, liver157, renal158 and


CE

pulmonary159 fibrosis. Moreover, it has been described that the cardiac

remodeling associated with hypertrophic cardiomyopathy in patients


AC

determines a release of hsa-miR-29a into the bloodstream to be significantly

associated with both hypertrophy and fibrosis. This data suggest that this miRNA

family has the potential to be used as a circulating biomarker for myocardial

remodeling assessment in patients with hypertrophic cardiomyopathy160.

Interestingly, ET seems to modify the expression levels of the different members

of the miR-29 family under pathological conditions. Swimming ET restores

cardiac rno-miR-29ac levels and prevents collagen deposition in the border zone,

as well as, in the remote myocardium, of the infarcted hearts, which may

contribute to the improvement in ventricular function induced by swimming


ACCEPTED MANUSCRIPT
15

ET112. The studies described above collectively suggest that swimming ET in

experimental animal models seems to change the cardiac levels of several

miRNAs (miR-27a, -27b, -143, -29ac, and -126) influencing a broad spectrum of

PT
targets, from components of the RAAS to the Raf-1/Erk1/2 or collagen pathways;

and, associated with different phenotypes, tissue remodeling, angiogenesis, etc.

RI
Nevertheless, the long-term effects of ET on cardiac miRNA expression are

SC
particularly dependent on the modalities, e.g., motorized treadmills, voluntary

NU
running wheel, and swimming or RT. Figure 2A compiled all the results

presented in the current review (Table 1 and 2) and shows a lack of overlap
MA
regarding cardiac miRNAs profile in different animal models (mice and rats).

This suggest that the molecular and cellular adaptations in the heart, at least at
ED

the miRNA level, are distinct according to the type of ET. Importantly, the
PT

current review sheds some lights in the especificities of exercise training

modalities and miRNA expression, and hopes to help scientists to choose the
CE

most appropriate ET modality and/or animal model for their investigation.


AC

Nevertheless, any inference regarding the influence of ET on cardiac miRNAs is

limited to data obtained from preclinical studies (mostly in small rodents). No

clinical study has been published so far, uncovering the effects of ET on the

cardiac miRNA profile. Changes in cardiac miRNA expression levels have been,

however, described in CVD patients. A few years ago, a randomized clinical trial

study was conducted by our group and demonstrated that remote ischemic

preconditioning (RIPC) preserves mitochondrial function and influences miRNA

expression in atrial myocardium during coronary bypass surgery161. MiR-133a


ACCEPTED MANUSCRIPT
16

and miR-133b were found upregulated after aortic cross-clamping in both RIPC

and control groups, whereas miR-1 was upregulated in control only, and miR-

338-3p expression level was higher in RIPC versus control after aortic cross-

PT
clamping.

RI
In line with these ideas, several biotech and pharmaceutical companies have

SC
invested in clinical trials on miRNA-based therapy in management of cancer,

metabolic diseases, chronic infections and mycosis (for reference cite162). In one

NU
of the first reports, miR‑122‑targeted locked nucleic acids (LNAs) inhibitor
MA
(Mirvirasen, Santaris Pharma A/S and Hoffmann‑La Roche) was used to treat

Hepatitis C-infected patients, in a multicentre phase IIa clinical trial, and


ED

demonstrated a significant reduction in viral titres163. More recently, a

multicentre phase I clinical trial (ongoing) has been conducted to treat


PT

Cutaneous T cell lymphoma and mycosis fungoides patients by using


CE

antimiR‑155 LNA-modified antisense inhibitor (MRG-106, miRagen

Therapeutics). In addition, antimiR‑103/-107 (RG-125/ AZD4076, Regulus


AC

Therapeutics) is in use to treat patients with type 2 diabetes and non-alcoholic

fatty liver diseases, in a single-centre phase I/IIa clinical trial (ongoing).

Skeletal Muscle MicroRNAs

Although miRNAs have been implicated in skeletal muscle development,

regeneration, and function, our understanding of the molecular mechanisms


ACCEPTED MANUSCRIPT
17

underlying the regulation of skeletal muscle mass by miRNAs is still limited.

Exercise, nutrition, and disease affect skeletal muscle mass postnatally, and they

were also recently shown to alter skeletal muscle miRNA expression164, 165.

PT
Several of miRNAs have been implicated in these processes (Tables 1, 2 and 3).

For instance, miR-206166, and miR-1115 and miR-133115 are grouped as

RI
“myomiRNAs” or myocytes miRNAs, as they are specific to or enrich the skeletal

SC
muscle tissue and play important roles in the regulation of muscle development

and differentiation167.

NU
MA
Either loss- and gain-of-function experiments have shown that miR-208b and

miR-499 play important and redundant roles in the specification of muscle fiber
ED

identity by activating slow and repressing fast myofiber gene programs168. The

mmu-miR-208b-/-; mmu-miR-499-/- double knockout (dKO) mouse presented a


PT

marked loss of slow myofibers in the soleus, and also reduced expression of slow
CE

-MHC at the protein and mRNA levels. Conversely, muscle-specific

overexpression of mmu-miR-499 was sufficient to induce a complete conversion


AC

of all fast myofibers in soleus to a slow, type I phenotype168.

Similarly to what has been described in the heart, miRNAs can mediate the

exercise training-induced changes in the skeletal muscle phenotype. In fact,

exercise can rapidly and transiently regulate several miRNAs in the skeletal

muscle. Russell and colleagues71 have performed measurements in muscle

biopsies from healthy untrained males at rest, after a single bout of moderate-

intensity endurance cycling and after chronic endurance cycling training. In the

3h period following the acute exercise bout hsa-miR-1, hsa-miR-133a, hsa-miR-


ACCEPTED MANUSCRIPT
18

133-b and hsa-miR-181a were all increased. Hsa-miR-9, hsa-miR-23a, hsa-miR-

23b and hsa-miR-31 were decreased. In contrast, short-term ET regime

increased hsa-miR-1 and hsa-miR-29b, while decreasing hsa-miR-31 in the

PT
skeletal muscle biopsies. Additionally, luciferase reporter activity assay validated

both HDAC4 and NRF1 as direct targets for hsa-miR-31. In another study, Nielsen

RI
and colleagues86 demonstrated that hsa-miR-1 and hsa-miR-133a expression

SC
levels in the vastus lateralis of healthy individuals were significantly upregulated

in response to a single bout of cycle ergometer exercise at 65% of Pmax86. On the

NU
other hand, 12 weeks of endurance ET on a cycle ergometer resulted in a
MA
decrease in the hsa-miR-1, hsa-miR-133a, hsa-miR-133b, and hsa-miR-206.
ED

Of interest, miR-1 and miR-133, clustered on the same chromosomal loci, are

transcribed together in a tissue-specific manner during development and have


PT

distinct roles in the modulating skeletal muscle proliferation and


CE

differentiation169. Therefore, miR-1 and miR-133 are both involved in cardiac-

related125, 140, 161 and skeletal-related169-171 human diseases.


AC

Moreover, mmu-miR-494 levels significantly decrease after endurance ET in

C57BL/6J mice, accompanied by an increase in expression of mtTfa and Foxj3

proteins. These results suggest that miR-494 regulates mitochondrial biogenesis

by downregulating mtTfa and Foxj3 during myocyte differentiation and skeletal

muscle adaptation to ET172.

The benefits of swimming ET in the skeletal muscle vasculature have also been

studied in an experimental model of arterial hypertension (i.e., spontaneously


ACCEPTED MANUSCRIPT
19

hypertensive rats, SHR). Fernandes and colleagues117 investigated the miRNAs

profile after ET, and the changes in the vascularization in the skeletal muscle. It

was found that ET reduced rno-miR-16 and rno-miR-21 expression and elevated

PT
vascular endothelial growth factor (Vegf) and Bcl-2 levels, key proteins in

angiogenesis and apoptosis, respectively. Moreover, rno-miR-126 level, which

RI
was reduced in the hypertensive sedentary rats, was normalized by ET in the

SC
hypertensive rats accompanied by changes in the phosphoinositol-3 kinase

regulatory subunit 2 (Pi3kr2) levels117.

NU
MA
Nevertheless, human subjects show considerable variation in the extent of ET

adaptation. Several factors, e.g.. age, sex, race, and initial fitness level, are known
ED

to play a role. In addition, genetics may certainly explain a large variability in the

ET response12, 173, 174. In order to verify the role of miRNAs in the ET


PT

responsiveness Davidsen and colleagues114 collected biopsies from the vastus


CE

lateralis muscle of healthy men, stratified according to their capacity to gain

muscle mass with RT (i.e., low and high responders), and showed a differential
AC

expression profile in hsa-miR-378, hsa-miR-29a, hsa-miR-26a, and hsa-miR-

451114. Interestingly, miR-133a/b and miR-1 expression levels, also known as

myomiRNAs, were found unaffected after a 12-wk RT program. The authors also

examined the changes in the mRNA expression levels of three genes (IGF-I, VEGF-

A, eIF4E2) that were predicted as targets of the miRNAs found in the study.

Taken together, the data suggested that miRNAs play an important role in

regulating the translation of key pathways responsible for skeletal muscle

growth in response to RT in high and low responder individuals.


ACCEPTED MANUSCRIPT
20

Taken together, the studies reported in the present review suggest that

myomiRNAs play an important role in the skeletal muscle adaptations to ET in

both human health and disease. Precisely, ET can modify the levels of several

PT
miRNAs, e.g.. miR-1, miR-16, miR-21, miR-26a, miR-29a, miR-126, miR-133a,

miR-133b, and miR-206, miR-378, miR-451, and miR-494. Coordinated changes

RI
in the expression levels of these myomiRNA contribute to skeletal muscle

SC
adaptations to acute and chronic ET.

NU
MA
Circulating MicroRNAs
ED

Recently, miRNAs have arisen as promising circulating biomarkers of disease, as

large amounts of stable miRNAs can enter the circulation106. Increased


PT

circulating levels of hsa-miR-1 have been associated with myocardial


CE

infarction175, while increased circulating levels of hsa-miR-423 have been

associated with HF176. Furthermore, increased circulating levels of the cardiac


AC

muscle-enriched hsa-miR-208 have been reported in patients with stable

coronary artery disease (CAD) and with myocardial damage caused by CVD80-82,

107. Elevation in another myocardial-enriched miRNA, hsa-miR-133a, has been

reported in the plasma of patients with stable CAD81. Additionally, increased

serum levels of hsa-miR-210 have been reported in patients with atherosclerosis

and kidney disease84, 177. On the other hand, several miRNAs have also been

reported to be downregulated in patients with stable CAD81, like the endothelial-


ACCEPTED MANUSCRIPT
21

associated hsa-miR-126, hsa-miR-17, and hsa-miR-92a; the inflammation-

associated hsa-miR-155; and the smooth muscle-enriched hsa-miR-145.

PT
With regard to physiological conditions, during the last few years there has been

a rise in the number of studies on circulating miRNAs in the bloodstream

RI
induced by both acute and chronic exercise76-79, 85, 90, 121, 123, 178. Release of

SC
miRNAs into the circulation after acute exhaustive exercise is likely to be a result

of exercise-induced tissue stress and/or damage. For instance, Baggish and

NU
colleagues77 reported that vascular endothelial cells are capable of releasing high
MA
levels of both hsa-miR-21 and hsa-miR-222 into the circulation immediately

after acute exhaustive cycling exercise.


ED

The same authors77 highlighted the rapid mobilization of circulating miRNAs in


PT

response to a cardiopulmonary exercise test, as they reported a rapid


CE

upregulation of miR-146a, miR-222, miR-21, and miR-221 in the setting of acute

exercise (cardiopulmonary exercise test), followed by a restoration of the plasma


AC

levels in some of the circulating miRNAs (miR-146a, miR-222 and miR-21) after

1 h of rest following acute exercise. Moreover, Nielsen and colleagues123

performed a screening for miRNAs in the circulation from young healthy men

before and after an acute endurance exercise bout (60 min cycle ergometer

exercise bout at 65% of Pmax) and following endurance ET (60-120 min/section

on cycle ergometer, 5x/wk, 12 wks, 55-91% of Pmax). These authors

demonstrated that ET orchestrates a dynamic regulation of miRNAs in the

circulation. For instance, immediately after the acute exercise bout, hsa-miR-

106a, hsa-miR-221, hsa-miR-30b, hsa-miR-151-5p, hsa-let-7i, hsa-miR-146, hsa-


ACCEPTED MANUSCRIPT
22

miR-652 and hsa-miR-151-3p were significantly downregulated in the

circulation, suggesting a bloodstream clearance as a result of uptake from

surrounding tissues. One hour after the acute bout, 5 miRNAs (miR-338-3p, miR-

PT
330-3p, miR-223, miR-139-5p, miR-143) were found upregulated and after three

hours only hsa-miR-1 was found increased.

RI
SC
A few studies have also reported changes in circulating miRNAs after long-term

exercise training protocols (Figs 1 and 2; Table 3). Baggish and colleagues77

NU
studied subjects that performed moderate-intensity endurance ET (rowing) for 3
MA
months and reported increased basal levels of hsa-miR-21, hsa-miR-146a, hsa-

miR-221 and hsa-miR-222. Another study by Taurino and colleagues178 reported


ED

that long-term endurance ET (10 weeks) caused an increase in both hsa-miR-92a

and hsa-miR-92b circulating levels in patients with CAD.


PT
CE

Uhlemann and colleagues103 showed that the mode of ET is important for the

miRNA-signature in the bloodstream. Whereas different protocols of endurance


AC

ET led to an acute increase in the endothelial-specific hsa-miR-126, RT caused an

increase in the skeletal muscle-specific hsa-miR-133. In another study, miRNAs

levels were measured in serum samples from twelve healthy males performing a

single resistance exercise session (bench press and leg press), consisting of five

sets of 10 repetitions at 70% of maximum strength, with a 1 min rest between

each set. Three days after the resistance exercise session the authors found

increased levels of hsa-miR-149 and decreased levels of hsa-miR-146a and hsa-

miR-22190. Comparing the acute against the chronic effects of exercise on the

miRNAs levels in the circulation, different results have been reported (Fig 2B).
ACCEPTED MANUSCRIPT
23

Nielsen and colleagues123 showed that chronic ET induced changes in a

completely different set of miRNAs in the circulation compared to an acute bout.

Differently, Aoi and colleagues76 found that hsa-miR-486 levels were

PT
significantly decreased following both acute and chronic exercise.

RI
Circulating miRNAs have also been the subject of a study in athletes78, 85.

Fourteen male marathon runners were investigated for myomiRNAs and

SC
inflammation related miRNAs in the plasma before, directly after, and 24h after a

NU
marathon run85. It was found that hsa-miR-1, hsa-miR-133a, and hsa-miR-206

correlated to aerobic performance parameters, such as VO2max and running


MA
speed at individual anaerobic lactate threshold. Interestingly, hsa-miR-1 showed

a moderate negative correlation with fractional shortening, whereas hsa-miR-


ED

133a was positively correlated with the thickness of intraventricular septum.


PT

None of the miRNAs correlated with cardiac injury markers, such as troponin T,

troponin I, and pro-brain natriuretic peptide85. Baggish and colleagues78


CE

evaluated the levels of muscle-enriched miRNAs (hsa-miR-1, hsa-miR-133a, hsa-

miR-499–5p), cardiac tissue (hsa-miR-208a), and the vascular endothelium (hsa-


AC

miR-126), as well as those important in inflammation (hsa-miR-146a) in male

marathon runners at rest, immediately after a marathon (42-km foot race), and

24 h after the race. All the measured miRNAs were found elevated after the

marathon, but hsa-miR-1, hsa-miR-133a, hsa-miR-208a and hsa-miR-146a were

the most robustly upregulated. More recently, it has been reported in data from

the Munich Marathon Study showing that both elite and non-elite runners

exhibited increased plasma levels of miR-30a mmediately after a marathon.


ACCEPTED MANUSCRIPT
24

Interestingly, miR-1 and miR-133a plasma levels also increased but showed

significant changes only in elite runners179.

PT
Changes in the miRNAs levels in the circulation not only reflect the effects of

both acute and chronic exercise, but also associate with inherited CRF. A recent

RI
study by Bye and colleagues121 reported that circulating levels of hsa-miR-210

SC
and hsa-miR-222 were significantly higher in individuals with low VO2max

compared to individuals with high VO2max. Furthermore, hsa-miR-21 was

NU
elevated in male participants with low VO2max. However, in this particular study
MA
no correlations were found between hsa-miR-210, hsa-miR-21 and hsa-miR-222

and frequency, intensity, and duration of self-reported regular ET. Another study
ED

conducted by Baggish and colleagues77 reported a positive correlation between

the circulating levels of hsa-miR-146a and VO2max in 10 male athletic subjects.


PT

These findings strengthen the hypothesis that CRF biomarkers also have a
CE

potential as circulating markers of future CV health independent of traditional

biomarkers.
AC

Taken together the studies reported that even brief bouts of exercise

substantially perturb the cellular homeostasis and dramatically change the

circulating miRNA signature. These changes are dynamic over short periods of

time in acute exercise, as well as over longer periods of time of endurance ET.

Furthermore, detection of miRNAs in the bloodstream after acute and chronic

exercise seems to be due to selective secretion rather than generalized passive

release due to muscle damage, for example. Therefore, detection of miRNAs in

the bloodstream may become a reliable marker for CRF or a diagnosis and
ACCEPTED MANUSCRIPT
25

prognosis marker for chronic diseases (i.e. CVD). In fact, the diagnostic and

prognostic value of circulating miRNAs is currently exploited by several groups

around the world74, 80-82, 89, 104, 160, 177. However, the mechanisms underlying the

PT
exercise-induced miRNA secretion in the bloodstream remain unclear. The vast

majority of the circulating miRNA data available in the literature is restricted to

RI
associations uncovered by observational studies, limiting any mechanistic

SC
inference. The synergy of mechanistic (preclinical) and multicentric, prospective

large-scale studies is therefore required to determine the potential use of

NU
circulating miRNAs as biomarkers for the development of complex traits and
MA
diseases.
ED

MicroRNAs and Single Nucleotide Polymorphisms (SNPs)


PT
CE

More recently, a myriad of studies180, 181 have been focused on identification and

description of single nucleotide polymorphisms (SNPs) located in non-coding


AC

regions and their role in the gene regulation in association with different

pathological conditions. Particularly, genetic variants influencing miRNA post-

transcriptional regulation have been investigated182-184. Single nucleotide

polymorphisms can influence miRNAs function in several ways, depending on

where positioned: if located in the pre-miRNA (outside the seed sequence); or, in

a 3’UTR of a target gene; or, in miRNAs promoter region; or, if a mutation

generates an alternative polyA site; or, even, if a gene responsible for the miRNA

biogenesis is mutated (i.e., Drosha, Dicer, and Argonaute).


ACCEPTED MANUSCRIPT
26

Interestingly, miRNA sequence variation can broadly affect miRNA function, even

when the nucleotide mismatch is located far away from the critical 5’ seed

PT
sequence generally considered to drive mRNA target recognition and

suppression. In an elegant study, Dorn and colleagues184 showed that a mutation

RI
outside the seed sequence of hsa-miR-499 alters cardiac mRNA targeting and

SC
end-organ function. Son and colleagues181 reported that a promoter

polymorphism (rs1042522) of pri-miR-34b/c is associated with an increase in

NU
the risk of hepatocellular carcinoma in the Korean population. In addition,
MA
genetic variations in the miRNA-processing machinery component (DROSHA)

can affect miRNA expression levels and lung cancer-specific survival185.


ED

Furthermore, AGO1 rs636832 A>G might also determine the susceptibility to

lung cancer, suggesting that the AGO1 and/or miRNAs might be involved in the
PT

development of lung cancer186.


CE

SNPs can interfere directly in the post-transcriptional regulation of miRNAs


AC

when is positioned in the mRNA target sites. Amin and colleagues182

demonstrated that rs2519184, rs8234, and rs10798 variants in the 3’UTR of the

KCNQ1 (Kv7.1 potassium channel) modify disease severity in patients with long

QT Syndrome type 1 in an allele-specific manner. Therefore, these authors

suggest that assessing these 3’UTR SNPs can predict disease severity of a given

KCNQ1 mutation in one family. Moreover, it has been shown that a functional

variant (rs2507800) in the 3’UTR of angiopoietin-1 gene might reduce risk of

stroke by interfering with the binding efficiency of hsa-miR-211180. Similarly, a

rs3735590 variant of PON1 3’UTR interferes with hsa-miR-616 binding to


ACCEPTED MANUSCRIPT
27

increase the risk of ischemic stroke and carotid atherosclerosis187. Skeletal

muscle phenotypes seem to be equally influenced by genetic variants in miRNA

target site. Clop and colleagues183 verified that a mutation in the myostatin

PT
(GDF8) 3’UTR creates a target site for hsa-miR-1 and hsa-miR-206, affecting

muscularity in sheep.

RI
SC
Polyadenylation signals can, for example, be created or disrupted by SNPs

altering gene regulation. Thomas and colleagues188, matching RNA-seq data to

NU
miRNA expression profile, showed an association between alternative polyA site
MA
strength (signal and GU-content) and loss of miRNA target sites, allelic imbalance,

and transcript expression. Accordingly, alternative polyA motifs created by SNPs


ED

can affect miRNA-based gene regulation and thereby are potential disease-

causative variants.
PT
CE

More recently, we were able to describe a new genetic variant (rs540) within a

miRNA target site in the TMEM8A (transmembrane protein 8A) 3’UTR associated
AC

with VO2max in adult healthy males126. Interestingly, a point mutation in the

TMEM8A 3’UTR creates a new target site for hsa-let-7a-3p and affects TMEM8A

gene expression through miRNA regulation (Table 3).

Other Conditions

MiRNAs changes induced by exercise training have been investigated in a few

other conditions or pathological diseases, e.g. spinal cord injury, senescence and
ACCEPTED MANUSCRIPT
28

the immune system. Here we detailed the exercise-induced miRNAs regulation in

those particular conditions (Fig 1; Tables 1 and 2).

PT
Spinal cord injury changes in the skeletal muscles are characterized by decrease

in gross muscle size, myofiber atrophy, and altered expression of myosin heavy

RI
chain proteins, and lead to loss of strength and endurance of the muscle189, 190.

SC
With regard to therapeutic treatment of spinal cord injury, it has generally been

accepted that exercise is an effective therapy that maintains muscle mass191,

NU
stabilizes rhythmic firing patterns of lumbar motoneurons192, 193 and improves
MA
functional motor and sensory recovery194, 195 after spinal cord injury. Over the

past years, we have witnessed some profiling attempts to uncover the miRNAs
ED

involved in this process. Liu and colleagues196 screened for miRNAs associated

with the damage to the spinal cord and treatment using exercise in an
PT

experimental model. Briefly, the spinal cord injury was experimentally induced
CE

in Sprague-Dawley rats by exposing the dorsal surface of spinal cord at the level

of the tenth thoracic vertebra and aspiration of the spinal cord through a
AC

micropipette in order to create a 2 mm long complete transection lesion; ET

consisted of a passive form of hind limb cycling exercise (45 rpm, 30 min, 2x per

day, starting 5 days after the spinal cord transection, 5 days per week, for 20

days). Spinal cord injury induced a significant decrease in the rno-miR-15b level.

Five days of exercise (passive form of hind limb exercise) after the injury

resulted in a significant increase in the expression of rno-miR-21 and an even

larger reduction of rno-miR-15b. In addition, changes in the microRNA levels

were accompanied by changes in the expression of target genes of these miRNAs

and a significant increase in the expression of mRNA for the anti-apoptotic


ACCEPTED MANUSCRIPT
29

marker Bcl-2196. In a subsequent article, the same group showed that the activity-

dependent plasticity in the injured spinal cord is modulated, at least in part,

through miRNAs (rno-miR-21 and rno-miR-199a) in spinal cord tissue that

PT
regulates Pten and mTor signaling and may indicate that exercise (cycle

ergometer, 5 days/week for 31 days) leads to an increase in the regenerative

RI
potential of neurons affected by a spinal cord injury119. Therefore, these studies

SC
provide evidences that cycling ET is a potent modulator of miRNAs expression

that affect both growth-associated Pten/mTor and apoptosis-associated

NU
pathways after spinal cord injury in rats. These pathways are a critical regulator
MA
of protein synthesis, axon regeneration and plasticity in the damaged central

nervous system.
ED

Exercise is beneficial for healthy aging and can improve cognitive function in
PT

older people, as well as people with existing neurological diseases, such as


CE

multiple sclerosis, Parkinson's and Alzheimer's disease197-199. However, the

cellular and molecular mechanisms underlying the beneficial effects of ET in


AC

people with neurological conditions are poorly understood. Recently, Cosín-

Tomás and colleagues200 studied the miRNAs expression profile in SAMP8 mice

and the modulation by exercise in the central nervous system, more precisely in

the hippocampus. The senescence-accelerated mouse prone (SAMP8) is an

animal model of age-related cognitive decline. The learning and memory

disturbances develop spontaneously and are due to overproduction of amyloid

precursor protein in the central nervous system, a similar mechanism

responsible for the pathophysiology of the Alzheimer’s disease201. The authors

found 84 differentially expressed miRNAs in the hippocampus that are suitable


ACCEPTED MANUSCRIPT
30

as senescence markers in this neurological condition. Furthermore, 8 weeks of

voluntary running wheel potentiated (mmu-miR-28a, mmu-miR-98, mmu-miR-

148b, mmu-miR-7a and mmu-miR-15b) the expression levels of few miRNAs, as

PT
well as restored (mmu-miR-105 and mmu-miR-133b) the transcription of other

miRNAs. Additionally, exercise training led to an increase in several neurogenic

RI
factors (i.e., Igf1, Bdnf, etc.) in the circulation and in the hippocampus in the

SC
SAMP8 strain. Data suggests that miRNAs are involved in the downregulation of

the target genes that control accelerated senescence. Therefore, miRNAs are

NU
increasingly recognized to play valuable roles in different pathological diseases
MA
and the development of new therapeutic strategies for senescence and

neurodegeneration.
ED

Exercise seems to also boost the immune system. In fact, it has been well
PT

demonstrated that ET modulates several parameters of both natural and


CE

acquired immunity202, 203, reducing the number of infections, inhibiting

tumourigenesis, and enhancing resistance to tumor implantation56, 202-205.


AC

Exercise seems to affect the immune system according to type, duration and

intensity of exercise, training or nutritional status, and environmental conditions

(temperature or altitude). In terms of mechanisms, a single brief bout of exercise

causes substantial changes in the expression of gene pathways in different

immune cells and inflammatory mediators. Recent publications have highlighted

the involvement of miRNAs in regulating the immune response induced by

exercise87, 88, 102, 118. Radom-Aizik and colleagues88 investigated the effects of an

acute bout of exercise (ten 2-min bouts of cycle ergometer exercise, at 76% of

VO2max) in the mRNA/miRNA transcriptional levels in circulating neutrophils.


ACCEPTED MANUSCRIPT
31

Neutrophils are the most abundant type of white blood cells in mammals,

comprising about 40-75% of all white blood cells, and form an essential part of

the innate immune system. Exercise seems to lead not just to increased numbers

PT
of neutrophils in the circulation, but also to substantial changes in expression

gene206. Pathway analysis revealed an enrichment of the gene associated with

RI
important inflammatory mechanisms (Ubiquitin-mediated proteolysis pathway,

SC
the JAK/STAT signaling pathway and the Hedgehog signaling pathway)88.

NU
In a subsequent study, Radom-Aizik and colleagues87 evaluated the effects of
MA
acute exercise (brief bouts of heavy exercise) on miRNAs expression in

circulating peripheral blood mononuclear cells, leukocytes, natural killer (NK)


ED

cells, and monocytes. The authors reported altered expression level of 34

miRNAs, mostly related to inflammatory processes. Among others, they reported


PT

a downregulation of hsa-miR-125b, hsa-let-7e and upregulation of hsa-miR-132.


CE

Furthermore, another paper by Radom-Aizik and colleagues102 studied the

effects of acute exercise on miRNA expression in natural killers cells. Briefly, NK


AC

cells are unique innate immune cells that increase up to fivefold in the circulating

blood with brief exercise and are known to play a key role in first-response

defense against pathogens and in cancer immunosurveillance. In total, 13

individuals have been submitted to an acute bout of exercise (cycle ergometer,

10x 2-min bouts, 1-min rest interval between each bout, 77% of VO2peak) and 23

differently expressed miRNAs and 986 genes altered by exercise have been

identified (Table 3)102. The authors identified 7 KEGG pathways (p53 signaling

pathway, melanoma, glioma, pathways in cancer, adherens junction, and focal

adhesion) found enriched with genes.


ACCEPTED MANUSCRIPT
32

More recently, the same group evaluated the transcriptomic

(mRNAs/microRNAs) profile in circulating monocytes immediately after a single

PT
bout of exercise118. In total, 20 healthy men were submitted to an acute exercise

session (ten 2-min bouts of cycle ergometer exercise at 82% of VO2max

RI
interspersed with 1-min rest) and 894 genes and 19 miRNAs were found

SC
differently expressed. The authors believe that the changes found in the

mRNAs/miRNAs are likely to direct monocytes in an anti-inflammatory, anti-

NU
atherogenic pathway. These data support the hypothesis that exercise affects the
MA
gene and miRNA expression pattern in different population of circulating cell

lines (monocytes, NK, neutrophils, etc.) and suggest mechanisms through which
ED

both acute and chronic exercise training could alter the immune system.
PT
CE

Concluding Remarks and Future Perspectives

Altogether, we have underlined the role of miRNAs as essential mediators of


AC

processes associated with exercise adaptation including cardiac and skeletal

muscle hypertrophies, angiogenesis, atherosclerosis, neuron regeneration,

metabolism, and CRF207, 208. Although a considerable number of preclinical

investigations on miRNA therapeutics have been conducted over the past years,

only a small number has so far moved into clinical application. Therefore,

specifically within the Exercise Physiology field, we can list few important points

that have to be properly addressed: First, a better understanding of miRNA

dynamic changes over shorts periods of time in acute exercise, as well as, over
ACCEPTED MANUSCRIPT
33

longer periods of time of endurance ET is not fully known; Secondly, we here

stressed how critical is the selection of ET protocols/modalities, as well as, a

choice of an animal model, regarding miRNA adjustments; Thus, a more detailed

PT
description of miRNA-mediated gene regulation induced by ET, especially into

preclinical and large clinical setups, is indispensable. Additionally, it is equally

RI
essential to address other more fundamental questions regarding miRNAs

SC
function, e.g. miRNA biogenesis (for referece see12) and the role of genetics (for

reference see179). Equally important, it is highly desirable to elucidate the use of

NU
miRNAs as circulating biomarkers, such as, miRNA release mechanisms (e.g.,
MA
extracellular vesicles, exosomes, and ceramide-dependent pathway)(for

reference see207, 208) and miRNA active role (e.g., miRNA communication through
ED

exosomes). Finally, in the near future it will be important to expand the miRNA

field of investigation towards other regulatory noncoding RNA (ncRNA)


PT

molecules (for reference see209). A better undertanding of these mechanisms can


CE

guide us to prevent and treat CVD, by uncovering new potencial therapeutic

targets. For instance, an in vivo therapeutic approach using a combination of key


AC

miRNAs, named “miR combo”210-212, has shown promising results.

Acknowledgments

We gratefully acknowledge Nina Zisko for critically proof-reading of the

manuscript. This research was supported by grants from the K.G. Jebsen

Foundation, The Research Council of Norway, and The Norwegian Council of

Cardiovascular Disease.
ACCEPTED MANUSCRIPT
34

Figure Legends

Fig 1 - miRNAs up and downregulated by exercise training in different tissues

and in the circulation. * other conditions included spinal cord lesion and brain

PT
phenotypes.

RI
Fig 2 - Effects of acute and chronic exercise training on miRNAs. (A) Exercise

SC
training modality (resistance training vs. aerobic exercise training on motorized

treadmill vs. voluntary running wheel vs. swimming exercise training)

NU
influencing miRNAs expression in the heart; (B) Acute and chronic exercise
MA
inducing changes in circulating miRNAs.
ED
PT
CE
AC
ACCEPTED MANUSCRIPT
35

PT
miR-1 miR-126
miR-17 miR-133b
miR-19b miR-144
miR-21 miR-145
miR-27a miR-150
miR-1 miR-181
miR-27b miR-208a

RI
miR-29a miR-216a miR-23a miR-200c miR-20a miR-146a
miR-29c miR-222 miR-27a miR-376a miR-21 miR-221 miR-7a miR-98
miR-30b miR-484 miR-107 miR-377 miR-103 miR-222 miR-15b miR-105
miR-30e miR-488 miR-126 miR-499b miR-107 miR-376a miR-21 miR-133b
miR-96 miR-136 miR-558 miR-126 miR-28a miR-148b

SC
Heart Skeletal Muscle Circulation Other Conditions*
(serum/plasma)
miR-1 miR-181a miR-1 miR-486 let-7d miR-185 miR-15b miR-199a
miR-22 miR-191a miR-16 miR-494 miR-16 miR-192
miR-99b miR-208a miR-21 miR-509 miR-21 miR-193b

NU
miR-100 miR-208b miR-23 miR-520a miR-25 miR-342
miR-124 miR-214 miR-28 miR-548an miR-27a miR-486
miR-143 miR-425 miR-30d miR-628 miR-28 miR-766
miR-133a miR-653 miR-148a
miR-133b miR-670
miR-204 miR-696
MA
miR-206 miR-761
miR-330 miR-889
miR-345 miR-1245a
miR-375 miR-1270
miR-378 miR-1280
miR-449c miR-1322
miR-483 miR-3180
ED
PT

Fig 1
CE
AC
ACCEPTED MANUSCRIPT
36

A Cardiac miRs Treadmill


Resistance
Swimming

↓mir-425 ↑ miR-96
↓ miR-214
↑ miR-216a
↓ miR-1 ↑ miR-1 ↑ miR-484

PT
↓ miR-22 ↑ miR-17 ↑ miR-488
↓ miR-99b ↑ miR-19b
↓ miR-100 ↑ miR-21
↓ miR-124 ↑ miR-27a
↓ miR-143 ↑ miR-27b
↓ miR-181a ↑ miR-29a
Voluntary wheel
↓ miR-191a ↑ miR-29c

RI
↓ miR-208a ↑ miR-30b
↓ miR-208b ↑ miR-30e
↓ miR-214 ↑ miR-126
↑ miR-150
↑ miR-133b
↑ miR-222
↑ miR-144

SC
↑ miR-145
↑ miR-208a

B c-miR (plasma)
Acute

NU
↑ miR-1 Chronic
↑ miR-133a
↑ miR-133b
↑ miR-139
↑ miR-143
↑ miR-181b ↓ let-7d
↑ miR-206 ↓ miR-16
MA
↓ let-7i
↑ miR-208b ↓ miR-21
↓ miR-30b ↑ miR-20a
↑ miR-214 ↑ miR-21 ↓ miR-25
↓ miR-106a ↑ miR-103
↑ miR-223 ↑ miR-146a ↓ miR-27a
↓ miR-146a ↑ miR-107
↑ miR-330 ↑ miR-221 ↓ miR-28
↓ miR-151 ↑ miR-126
↑ miR-338 ↑ miR-222 ↓ miR-148a
↓ miR-221 ↑ miR-376a
↑ miR-485 ↓ miR-185
↓ miR-652
↑ miR-509 ↓ miR-342
↑ miR-517a ↓ miR-766
↑ miR-518f
↑ miR-520f
ED

↑ miR-522
↑ miR-553
↑ miR-888

Fig 2
PT
CE
AC
ACCEPTED MANUSCRIPT
37

Table 1. Reported changes in miRNA expression in mouse models.

miRNA

PT
Phenotype
Target Organ/System Type of Exercise* Reference
Up Down Influenced
mmu-miR-150 ___ ___ Heart Cardiac hypertrophy Voluntary wheel (35 days) 213

RI
Hipk1 Cardiomyocyte growth 214
mmu-miR-222 ___ Heart Voluntary wheel (3 wks)
Hmbox1 and proliferation

SC
Bcl-2
mmu-miR-21 p53 Swimming (2 times/day, 5 215
mmu-miR-1 Heart Cardiac apoptosis

NU
mmu-miR-30b Pdcd4 days/week, 8 weeks)
Drp-1
Swimming (90 min, 2
Cardiac growth

MA
Timp-3 times/day, 21 days) or 216
mmu- miR-17 ___ Heart and myocyte
Pten Voluntary wheel exercise (21
proliferation
days)

ED
mmu-miR-1 Skeletal muscle Acute exercise bout
mmu-miR-107 mmu-miR-23 Pgc-1α (quadriceps ___ (motorized treadmill, 15 122

mmu-miR-181 femoris) m/min for 90 min)

PT
Chronic training (motorized
___ mmu-miR-696 Pgc-1α CE
Skeletal muscle
(gastrocnemius)
Metabolism
treadmill, 5 times/wk, 18-32
m/min, 20-60 min per
120

session, 4 wk)
AC
Chronic training (motorized
Pgc-1α Skeletal muscle treadmill, 5 days/wk, 28 217
___ mmu-miR-761 Metabolism
p-Mk2 (gastrocnemius) m/min, 45 min per session, 4
wk)
Nrf1
Mitochondrial
mmu-miR-494 Bim Skeletal muscle 218
___ biogenesis and Voluntary wheel (8 wks)
mmu-miR-696 Bcl-xl (gastrocnemius)
apoptosis
Pgc-1α
Acute (motorized treadmill,
Skeletal muscle
Igf-1 Mitochondrial 14 m/min, 10% grade, 60 219
mmu-miR-133a ___ (extensor
Akt biogenesis min) or Chronic (motorized
digitorum longus)
treadmill, 14 m/min, 10%
ACCEPTED MANUSCRIPT
38

grade, 60 min, 5 d/wk, 6 wks)


exercises
Pten

PT
mmu-miR-23a Skeletal muscle Resistance exercise (muscle 220
___ Casp7 Muscle atrophy
mmu-miR-27a (tibialis anterior) overload)
FoxO1

RI
Chronic training (motorized
mmu-miR-192 Circulation 221
___ ___ ___ treadmill, 1 h/d, 5 d/wk,
mmu-miR-193b (serum)

SC
22m/min, 5 wks)
mmu-miR-7a
mmu-miR-15b

NU
mmu-miR-28a
Hdac3 Brain Voluntary wheel running
mmu-miR-98 ___ Senescence 200
Hdac5 (hippocampus) (8 weeks)
mmu-miR-105

MA
mmu-miR-133b
mmu-miR-148b

ED
PT
CE
AC
ACCEPTED MANUSCRIPT
39

Table 2. Reported changes in miRNA expression in rat models.

miRNA

PT
Phenotype
Target Organ/System Type of Exercise* Reference
Up Down Influenced
Swimming (60 min, 5

RI
rno-miR-27a Ace
rno-miR-143 Heart Cardiac hypertrophy days/week, 5% caudal body 110
rno-miR-27b Ace2
weight workload, 10 wks)

SC
Swimming (60 min, 5 d/wk,
ColIaI
rno-miR-29c ___ Heart Ventricular compliance 5% caudal body weight 113
ColIIIaI
workload, 10 wks)

NU
Swimming (60 min, 5 d/wk,
Spred-1
rno-miR-126 ___ Heart Angiogenesis 5% caudal body weight 116
Pi3kr2
workload, 10 wks)

MA
rno-miR-21 Pten Swimming (60 min, 5 d/wk,
rno-miR-144 rno-miR-124 Pik3a Heart Cardiac hypertrophy 5% caudal body weight 111

rno-miR-145 Tsc2 workload, 8 wks)

ED
Cardiac fibrosis Swimming (60 min, 5 d/wk,
rno-miR-29a ColIaI
___ Heart induced by myocardial 5% caudal body weight 112
rno-miR-29c ColIIIaI
infarction workload, 10 wks)

PT
Swimming (60 min, 5
Ncx 222
rno-miR-1 rno-miR-214 Heart Ca+2 handling days/wk, 3% caudal body
Serca2a
miR-22
CE weight workload, 10 wks)
rno-miR-19b Igf1
miR-99b Swimming (90 min, 2 x/day,
rno-miR-30e PI3/Akt/mTor
AC

miR-100 Heart Apoptosis 5 d/week, 5% caudal body 223


rno-miR-208a Mapk
miR-181a weight workload, 8 wks)
rno-miR-133b p53
miR-191a
Med13
Pur Swimming (60 min, 5
rno-miR-208a Cardiac hypertrophy 224
___ Sox6 Heart days/week, 5% caudal body
rno-miR-208b and metabolism
Hp1 weight workload, 10 wks)
Sp3
rno-miR-96
rno-miR-216a Treadmill (1 h/d, 5 d/wk, 15 225
rno-mir-425 ___ Heart Cardiad protection
rno-miR-484 m/min, 10 wks)
rno-miR-488
ACCEPTED MANUSCRIPT
40

Resistance exercise (4 × 12
___ rno-miR-214 Serca2a Heart Ca+2 handling bouts, 5 x/wk, 8 wks, 80% 226
1-repetition maximum)

PT
Swimming (60 min, 5
rno-miR-16 Bcl-2 Skeletal Muscle
rno-miR-126 Vascularization days/week, 4% caudal body 117
rno-miR-21 Pi3kr2 (soleus)

RI
weight workload, 10 weeks)
Pten

SC
Pdcd4 Cycling (5 days/week, 45
rno-miR-21 rno-miR-15b Ras Spinal cord Apoptosis rpm, 2x 30 min sessions, 10- 196

Casp7 31 days)

NU
Casp9
Cycling (5 days/week, 45
Pten
rno-miR-21 rno-miR-199a Spinal cord Neuron regeneration rpm, 2x 30 min sessions, 10- 119

MA
Mtor
31 days)

ED
PT
CE
AC
ACCEPTED MANUSCRIPT
41

Table 3. Reported changes in miRNA expression in human.

miRNA

PT
Phenotype
Target Organ/System Type of Exercise* Reference
Up Down Influenced
hsa-miR-1 hsa-miR-9

RI
Acute exercise bout (cycle
hsa-miR-133a hsa-miR-23a HDAC4 Skeletal Muscle
___ ergometer, 60 min, 70% 71
hsa-miR-133b hsa-miR-23b NRF1 (vastus lateralis)

SC
VO2peak)
hsa-miR-181a hsa-miR-31
hsa-miR-1 Acute resistance exercise
Skeletal Muscle
___ hsa-miR-23a ___ ___ (one-legged knee extension, 227

NU
(vastus lateralis)
hsa-miR-133a 45 min, 60% of wattmax)
Acute resistance exercise
hsa-miR-1
Skeletal Muscle (two-legged knee extension, 8

MA
___ hsa-miR-133a ___ ___ 228
(vastus lateralis) sets of 10 repetitions, 70% of
hsa-miR-206
their 1 RM)
hsa-miR-1 Chronic training (cycle

ED
hsa-miR-133a Skeletal Muscle ergometer, 5x/wk, 12 wks,
___ ___ ___ 86
hsa-miR-133b (vastus lateralis) 60-120min/section, 55-91%
hsa-miR-206 of Pmax)

PT
Chronic resistance exercise
Skeletal Muscle (leg press, knee extension, leg
___ hsa-miR-1 IGF-1 Hypertrophy 124
CE
(vastus lateralis) curl, hip extension, 3x/8–10
repetitions, 12 wks)
Muscle regeneration,
AC

hsa-miR-1 HDAC4 Skeletal Muscle gene transcription and 71


___ Endurance (cyclying, 10 days)
hsa-miR-29b NRF1 (vastus lateralis) mitochondrial
biogenesis
MCT (cycling, 30 min, 80%
lactate threshold), HIIT
hsa-miR-133a (cycling, 10 times/2 min,
Skeletal Muscle 229
___ hsa-miR-378 mTORC1 ___ 120% lactate threshold) or
(vastus lateralis)
hsa-miR-486 Resistance exercise (8 x 5 leg
press repetitions, 80% of 1-
repetition maximum)
hsa-miR-136 hsa-miR-28 Skeletal Muscle Hypertrophy Chronic resistance exercise (8 230
___
hsa-miR-200c hsa-miR-30d (vastus lateralis) (high or low x 5 leg press repetitions, 80%
ACCEPTED MANUSCRIPT
42

hsa-miR-376a hsa-miR-204 responders) of 1-repetition maximum, 12


hsa-miR-377 hsa-miR-330 wks)
hsa-miR-499b hsa-miR-345

PT
hsa-miR-558 hsa-miR-375
hsa-miR-449c

RI
hsa-miR-483
hsa-miR-509

SC
hsa-miR-520a
hsa-miR-548an
hsa-miR-628

NU
hsa-miR-653
hsa-miR-670
hsa-miR-889

MA
hsa-miR-1245a
hsa-miR-1270
hsa-miR-1280
hsa-miR-1322

ED
hsa-miR-3180
hsa-miR-26a
Skeletal Muscle

PT
hsa-miR-451 hsa-miR-29a ___ Skeletal mass gain High vs. low responders 114
(vastus lateralis)
hsa-miR-378
hsa-miR-125a hsa-let-7i
hsa-miR-145 hsa-miR-16
CE
hsa-miR-181b hsa-miR-17
hsa-miR-193a hsa-miR-18a
AC

hsa-miR-197 hsa-miR-18b
hsa-miR-212 hsa-miR-20a
hsa-miR-223 hsa-miR-20b Acute exercise bout (cycle
Circulating
hsa-miR-340 hsa-miR-22 ergometer, 10x 2-min bouts,
___ (neutrophils Immune system 88
hsa-miR-365 hsa-miR-93 1-min rest interval between
cells)
hsa-miR-485 hsa-miR-96 each bout, 76% VO2peak)
hsa-miR-505 hsa-miR-106a
hsa-miR-520d hsa-miR-107
hsa-miR-629 hsa-miR-126
hsa-miR-638 hsa-miR-130a
hsa-miR-939 hsa-miR-130b
hsa-miR-940 hsa-miR-151
ACCEPTED MANUSCRIPT
43

hsa-miR-1225 hsa-miR-185
hsa-miR-1238 hsa-miR-194
hsa-miR-363

PT
hsa-miR-660
hsa-let-7e

RI
hsa-miR-23b
hsa-miR-7 hsa-miR-31

SC
hsa-miR-15a hsa-miR-99a
hsa-miR-21 hsa-miR-125a
hsa-miR-26b hsa-miR-125b

NU
hsa-miR-132 hsa-miR-126
Circulating
hsa-miR-140 hsa-miR-130a Acute exercise bout (cycle
(peripheral
hsa-miR-181a hsa-miR-145 ergometer, 10x 2-min bouts,

MA
___ blood Immune system 87
hsa-miR-181b hsa-miR-151 1-min rest interval between
mononuclear
hsa-miR-181c hsa-miR-199a each bout, 76% VO2peak)
cells)
hsa-miR-338 hsa-miR-199b
hsa-miR-363 hsa-miR-221

ED
hsa-miR-939 hsa-miR-320
hsa-miR-940 hsa-miR-451

PT
hsa-miR-1225 hsa-miR-486
hsa-miR-584
hsa-miR-652
hsa-miR-let-7f-1 hsa-miR-let-7f-1
CE
Circulating
(peripheral Chronic exercise training
hsa-miR-21 hsa-miR-21 VO2peak response to 231
___ blood (running, 3 x/wk, 60mi, 18
AC

hsa-miR-29c hsa-miR-29c exercise training


mononuclear wks)
hsa-miR-223 hsa-miR-223
cells
hsa-miR-7 hsa-let-7e
hsa-miR-29a hsa-miR-126
hsa-miR-29b hsa-miR-130a
hsa-miR-29c hsa-miR-151 Acute exercise bout (cycle
Circulating
hsa-miR-30e hsa-miR-199a ergometer, 10x 2-min bouts,
___ (natural killers Immune system 102
hsa-miR-142 hsa-miR-221 1-min rest interval between
cells)
hsa-miR-192 hsa-miR-223 each bout, 77% VO2peak)
hsa-miR-338 hsa-miR-326
hsa-miR-363 hsa-miR-328
hsa-miR-590 hsa-miR-652
ACCEPTED MANUSCRIPT
44

hsa-miR-15a
hsa-miR-29b
hsa-miR-29c

PT
hsa-miR-30e
hsa-miR-23b
hsa-miR-140
hsa-miR-130a Acute exercise bout (10x 2

RI
hsa-miR-324 Circulating
hsa-miR-151 ___ Immune system min bouts of cycle ergometer 118
hsa-miR-338 (Monocytes)
hsa-miR-199a exercise, 82% VO2max)

SC
hsa-miR-362
hsa-miR-221
hsa-miR-532
hsa-miR-660

NU
hsa-miR-1202
hsa-miR-1305
Circulation

MA
(skeletal muscle Acute exercise bout
hsa-miR-133b 232
___ ___ releases CRF (treadmill, 40 min, 1% grade,
hsa-miR-181a
extracellular 80% VO2max)
vesicles)

ED
Cardiac rehabilitation
hsa-miR-92a Circulating
___ ___ ___ programme (60 min, 2/wk, 178
hsa-miR-92b (whole blood)

PT
10 wks)
hsa-miR-1
Circulating Endurance athletes (runners,
hsa-miR-486 ___ ___ VO2max 233

hsa-miR-494
CE
(whole blood) cyclists, and triathletes)
hsa-miR-21
Acute exhaustive
hsa-miR-146a
AC

cardiopulmonary exercise
hsa-miR-221
test
hsa-miR-222
Circulating
___ ___ ___ 77
hsa-miR-20a (plasma)
Chronic exercise training
hsa-miR-21
(rowing training, 5 km, 1-3 h
hsa-miR-146a
per session, 20-24
hsa-miR-221
strokes/min, 90 days)
hsa-miR-222
hsa-miR-1
Acute exercise bout (30-min
hsa-miR-133a Circulating
___ ___ ___ downhill and uphill walking 79
hsa-miR-133b (plasma)
exercises)
hsa-miR-181b
ACCEPTED MANUSCRIPT
45

hsa-miR-208b
hsa-miR-214
hsa-let-7i Acute exercise bout (60min

PT
hsa-miR-139 hsa-miR-30b cycle ergometer exercise bout
hsa-miR-143 hsa-miR-106a at 65% of Pmax)

RI
hsa-miR-223 hsa-miR-146
hsa-miR-330 hsa-miR-151

SC
hsa-miR-338 hsa-miR-221
hsa-miR-652
Circulating
___ ___ Chronic training (cycle 123
(plasma)

NU
hsa-miR-103 hsa-let-7d ergometer, 5x/wk, 12 wks,
hsa-miR-107 hsa-miR-21 60-120min/section, 55-91%
hsa-miR-25 of Pmax)

MA
hsa-miR-148a
hsa-miR-185
hsa-miR-342
hsa-miR-766

ED
hsa-miR-1
hsa-miR-133a

PT
hsa-miR-133b
hsa-miR-206 Acute HIIE (motorized
hsa-miR-485 treadmill, 7 x 4 min, ∼85–
hsa-miR-509
___ ___
CE
Circulating
___
95% of HRmax) vs. VICE 234
hsa-miR-517a (plasma) (motorized treadmill,
hsa-miR-518f matched distance covered
AC

hsa-miR-520f during the HIIE trial)


hsa-miR-522
hsa-miR-553
hsa-miR-888
High-Volume Training (130
min, 55% peak power output)
hsa-miR-21 Circulating vs. High-Intensity Training
___ ___ ___ 235
hsa-miR-126 (plasma) (4×4 min, 95% peak power
output) vs. Sprint-Interval
Training (4×30s all-out).
hsa-miR-16 Circulating Aerobic exercise training
hsa-miR-376a ___ ___ 236
hsa-miR-27a (plasma) (motorized treadmill, 4
ACCEPTED MANUSCRIPT
46

hsa-miR-28 days/wk, 5 months)

hsa-miR-140

PT
Circulating High vs. low responders
hsa-miR-221 ___ ___ Body weight 237
(plasma) (losses in body weight)
hsa-miR-223

RI
hsa-miR-1
hsa-miR-133a
Circulating

SC
hsa-miR-206 ___ ___ VO2max Marathon run 85
(plasma)
hsa-miR-208b
hsa-miR-499

NU
hsa-miR-1
hsa-miR-126
hsa-miR-133a Circulating

MA
___ ___ ___ Marathon run 78
hsa-miR-134 (plasma)
hsa-miR-146a
hsa-miR-499
Acute exercise bout (single

ED
bout of steady-state cycling
Circulating exercise, 70% VO2max, 60min)
___ hsa-miR-486 ___ ___ 76

PT
(serum)
Chronic exercise (cycling
training 3 d/wk, 4 wks)
CE Acute resistance exercise
(bench press and leg press, 5
hsa-miR-146a Circulating
hsa-miR-149 ___ ___ sets of 10 repetitions, 70% of
AC
90
hsa-miR-221 (serum)
maximum strength, 1 min rest
between sets)
hsa-miR-19a Acute resistance exercise
hsa-miR-19b bout (3 times, bilateral knee
hsa-miR-20a p-AKTSer473 Circulating extension exercise + bilateral
___ ___ 238
hsa-miR-26b p-S6K1Thr389 (serum) leg press exercise, 10
hsa-miR-143 repetitions, 80% of 1-
hsa-miR-195 repetition maximum.)
Acute exhaustive exercise
hsa-miR-21
Circulating (cycle ergometer, starting
hsa-miR-378 ___ ___ ___ 239
(serum) from 20 J/s for 2 min and
hsa-miR-940
increased by 5J/s every 30s,
ACCEPTED MANUSCRIPT
47

60 rpm)
Chronic training (cycle
hsa-miR-192 Circulation 221
___ ___ ___ ergometer, 60-85% VO2max, 2

PT
hsa-miR-193b (serum)
times/wk, 16 wks)
hsa-miR-210 Circulating
hsa-miR-21 ___ VO2max High vs. low VO2max 121

RI
hsa-miR-222 (serum)

SC
Genetic
Markers
hsa-let-7a TMEM8A SNP (rs540) VO2max ___ 126

NU
MCT, moderate-intensity continuous training; HIIT, high-intensity interval training.
HIIE, high-intensity interval exercise; VICE, Vigorous-intensity continuous exercise.

MA
CRF, Cardiorespiratory fitness.

ED
PT
CE
AC
ACCEPTED MANUSCRIPT

References

1. Malik S, Wong ND, Franklin SS, et al. Impact of the metabolic syndrome on
mortality from coronary heart disease, cardiovascular disease, and all
causes in United States adults. Circulation. 2004;110:1245-1250.
2. Kavanagh T, Mertens DJ, Hamm LF, et al. Peak oxygen intake and cardiac

PT
mortality in women referred for cardiac rehabilitation. J Am Coll Cardiol.
2003;42:2139-2143.
3. Myers J, Prakash M, Froelicher V, Do D, Partington S, Atwood JE. Exercise

RI
capacity and mortality among men referred for exercise testing. N Engl J
Med. 2002;346:793-801.
4. Yusuf S, Hawken S, Ounpuu S, et al. Effect of potentially modifiable risk

SC
factors associated with myocardial infarction in 52 countries (the
INTERHEART study): case-control study. Lancet. 2004;364:937-952.
5. Hakim AA, Petrovitch H, Burchfiel CM, et al. Effects of walking on

NU
mortality among nonsmoking retired men. N Engl J Med. 1998;338:94-99.
6. Mora S, Cook N, Buring JE, Ridker PM, Lee IM. Physical activity and
reduced risk of cardiovascular events: potential mediating mechanisms.
MA
Circulation. 2007;116:2110-2118.
7. Farrell SW, Cheng YJ, Blair SN. Prevalence of the metabolic syndrome
across cardiorespiratory fitness levels in women. Obes Res. 2004;12:824-
830.
ED

8. Hatle H, Stobakk PK, Molmen HE, et al. Effect of 24 sessions of high-


intensity aerobic interval training carried out at either high or moderate
frequency, a randomized trial. PLoS One. 2014;9:e88375.
PT

9. Kavanagh T, Mertens DJ, Hamm LF, et al. Prediction of long-term


prognosis in 12 169 men referred for cardiac rehabilitation. Circulation.
2002;106:666-671.
CE

10. LaMonte MJ, Barlow CE, Jurca R, Kampert JB, Church TS, Blair SN.
Cardiorespiratory fitness is inversely associated with the incidence of
metabolic syndrome: a prospective study of men and women. Circulation.
2005;112:505-512.
AC

11. Weston KS, Wisloff U, Coombes JS. High-intensity interval training in


patients with lifestyle-induced cardiometabolic disease: a systematic
review and meta-analysis. Br J Sports Med. 2013.
12. Bouchard C, Antunes-Correa LM, Ashley EA, et al. Personalized preventive
medicine: genetics and the response to regular exercise in preventive
interventions. Prog Cardiovasc Dis. 2015;57:337-346.
13. Fletcher GF, Balady GJ, Amsterdam EA, et al. Exercise standards for testing
and training: a statement for healthcare professionals from the American
Heart Association. Circulation. 2001;104:1694-1740.
14. Leon AS, Franklin BA, Costa F, et al. Cardiac rehabilitation and secondary
prevention of coronary heart disease: an American Heart Association
scientific statement from the Council on Clinical Cardiology
(Subcommittee on Exercise, Cardiac Rehabilitation, and Prevention) and
the Council on Nutrition, Physical Activity, and Metabolism
(Subcommittee on Physical Activity), in collaboration with the American
association of Cardiovascular and Pulmonary Rehabilitation. Circulation.
2005;111:369-376.
ACCEPTED MANUSCRIPT
49

15. Adair TH, Gay WJ, Montani JP. Growth regulation of the vascular system:
evidence for a metabolic hypothesis. Am J Physiol. 1990;259:R393-404.
16. Gute D, Fraga C, Laughlin MH, Amann JF. Regional changes in capillary
supply in skeletal muscle of high-intensity endurance-trained rats. J Appl
Physiol. 1996;81:619-626.
17. Howald H, Hoppeler H, Claassen H, Mathieu O, Straub R. Influences of

PT
endurance training on the ultrastructural composition of the different
muscle fiber types in humans. Pflugers Arch. 1985;403:369-376.
18. Saltin B, Gollnik PD. Skeletal muscle adaptability: significance for

RI
metabolism and performance. In: Soc AP, ed. Handbook of Physiology.
Bethesda, MD1983:555-632.
19. Tjonna AE, Leinan IM, Bartnes AT, et al. Low- and high-volume of

SC
intensive endurance training significantly improves maximal oxygen
uptake after 10-weeks of training in healthy men. PLoS One.
2013;8:e65382.

NU
20. Pedersen LR, Olsen RH, Frederiksen M, et al. Copenhagen study of
overweight patients with coronary artery disease undergoing low energy
diet or interval training: the randomized CUT-IT trial protocol. BMC
MA
Cardiovasc Disord. 2013;13:106.
21. Larsen I, Welde B, Martins C, Tjonna AE. High- and moderate-intensity
aerobic exercise and excess post-exercise oxygen consumption in men
with metabolic syndrome. Scand J Med Sci Sports. 2013.
ED

22. Bronstad E, Tjonna AE, Rognmo O, et al. Aerobic exercise training


improves right- and left ventricular systolic function in patients with
COPD. COPD. 2013;10:300-306.
PT

23. Rognmo O, Moholdt T, Bakken H, et al. Cardiovascular risk of high- versus


moderate-intensity aerobic exercise in coronary heart disease patients.
Circulation. 2012;126:1436-1440.
CE

24. Moholdt T, Bekken Vold M, Grimsmo J, Slordahl SA, Wisloff U. Home-


based aerobic interval training improves peak oxygen uptake equal to
residential cardiac rehabilitation: a randomized, controlled trial. PLoS One.
2012;7:e41199.
AC

25. Moholdt T, Aamot IL, Granoien I, et al. Aerobic interval training increases
peak oxygen uptake more than usual care exercise training in myocardial
infarction patients: a randomized controlled study. Clin Rehabil.
2012;26:33-44.
26. Bronstad E, Rognmo O, Tjonna AE, et al. High-intensity knee extensor
training restores skeletal muscle function in COPD patients. Eur Respir J.
2012;40:1130-1136.
27. Moholdt T, Aamot IL, Granoien I, et al. Long-term follow-up after cardiac
rehabilitation: a randomized study of usual care exercise training versus
aerobic interval training after myocardial infarction. Int J Cardiol.
2011;152:388-390.
28. Stensvold D, Tjonna AE, Skaug EA, et al. Strength training versus aerobic
interval training to modify risk factors of metabolic syndrome. J Appl
Physiol (1985). 2010;108:804-810.
29. Tjonna AE, Stolen TO, Bye A, et al. Aerobic interval training reduces
cardiovascular risk factors more than a multitreatment approach in
overweight adolescents. Clin Sci (Lond). 2009;116:317-326.
ACCEPTED MANUSCRIPT
50

30. Moholdt TT, Amundsen BH, Rustad LA, et al. Aerobic interval training
versus continuous moderate exercise after coronary artery bypass
surgery: a randomized study of cardiovascular effects and quality of life.
Am Heart J. 2009;158:1031-1037.
31. Tjonna AE, Lee SJ, Rognmo O, et al. Aerobic interval training versus
continuous moderate exercise as a treatment for the metabolic syndrome:

PT
a pilot study. Circulation. 2008;118:346-354.
32. Wisloff U, Stoylen A, Loennechen JP, et al. Superior cardiovascular effect
of aerobic interval training versus moderate continuous training in heart

RI
failure patients: a randomized study. Circulation. 2007;115:3086-3094.
33. Ferguson S, Gledhill N, Jamnik VK, Wiebe C, Payne N. Cardiac performance
in endurance-trained and moderately active young women. Med Sci Sports

SC
Exerc. 2001;33:1114-1119.
34. Gledhill N, Cox D, Jamnik R. Endurance athletes' stroke volume does not
plateau: major advantage is diastolic function. Med Sci Sports Exerc.

NU
1994;26:1116-1121.
35. Schaible TF, Scheuer J. Cardiac function in hypertrophied hearts from
chronically exercised female rats. J Appl Physiol Respir Environ Exerc
MA
Physiol. 1981;50:1140-1145.
36. Wisloff U, Najjar SM, Ellingsen O, et al. Cardiovascular risk factors emerge
after artificial selection for low aerobic capacity. Science. 2005;307:418-
420.
ED

37. Wisloff U, Loennechen JP, Currie S, Smith GL, Ellingsen O. Aerobic exercise
reduces cardiomyocyte hypertrophy and increases contractility, Ca2+
sensitivity and SERCA-2 in rat after myocardial infarction. Cardiovasc Res.
PT

2002;54:162-174.
38. Belke DD, Swanson EA, Dillmann WH. Decreased sarcoplasmic reticulum
activity and contractility in diabetic db/db mouse heart. Diabetes.
CE

2004;53:3201-3208.
39. Bers DM, Despa S. Cardiac myocytes Ca2+ and Na+ regulation in normal
and failing hearts. J Pharmacol Sci. 2006;100:315-322.
40. Brum PC, Kosek J, Patterson A, Bernstein D, Kobilka B. Abnormal cardiac
AC

function associated with sympathetic nervous system hyperactivity in


mice. Am J Physiol Heart Circ Physiol. 2002;283:H1838-1845.
41. Respress JL, van Oort RJ, Li N, et al. Role of RyR2 phosphorylation at
S2814 during heart failure progression. Circ Res. 2012;110:1474-1483.
42. Kemi OJ, Ceci M, Condorelli G, Smith GL, Wisloff U. Myocardial
sarcoplasmic reticulum Ca2+ ATPase function is increased by aerobic
interval training. Eur J Cardiovasc Prev Rehabil. 2008;15:145-148.
43. Kemi OJ, Ceci M, Wisloff U, et al. Activation or inactivation of cardiac
Akt/mTOR signaling diverges physiological from pathological
hypertrophy. J Cell Physiol. 2008;214:316-321.
44. Kemi OJ, Haram PM, Wisloff U, Ellingsen O. Aerobic fitness is associated
with cardiomyocyte contractile capacity and endothelial function in
exercise training and detraining. Circulation. 2004;109:2897-2904.
45. Kemi OJ, Loennechen JP, Wisloff U, Ellingsen O. Intensity-controlled
treadmill running in mice: cardiac and skeletal muscle hypertrophy. J Appl
Physiol (1985). 2002;93:1301-1309.
ACCEPTED MANUSCRIPT
51

46. Wisloff U, Helgerud J, Kemi OJ, Ellingsen O. Intensity-controlled treadmill


running in rats: VO(2 max) and cardiac hypertrophy. Am J Physiol Heart
Circ Physiol. 2001;280:H1301-1310.
47. Brum PC, Bacurau AV, Cunha TF, Bechara LR, Moreira JB. Skeletal
myopathy in heart failure: effects of aerobic exercise training. Exp Physiol.
2014;99:616-620.

PT
48. Brum PC, Bacurau AV, Medeiros A, Ferreira JC, Vanzelli AS, Negrao CE.
Aerobic exercise training in heart failure: impact on sympathetic
hyperactivity and cardiac and skeletal muscle function. Braz J Med Biol

RI
Res. 2011;44:827-835.
49. Middlekauff HR. Making the case for skeletal myopathy as the major
limitation of exercise capacity in heart failure. Circ Heart Fail. 2010;3:537-

SC
546.
50. Schiaffino S, Reggiani C. Fiber types in mammalian skeletal muscles.
Physiol Rev. 2011;91:1447-1531.

NU
51. Zizola C, Schulze PC. Metabolic and structural impairment of skeletal
muscle in heart failure. Heart Fail Rev. 2013;18:623-630.
52. Cunha TF, Bacurau AV, Moreira JB, et al. Exercise training prevents
MA
oxidative stress and ubiquitin-proteasome system overactivity and
reverse skeletal muscle atrophy in heart failure. PLoS One. 2012;7:e41701.
53. Jannig PR, Moreira JB, Bechara LR, et al. Autophagy signaling in skeletal
muscle of infarcted rats. PLoS One. 2014;9:e85820.
ED

54. Moreira JB, Bechara LR, Bozi LH, et al. High- versus moderate-intensity
aerobic exercise training effects on skeletal muscle of infarcted rats. J Appl
Physiol (1985). 2013;114:1029-1041.
PT

55. Voltarelli VA, Bechara LR, Bacurau AV, et al. Lack of beta -adrenoceptors
aggravates heart failure-induced skeletal muscle myopathy in mice. J Cell
Mol Med. 2014.
CE

56. Bacurau AV, Belmonte MA, Navarro F, et al. Effect of a high-intensity


exercise training on the metabolism and function of macrophages and
lymphocytes of walker 256 tumor bearing rats. Exp Biol Med (Maywood).
2007;232:1289-1299.
AC

57. Bueno CR, Jr., Ferreira JC, Pereira MG, Bacurau AV, Brum PC. Aerobic
exercise training improves skeletal muscle function and Ca2+ handling-
related protein expression in sympathetic hyperactivity-induced heart
failure. J Appl Physiol (1985). 2010;109:702-709.
58. Kraljevic J, Marinovic J, Pravdic D, et al. Aerobic interval training
attenuates remodelling and mitochondrial dysfunction in the post-
infarction failing rat heart. Cardiovasc Res. 2013;99:55-64.
59. Oliveira RS, Ferreira JC, Gomes ER, et al. Cardiac anti-remodelling effect of
aerobic training is associated with a reduction in the calcineurin/NFAT
signalling pathway in heart failure mice. J Physiol. 2009;587:3899-3910.
60. Rolim NP, Medeiros A, Rosa KT, et al. Exercise training improves the net
balance of cardiac Ca2+ handling protein expression in heart failure.
Physiol Genomics. 2007;29:246-252.
61. Stolen TO, Hoydal MA, Kemi OJ, et al. Interval training normalizes
cardiomyocyte function, diastolic Ca2+ control, and SR Ca2+ release
synchronicity in a mouse model of diabetic cardiomyopathy. Circ Res.
2009;105:527-536.
ACCEPTED MANUSCRIPT
52

62. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function.


Cell. 2004;116:281-297.
63. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of
novel genes coding for small expressed RNAs. Science. 2001;294:853-858.
64. Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs
with probable regulatory roles in Caenorhabditis elegans. Science.

PT
2001;294:858-862.
65. Lee RC, Ambros V. An extensive class of small RNAs in Caenorhabditis
elegans. Science. 2001;294:862-864.

RI
66. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4
encodes small RNAs with antisense complementarity to lin-14. Cell.
1993;75:843-854.

SC
67. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the
heterochronic gene lin-14 by lin-4 mediates temporal pattern formation
in C. elegans. Cell. 1993;75:855-862.

NU
68. Chalfie M, Horvitz HR, Sulston JE. Mutations that lead to reiterations in the
cell lineages of C. elegans. Cell. 1981;24:59-69.
69. van Rooij E, Marshall WS, Olson EN. Toward microRNA-based
MA
therapeutics for heart disease: the sense in antisense. Circ Res.
2008;103:919-928.
70. Snyder LL, Ahmed I, Steel LF. RNA polymerase III can drive polycistronic
expression of functional interfering RNAs designed to resemble
ED

microRNAs. Nucleic Acids Res. 2009;37:e127.


71. Russell AP, Lamon S, Boon H, et al. Regulation of miRNAs in human
skeletal muscle following acute endurance exercise and short-term
PT

endurance training. J Physiol. 2013;591:4637-4653.


72. Chen X, Ba Y, Ma L, et al. Characterization of microRNAs in serum: a novel
class of biomarkers for diagnosis of cancer and other diseases. Cell Res.
CE

2008;18:997-1006.
73. Lawrie CH, Gal S, Dunlop HM, et al. Detection of elevated levels of tumour-
associated microRNAs in serum of patients with diffuse large B-cell
lymphoma. Br J Haematol. 2008;141:672-675.
AC

74. Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable
blood-based markers for cancer detection. Proc Natl Acad Sci U S A.
2008;105:10513-10518.
75. Weber JA, Baxter DH, Zhang S, et al. The microRNA spectrum in 12 body
fluids. Clin Chem. 2010;56:1733-1741.
76. Aoi W, Ichikawa H, Mune K, et al. Muscle-enriched microRNA miR-486
decreases in circulation in response to exercise in young men. Front
Physiol. 2013;4:80.
77. Baggish AL, Hale A, Weiner RB, et al. Dynamic regulation of circulating
microRNA during acute exhaustive exercise and sustained aerobic
exercise training. J Physiol. 2011;589:3983-3994.
78. Baggish AL, Park J, Min PK, et al. Rapid upregulation and clearance of
distinct circulating microRNAs after prolonged aerobic exercise. J Appl
Physiol (1985). 2014;116:522-531.
79. Banzet S, Chennaoui M, Girard O, et al. Changes in circulating microRNAs
levels with exercise modality. J Appl Physiol (1985). 2013;115:1237-1244.
ACCEPTED MANUSCRIPT
53

80. Corsten MF, Dennert R, Jochems S, et al. Circulating MicroRNA-208b and


MicroRNA-499 reflect myocardial damage in cardiovascular disease. Circ
Cardiovasc Genet. 2010;3:499-506.
81. Fichtlscherer S, De Rosa S, Fox H, et al. Circulating microRNAs in patients
with coronary artery disease. Circ Res. 2010;107:677-684.
82. Ji X, Takahashi R, Hiura Y, Hirokawa G, Fukushima Y, Iwai N. Plasma miR-

PT
208 as a biomarker of myocardial injury. Clin Chem. 2009;55:1944-1949.
83. Laterza OF, Lim L, Garrett-Engele PW, et al. Plasma MicroRNAs as
sensitive and specific biomarkers of tissue injury. Clin Chem.

RI
2009;55:1977-1983.
84. Lorenzen JM, Kielstein JT, Hafer C, et al. Circulating miR-210 predicts
survival in critically ill patients with acute kidney injury. Clin J Am Soc

SC
Nephrol. 2011;6:1540-1546.
85. Mooren FC, Viereck J, Kruger K, Thum T. Circulating micrornas as
potential biomarkers of aerobic exercise capacity. Am J Physiol Heart Circ

NU
Physiol. 2014;306:H557-563.
86. Nielsen S, Scheele C, Yfanti C, et al. Muscle specific microRNAs are
regulated by endurance exercise in human skeletal muscle. J Physiol.
MA
2010;588:4029-4037.
87. Radom-Aizik S, Zaldivar F, Jr., Leu SY, Adams GR, Oliver S, Cooper DM.
Effects of exercise on microRNA expression in young males peripheral
blood mononuclear cells. Clin Transl Sci. 2012;5:32-38.
ED

88. Radom-Aizik S, Zaldivar F, Jr., Oliver S, Galassetti P, Cooper DM. Evidence


for microRNA involvement in exercise-associated neutrophil gene
expression changes. J Appl Physiol. 2010;109:252-261.
PT

89. Rosjo H, Dahl MB, Bye A, et al. Prognostic Value of Circulating MicroRNA-
210 Levels in Patients with Moderate to Severe Aortic Stenosis. PLoS One.
2014;9:e91812.
CE

90. Sawada S, Kon M, Wada S, Ushida T, Suzuki K, Akimoto T. Profiling of


Circulating MicroRNAs after a Bout of Acute Resistance Exercise in
Humans. PLoS One. 2013;8:e70823.
91. Hunter MP, Ismail N, Zhang X, et al. Detection of microRNA expression in
AC

human peripheral blood microvesicles. PLoS One. 2008;3:e3694.


92. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-
mediated transfer of mRNAs and microRNAs is a novel mechanism of
genetic exchange between cells. Nat Cell Biol. 2007;9:654-659.
93. Zernecke A, Bidzhekov K, Noels H, et al. Delivery of microRNA-126 by
apoptotic bodies induces CXCL12-dependent vascular protection. Sci
Signal. 2009;2:ra81.
94. Arroyo JD, Chevillet JR, Kroh EM, et al. Argonaute2 complexes carry a
population of circulating microRNAs independent of vesicles in human
plasma. Proc Natl Acad Sci U S A. 2011;108:5003-5008.
95. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT.
MicroRNAs are transported in plasma and delivered to recipient cells by
high-density lipoproteins. Nat Cell Biol. 2011;13:423-433.
96. Kosaka N, Iguchi H, Ochiya T. Circulating microRNA in body fluid: a new
potential biomarker for cancer diagnosis and prognosis. Cancer Sci.
2010;101:2087-2092.
ACCEPTED MANUSCRIPT
54

97. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T.


Secretory mechanisms and intercellular transfer of microRNAs in living
cells. J Biol Chem. 2010;285:17442-17452.
98. Halkein J, Tabruyn SP, Ricke-Hoch M, et al. MicroRNA-146a is a
therapeutic target and biomarker for peripartum cardiomyopathy. J Clin
Invest. 2013;123:2143-2154.

PT
99. Hergenreider E, Heydt S, Treguer K, et al. Atheroprotective
communication between endothelial cells and smooth muscle cells
through miRNAs. Nat Cell Biol. 2012;14:249-256.

RI
100. Chim SS, Shing TK, Hung EC, et al. Detection and characterization of
placental microRNAs in maternal plasma. Clin Chem. 2008;54:482-490.
101. Gilad S, Meiri E, Yogev Y, et al. Serum microRNAs are promising novel

SC
biomarkers. PLoS One. 2008;3:e3148.
102. Radom-Aizik S, Zaldivar F, Haddad F, Cooper DM. Impact of brief exercise
on peripheral blood NK cell gene and microRNA expression in young

NU
adults. J Appl Physiol (1985). 2013;114:628-636.
103. Uhlemann M, Mobius-Winkler S, Fikenzer S, et al. Circulating microRNA-
126 increases after different forms of endurance exercise in healthy
MA
adults. Eur J Prev Cardiol. 2012;21:484-491.
104. Adachi T, Nakanishi M, Otsuka Y, et al. Plasma microRNA 499 as a
biomarker of acute myocardial infarction. Clin Chem. 2010;56:1183-1185.
105. Ai J, Zhang R, Li Y, et al. Circulating microRNA-1 as a potential novel
ED

biomarker for acute myocardial infarction. Biochem Biophys Res Commun.


2010;391:73-77.
106. Cortez MA, Calin GA. MicroRNA identification in plasma and serum: a new
PT

tool to diagnose and monitor diseases. Expert Opin Biol Ther. 2009;9:703-
711.
107. Wang GK, Zhu JQ, Zhang JT, et al. Circulating microRNA: a novel potential
CE

biomarker for early diagnosis of acute myocardial infarction in humans.


Eur Heart J. 2010;31:659-666.
108. Wong TS, Liu XB, Wong BY, Ng RW, Yuen AP, Wei WI. Mature miR-184 as
Potential Oncogenic microRNA of Squamous Cell Carcinoma of Tongue.
AC

Clin Cancer Res. 2008;14:2588-2592.


109. Bye A, Rosjo H, Nauman J, et al. Circulating microRNAs predict future fatal
myocardial infarction in healthy individuals - The HUNT study. J Mol Cell
Cardiol. 2016;97:162-168.
110. Fernandes T, Hashimoto NY, Magalhaes FC, et al. Aerobic exercise
training-induced left ventricular hypertrophy involves regulatory
MicroRNAs, decreased angiotensin-converting enzyme-angiotensin ii, and
synergistic regulation of angiotensin-converting enzyme 2-angiotensin
(1-7). Hypertension. 2011;58:182-189.
111. Ma Z, Qi J, Meng S, Wen B, Zhang J. Swimming exercise training-induced
left ventricular hypertrophy involves microRNAs and synergistic
regulation of the PI3K/AKT/mTOR signaling pathway. Eur J Appl Physiol.
2013;113:2473-2486.
112. Melo SF, Fernandes T, Barauna VG, et al. Expression of MicroRNA-29 and
Collagen in Cardiac Muscle after Swimming Training in Myocardial-
Infarcted Rats. Cell Physiol Biochem. 2014;33:657-669.
ACCEPTED MANUSCRIPT
55

113. Soci UP, Fernandes T, Hashimoto NY, et al. MicroRNAs 29 are involved in
the improvement of ventricular compliance promoted by aerobic exercise
training in rats. Physiol Genomics. 2011;43:665-673.
114. Davidsen PK, Gallagher IJ, Hartman JW, et al. High responders to
resistance exercise training demonstrate differential regulation of skeletal
muscle microRNA expression. J Appl Physiol. 2011;110:309-317.

PT
115. McCarthy JJ, Esser KA. MicroRNA-1 and microRNA-133a expression are
decreased during skeletal muscle hypertrophy. J Appl Physiol.
2007;102:306-313.

RI
116. da Silva ND J, Fernandes T, Soci UP, Monteiro AW, Phillips MI, EM DEO.
Swimming training in rats increases cardiac MicroRNA-126 expression
and angiogenesis. Med Sci Sports Exerc. 2012;44:1453-1462.

SC
117. Fernandes T, Magalhaes FC, Roque FR, Phillips MI, Oliveira EM. Exercise
Training Prevents the Microvascular Rarefaction in Hypertension
Balancing Angiogenic and Apoptotic Factors: Role of MicroRNAs-16, -21,

NU
and -126. Hypertension. 2012;59:513-520.
118. Radom-Aizik S, Zaldivar FP, Jr., Haddad F, Cooper DM. Impact of brief
exercise on circulating monocyte gene and microRNA expression:
MA
Implications for atherosclerotic vascular disease. Brain Behav Immun.
2014.
119. Liu G, Detloff MR, Miller KN, Santi L, Houle JD. Exercise modulates
microRNAs that affect the PTEN/mTOR pathway in rats after spinal cord
ED

injury. Exp Neurol. 2012;233:447-456.


120. Aoi W, Naito Y, Mizushima K, et al. The microRNA miR-696 regulates PGC-
1{alpha} in mouse skeletal muscle in response to physical activity. Am J
PT

Physiol Endocrinol Metab. 2010;298:E799-806.


121. Bye A, Rosjo H, Aspenes ST, Condorelli G, Omland T, Wisloff U. Circulating
microRNAs and aerobic fitness--the HUNT-Study. PLoS One.
CE

2013;8:e57496.
122. Safdar A, Abadi A, Akhtar M, Hettinga BP, Tarnopolsky MA. miRNA in the
regulation of skeletal muscle adaptation to acute endurance exercise in
C57Bl/6J male mice. PLoS One. 2009;4:e5610.
AC

123. Nielsen S, Akerstrom T, Rinnov A, et al. The miRNA Plasma Signature in


Response to Acute Aerobic Exercise and Endurance Training. PLoS One.
2014;9:e87308.
124. Mueller M, Breil FA, Lurman G, et al. Different molecular and structural
adaptations with eccentric and conventional strength training in elderly
men and women. Gerontology. 2011;57:528-538.
125. Care A, Catalucci D, Felicetti F, et al. MicroRNA-133 controls cardiac
hypertrophy. Nat Med. 2007;13:613-618.
126. Silva GJJ, Almeida M, Bye A, et al. Non-Coding Genetic Variants Associated
with Aerobic Capacity: the HUNT Study. (unpublished).
127. Leptidis S, El Azzouzi H, Lok SI, et al. A deep sequencing approach to
uncover the miRNOME in the human heart. PLoS One. 2013;8:e57800.
128. Chen JF, Murchison EP, Tang R, et al. Targeted deletion of Dicer in the
heart leads to dilated cardiomyopathy and heart failure. Proc Natl Acad
Sci U S A. 2008;105:2111-2116.
ACCEPTED MANUSCRIPT
56

129. da Costa Martins PA, Bourajjaj M, Gladka M, et al. Conditional dicer gene
deletion in the postnatal myocardium provokes spontaneous cardiac
remodeling. Circulation. 2008;118:1567-1576.
130. Kwon C, Han Z, Olson EN, Srivastava D. MicroRNA1 influences cardiac
differentiation in Drosophila and regulates Notch signaling. Proc Natl
Acad Sci U S A. 2005;102:18986-18991.

PT
131. Sokol NS, Ambros V. Mesodermally expressed Drosophila microRNA-1 is
regulated by Twist and is required in muscles during larval growth. Genes
Dev. 2005;19:2343-2354.

RI
132. Zhao Y, Ransom JF, Li A, et al. Dysregulation of cardiogenesis, cardiac
conduction, and cell cycle in mice lacking miRNA-1-2. Cell. 2007;129:303-
317.

SC
133. da Costa Martins PA, Salic K, Gladka MM, et al. MicroRNA-199b targets the
nuclear kinase Dyrk1a in an auto-amplification loop promoting
calcineurin/NFAT signalling. Nat Cell Biol. 2010;12:1220-1227.

NU
134. Dirkx E, Gladka MM, Philippen LE, et al. Nfat and miR-25 cooperate to
reactivate the transcription factor Hand2 in heart failure. Nat Cell Biol.
2013;15:1282-1293.
MA
135. el Azzouzi H, Leptidis S, Dirkx E, et al. The hypoxia-inducible microRNA
cluster miR-199a approximately 214 targets myocardial PPARdelta and
impairs mitochondrial fatty acid oxidation. Cell Metab. 2013;18:341-354.
136. Heymans S, Corsten MF, Verhesen W, et al. Macrophage microRNA-155
ED

promotes cardiac hypertrophy and failure. Circulation. 2013;128:1420-


1432.
137. Schober A, Nazari-Jahantigh M, Wei Y, et al. MicroRNA-126-5p promotes
PT

endothelial proliferation and limits atherosclerosis by suppressing Dlk1.


Nat Med. 2014.
138. Thum T, Gross C, Fiedler J, et al. MicroRNA-21 contributes to myocardial
CE

disease by stimulating MAP kinase signalling in fibroblasts. Nature.


2008;456:980-984.
139. van Rooij E, Sutherland LB, Thatcher JE, et al. Dysregulation of microRNAs
after myocardial infarction reveals a role of miR-29 in cardiac fibrosis.
AC

Proc Natl Acad Sci U S A. 2008;105:13027-13032.


140. Yang B, Lin H, Xiao J, et al. The muscle-specific microRNA miR-1 regulates
cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med.
2007;13:486-491.
141. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an
essential role in the development of cardiac hypertrophy. Circ Res.
2007;100:416-424.
142. Ikeda S, Kong SW, Lu J, et al. Altered microRNA expression in human heart
disease. Physiol Genomics. 2007;31:367-373.
143. van Rooij E, Sutherland LB, Liu N, et al. A signature pattern of stress-
responsive microRNAs that can evoke cardiac hypertrophy and heart
failure. Proc Natl Acad Sci U S A. 2006;103:18255-18260.
144. Brum PC, Da Silva GJ, Moreira ED, Ida F, Negrao CE, Krieger EM. Exercise
training increases baroreceptor gain sensitivity in normal and
hypertensive rats. Hypertension. 2000;36:1018-1022.
ACCEPTED MANUSCRIPT
57

145. Silva GJ, Brum PC, Negrao CE, Krieger EM. Acute and chronic effects of
exercise on baroreflexes in spontaneously hypertensive rats. Hypertension.
1997;30:714-719.
146. Allen DL, Harrison BC, Maass A, Bell ML, Byrnes WC, Leinwand LA.
Cardiac and skeletal muscle adaptations to voluntary wheel running in
the mouse. J Appl Physiol (1985). 2001;90:1900-1908.

PT
147. Konhilas JP, Maass AH, Luckey SW, Stauffer BL, Olson EN, Leinwand LA.
Sex modifies exercise and cardiac adaptation in mice. Am J Physiol Heart
Circ Physiol. 2004;287:H2768-2776.

RI
148. Evangelista FS, Brum PC, Krieger JE. Duration-controlled swimming
exercise training induces cardiac hypertrophy in mice. Braz J Med Biol Res.
2003;36:1751-1759.

SC
149. Medeiros A, Oliveira EM, Gianolla R, Casarini DE, Negrao CE, Brum PC.
Swimming training increases cardiac vagal activity and induces cardiac
hypertrophy in rats. Braz J Med Biol Res. 2004;37:1909-1917.

NU
150. Barauna VG, Batista ML, Jr., Costa Rosa LF, Casarini DE, Krieger JE,
Oliveira EM. Cardiovascular adaptations in rats submitted to a resistance-
training model. Clin Exp Pharmacol Physiol. 2005;32:249-254.
MA
151. Barauna VG, Rosa KT, Irigoyen MC, de Oliveira EM. Effects of resistance
training on ventricular function and hypertrophy in a rat model. Clin Med
Res. 2007;5:114-120.
152. Mostarda CT, Rodrigues B, de Cxu Moraes OA, et al. Low intensity
ED

resistance training improves systolic function and cardiovascular


autonomic control in diabetic rats. J Diabetes Complications. 2013.
153. Fenning A, Harrison G, Dwyer D, Rose'Meyer R, Brown L. Cardiac
PT

adaptation to endurance exercise in rats. Mol Cell Biochem. 2003;251:51-


59.
154. Zhang XQ, Song J, Carl LL, et al. Effects of sprint training on contractility
CE

and [Ca(2+)](i) transients in adult rat myocytes. J Appl Physiol (1985).


2002;93:1310-1317.
155. Fernandes T, Soci UP, Oliveira EM. Eccentric and concentric cardiac
hypertrophy induced by exercise training: microRNAs and molecular
AC

determinants. Braz J Med Biol Res. 2011;44:836-847.


156. Patel V, Noureddine L. MicroRNAs and fibrosis. Curr Opin Nephrol
Hypertens. 2012;21:410-416.
157. Roderburg C, Urban GW, Bettermann K, et al. Micro-RNA profiling reveals
a role for miR-29 in human and murine liver fibrosis. Hepatology.
2011;53:209-218.
158. Wang B, Komers R, Carew R, et al. Suppression of microRNA-29
expression by TGF-beta1 promotes collagen expression and renal fibrosis.
J Am Soc Nephrol. 2012;23:252-265.
159. Cushing L, Kuang PP, Qian J, et al. miR-29 is a major regulator of genes
associated with pulmonary fibrosis. Am J Respir Cell Mol Biol.
2011;45:287-294.
160. Roncarati R, Viviani Anselmi C, Losi MA, et al. Circulating miR-29a, Among
Other Up-Regulated MicroRNAs, Is the Only Biomarker for Both
Hypertrophy and Fibrosis in Patients With Hypertrophic Cardiomyopathy.
J Am Coll Cardiol. 2014;63:920-927.
ACCEPTED MANUSCRIPT
58

161. Slagsvold KH, Rognmo O, Hoydal M, Wisloff U, Wahba A. Remote Ischemic


Preconditioning Preserves Mitochondrial Function and Influences
Myocardial MicroRNA Expression in Atrial Myocardium During Coronary
Bypass Surgery. Circ Res. 2014;114:851-859.
162. Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the
management of cancer and other diseases. Nat Rev Drug Discov.

PT
2017;16:203-222.
163. Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by
targeting microRNA. N Engl J Med. 2013;368:1685-1694.

RI
164. Pasiakos SM, McClung JP. miRNA analysis for the assessment of exercise
and amino acid effects on human skeletal muscle. Adv Nutr. 2013;4:412-
417.

SC
165. Zacharewicz E, Lamon S, Russell AP. MicroRNAs in skeletal muscle and
their regulation with exercise, ageing, and disease. Front Physiol.
2013;4:266.

NU
166. McCarthy JJ. MicroRNA-206: the skeletal muscle-specific myomiR. Biochim
Biophys Acta. 2008;1779:682-691.
167. McCarthy JJ. The MyomiR network in skeletal muscle plasticity. Exerc
MA
Sport Sci Rev. 2011;39:150-154.
168. van Rooij E, Quiat D, Johnson BA, et al. A family of microRNAs encoded by
myosin genes governs myosin expression and muscle performance. Dev
Cell. 2009;17:662-673.
ED

169. Chen JF, Mandel EM, Thomson JM, et al. The role of microRNA-1 and
microRNA-133 in skeletal muscle proliferation and differentiation. Nat
Genet. 2006;38:228-233.
PT

170. Cacchiarelli D, Martone J, Girardi E, et al. MicroRNAs involved in


molecular circuitries relevant for the Duchenne muscular dystrophy
pathogenesis are controlled by the dystrophin/nNOS pathway. Cell Metab.
CE

2010;12:341-351.
171. Liu N, Williams AH, Kim Y, et al. An intragenic MEF2-dependent enhancer
directs muscle-specific expression of microRNAs 1 and 133. Proc Natl
Acad Sci U S A. 2007;104:20844-20849.
AC

172. Yamamoto H, Morino K, Nishio Y, et al. MicroRNA-494 regulates


mitochondrial biogenesis in skeletal muscle through mitochondrial
transcription factor A and Forkhead box j3. Am J Physiol Endocrinol Metab.
2012;303:E1419-1427.
173. Bouchard C, An P, Rice T, et al. Familial aggregation of VO(2max) response
to exercise training: results from the HERITAGE Family Study. J Appl
Physiol (1985). 1999;87:1003-1008.
174. Skinner JS, Jaskolski A, Jaskolska A, et al. Age, sex, race, initial fitness, and
response to training: the HERITAGE Family Study. J Appl Physiol (1985).
2001;90:1770-1776.
175. Ai J, Zhang R, Li Y, et al. Circulating microRNA-1 as a potential novel
biomarker for acute myocardial infarction. Biochem Biophys Res Commun.
2010;391:73-77.
176. Tijsen AJ, Creemers EE, Moerland PD, et al. MiR423-5p as a circulating
biomarker for heart failure. Circ Res. 2010;106:1035-1039.
ACCEPTED MANUSCRIPT
59

177. Li T, Cao H, Zhuang J, et al. Identification of miR-130a, miR-27b and miR-


210 as serum biomarkers for atherosclerosis obliterans. Clin Chim Acta.
2011;412:66-70.
178. Taurino C, Miller WH, McBride MW, et al. Gene expression profiling in
whole blood of patients with coronary artery disease. Clin Sci (Lond).
2010;119:335-343.

PT
179. Clauss S, Wakili R, Hildebrand B, et al. MicroRNAs as Biomarkers for Acute
Atrial Remodeling in Marathon Runners (The miRathon Study--A Sub-
Study of the Munich Marathon Study). PLoS One. 2016;11:e0148599.

RI
180. Chen J, Yang T, Yu H, et al. A functional variant in the 3'-UTR of
angiopoietin-1 might reduce stroke risk by interfering with the binding
efficiency of microRNA 211. Hum Mol Genet. 2010;19:2524-2533.

SC
181. Son MS, Jang MJ, Jeon YJ, et al. Promoter polymorphisms of pri-miR-34b/c
are associated with hepatocellular carcinoma. Gene. 2013;524:156-160.
182. Amin AS, Giudicessi JR, Tijsen AJ, et al. Variants in the 3' untranslated

NU
region of the KCNQ1-encoded Kv7.1 potassium channel modify disease
severity in patients with type 1 long QT syndrome in an allele-specific
manner. Eur Heart J. 2012;33:714-723.
MA
183. Clop A, Marcq F, Takeda H, et al. A mutation creating a potential
illegitimate microRNA target site in the myostatin gene affects
muscularity in sheep. Nat Genet. 2006;38:813-818.
184. Dorn GW, 2nd, Matkovich SJ, Eschenbacher WH, Zhang Y. A human 3' miR-
ED

499 mutation alters cardiac mRNA targeting and function. Circ Res.
2012;110:958-967.
185. Rotunno M, Zhao Y, Bergen AW, et al. Inherited polymorphisms in the
PT

RNA-mediated interference machinery affect microRNA expression and


lung cancer survival. Br J Cancer. 2010;103:1870-1874.
186. Kim JS, Choi YY, Jin G, et al. Association of a common AGO1 variant with
CE

lung cancer risk: a two-stage case-control study. Mol Carcinog.


2010;49:913-921.
187. Liu ME, Liao YC, Lin RT, et al. A functional polymorphism of PON1
interferes with microRNA binding to increase the risk of ischemic stroke
AC

and carotid atherosclerosis. Atherosclerosis. 2013;228:161-167.


188. Thomas LF, Saetrom P. Single nucleotide polymorphisms can create
alternative polyadenylation signals and affect gene expression through
loss of microRNA-regulation. PLoS Comput Biol. 2012;8:e1002621.
189. McComas AJ. Neuromuscular Functional and Disorders. London, England:
Butterworth & Co Ltd; 1977.
190. Vrbová G, Gordon T, Jones R. Nerve-Muscle Interaction: Springer; 1994.
191. Houle JD, Morris K, Skinner RD, Garcia-Rill E, Peterson CA. Effects of fetal
spinal cord tissue transplants and cycling exercise on the soleus muscle in
spinalized rats. Muscle Nerve. 1999;22:846-856.
192. Beaumont E, Houle JD, Peterson CA, Gardiner PF. Passive exercise and
fetal spinal cord transplant both help to restore motoneuronal properties
after spinal cord transection in rats. Muscle Nerve. 2004;29:234-242.
193. Ollivier-Lanvin K, Keeler BE, Siegfried R, Houle JD, Lemay MA.
Proprioceptive neuropathy affects normalization of the H-reflex by
exercise after spinal cord injury. Exp Neurol. 2010;221:198-205.
ACCEPTED MANUSCRIPT
60

194. Hutchinson KJ, Gomez-Pinilla F, Crowe MJ, Ying Z, Basso DM. Three
exercise paradigms differentially improve sensory recovery after spinal
cord contusion in rats. Brain. 2004;127:1403-1414.
195. Sandrow-Feinberg HR, Izzi J, Shumsky JS, Zhukareva V, Houle JD. Forced
exercise as a rehabilitation strategy after unilateral cervical spinal cord
contusion injury. J Neurotrauma. 2009;26:721-731.

PT
196. Liu G, Keeler BE, Zhukareva V, Houle JD. Cycling exercise affects the
expression of apoptosis-associated microRNAs after spinal cord injury in
rats. Exp Neurol. 2010;226:200-206.

RI
197. Baker LD, Frank LL, Foster-Schubert K, et al. Effects of aerobic exercise on
mild cognitive impairment: a controlled trial. Arch Neurol. 2010;67:71-79.
198. Erickson KI, Voss MW, Prakash RS, et al. Exercise training increases size of

SC
hippocampus and improves memory. Proc Natl Acad Sci U S A.
2011;108:3017-3022.
199. Lautenschlager NT, Cox KL, Flicker L, et al. Effect of physical activity on

NU
cognitive function in older adults at risk for Alzheimer disease: a
randomized trial. JAMA. 2008;300:1027-1037.
200. Cosin-Tomas M, Alvarez-Lopez MJ, Sanchez-Roige S, et al. Epigenetic
MA
alterations in hippocampus of SAMP8 senescent mice and modulation by
voluntary physical exercise. Front Aging Neurosci. 2014;6:51.
201. Morley JE, Farr SA, Kumar VB, Armbrecht HJ. The SAMP8 mouse: a model
to develop therapeutic interventions for Alzheimer's disease. Curr Pharm
ED

Des. 2012;18:1123-1130.
202. Walsh NP, Gleeson M, Pyne DB, et al. Position statement. Part two:
Maintaining immune health. Exerc Immunol Rev. 2011;17:64-103.
PT

203. Walsh NP, Gleeson M, Shephard RJ, et al. Position statement. Part one:
Immune function and exercise. Exerc Immunol Rev. 2011;17:6-63.
204. Bacurau RF, Belmonte MA, Seelaender MC, Costa Rosa LF. Effect of a
CE

moderate intensity exercise training protocol on the metabolism of


macrophages and lymphocytes of tumour-bearing rats. Cell Biochem Funct.
2000;18:249-258.
205. Navarro F, Bacurau AV, Pereira GB, et al. Moderate exercise increases the
AC

metabolism and immune function of lymphocytes in rats. Eur J Appl


Physiol. 2013;113:1343-1352.
206. Radom-Aizik S, Zaldivar F, Jr., Leu SY, Galassetti P, Cooper DM. Effects of
30 min of aerobic exercise on gene expression in human neutrophils. J
Appl Physiol. 2008;104:236-243.
207. Fernhall B, Borghi-Silva A, Babu AS. The future of physical activity
research: funding, opportunities and challenges. Prog Cardiovasc Dis.
2015;57:299-305.
208. O'Keefe JH, Lavie CJ, Guazzi M. Part 1: potential dangers of extreme
endurance exercise: how much is too much? Part 2: screening of school-
age athletes. Prog Cardiovasc Dis. 2015;57:396-405.
209. Ounzain S, Burdet F, Ibberson M, Pedrazzini T. Discovery and functional
characterization of cardiovascular long noncoding RNAs. J Mol Cell Cardiol.
2015;89:17-26.
210. Dal-Pra S, Hodgkinson CP, Mirotsou M, Kirste I, Dzau VJ. Demethylation of
H3K27 Is Essential for the Induction of Direct Cardiac Reprogramming by
miR Combo. Circ Res. 2017;120:1403-1413.
ACCEPTED MANUSCRIPT
61

211. Jayawardena TM, Finch EA, Zhang L, et al. MicroRNA induced cardiac
reprogramming in vivo: evidence for mature cardiac myocytes and
improved cardiac function. Circ Res. 2015;116:418-424.
212. Li Y, Dal-Pra S, Mirotsou M, et al. Tissue-engineered 3-dimensional (3D)
microenvironment enhances the direct reprogramming of fibroblasts into
cardiomyocytes by microRNAs. Sci Rep. 2016;6:38815.

PT
213. Martinelli NC, Cohen CR, Santos KG, et al. An analysis of the global
expression of microRNAs in an experimental model of physiological left
ventricular hypertrophy. PLoS One. 2014;9:e93271.

RI
214. Liu X, Xiao J, Zhu H, et al. miR-222 Is Necessary for Exercise-Induced
Cardiac Growth and Protects against Pathological Cardiac Remodeling.
Cell Metab. 2015;21:584-595.

SC
215. Zhao Y, Ma Z. Swimming training affects apoptosis-related microRNAs
and reduces cardiac apoptosis in mice. Gen Physiol Biophys. 2016;35:443-
450.

NU
216. Shi J, Bei Y, Kong X, et al. miR-17-3p Contributes to Exercise-Induced
Cardiac Growth and Protects against Myocardial Ischemia-Reperfusion
Injury. Theranostics. 2017;7:664-676.
MA
217. Xu Y, Zhao C, Sun X, Liu Z, Zhang J. MicroRNA-761 regulates mitochondrial
biogenesis in mouse skeletal muscle in response to exercise. Biochem
Biophys Res Commun. 2015;467:103-108.
218. Sun Y, Cui D, Zhang Z, Zhang Q, Ji L, Ding S. Voluntary wheel exercise
ED

alters the levels of miR-494 and miR-696 in the skeletal muscle of


C57BL/6 mice. Comp Biochem Physiol B Biochem Mol Biol. 2016;202:16-22.
219. Nie Y, Sato Y, Wang C, Yue F, Kuang S, Gavin TP. Impaired exercise
PT

tolerance, mitochondrial biogenesis, and muscle fiber maintenance in


miR-133a-deficient mice. FASEB J. 2016;30:3745-3758.
220. Wang B, Zhang C, Zhang A, Cai H, Price SR, Wang XH. MicroRNA-23a and
CE

MicroRNA-27a Mimic Exercise by Ameliorating CKD-Induced Muscle


Atrophy. J Am Soc Nephrol. 2017.
221. Parrizas M, Brugnara L, Esteban Y, et al. Circulating miR-192 and miR-
193b are markers of prediabetes and are modulated by an exercise
AC

intervention. J Clin Endocrinol Metab. 2015;100:E407-415.


222. Melo SF, Barauna VG, Neves VJ, et al. Exercise training restores the cardiac
microRNA-1 and -214 levels regulating Ca2+ handling after myocardial
infarction. BMC Cardiovasc Disord. 2015;15:166.
223. Ramasamy S, Velmurugan G, Shanmugha Rajan K, Ramprasath T, Kalpana
K. MiRNAs with Apoptosis Regulating Potential Are Differentially
Expressed in Chronic Exercise-Induced Physiologically Hypertrophied
Hearts. PLoS One. 2015;10:e0121401.
224. Soci UP, Fernandes T, Barauna VG, et al. Epigenetic control of exercise
training-induced cardiac hypertrophy by miR-208. Clin Sci (Lond). 2016.
225. Souza RW, Fernandez GJ, Cunha JP, et al. Regulation of cardiac microRNAs
induced by aerobic exercise training during heart failure. Am J Physiol
Heart Circ Physiol. 2015;309:H1629-1641.
226. Melo SF, Barauna VG, Junior MA, et al. Resistance training regulates
cardiac function through modulation of miRNA-214. Int J Mol Sci.
2015;16:6855-6867.
ACCEPTED MANUSCRIPT
62

227. Ringholm S, Bienso RS, Kiilerich K, et al. Bed rest reduces metabolic
protein content and abolishes exercise-induced mRNA responses in
human skeletal muscle. Am J Physiol Endocrinol Metab. 2011;301:E649-
658.
228. Drummond MJ, McCarthy JJ, Fry CS, Esser KA, Rasmussen BB. Aging
differentially affects human skeletal muscle microRNA expression at rest

PT
and after an anabolic stimulus of resistance exercise and essential amino
acids. Am J Physiol Endocrinol Metab. 2008;295:E1333-1340.
229. Fyfe JJ, Bishop DJ, Zacharewicz E, Russell AP, Stepto NK. Concurrent

RI
exercise incorporating high-intensity interval or continuous training
modulates mTORC1 signaling and microRNA expression in human
skeletal muscle. Am J Physiol Regul Integr Comp Physiol. 2016;310:R1297-

SC
1311.
230. Ogasawara R, Akimoto T, Umeno T, Sawada S, Hamaoka T, Fujita S.
MicroRNA expression profiling in skeletal muscle reveals different

NU
regulatory patterns in high and low responders to resistance training.
Physiol Genomics. 2016;48:320-324.
231. Dias RG, Silva MS, Duarte NE, et al. PBMCs express a transcriptome
MA
signature predictor of oxygen uptake responsiveness to endurance
exercise training in men. Physiol Genomics. 2015;47:13-23.
232. Guescini M, Canonico B, Lucertini F, et al. Muscle Releases Alpha-
Sarcoglycan Positive Extracellular Vesicles Carrying miRNAs in the
ED

Bloodstream. PLoS One. 2015;10:e0125094.


233. Denham J, Prestes PR. Muscle-Enriched MicroRNAs Isolated from Whole
Blood Are Regulated by Exercise and Are Potential Biomarkers of
PT

Cardiorespiratory Fitness. Front Genet. 2016;7:196.


234. Cui SF, Wang C, Yin X, et al. Similar Responses of Circulating MicroRNAs to
Acute High-Intensity Interval Exercise and Vigorous-Intensity Continuous
CE

Exercise. Front Physiol. 2016;7:102.


235. Wahl P, Wehmeier UF, Jansen FJ, et al. Acute Effects of Different Exercise
Protocols on the Circulating Vascular microRNAs -16, -21, and -126 in
Trained Subjects. Front Physiol. 2016;7:643.
AC

236. Zhang T, Brinkley TE, Liu K, et al. Circulating MiRNAs as biomarkers of


gait speed responses to aerobic exercise training in obese older adults.
Aging (Albany NY). 2017;9:900-913.
237. Parr EB, Camera DM, Burke LM, Phillips SM, Coffey VG, Hawley JA.
Circulating MicroRNA Responses between 'High' and 'Low' Responders to
a 16-Wk Diet and Exercise Weight Loss Intervention. PLoS One.
2016;11:e0152545.
238. Margolis LM, Lessard SJ, Ezzyat Y, Fielding RA, Rivas DA. Circulating
MicroRNA Are Predictive of Aging and Acute Adaptive Response to
Resistance Exercise in Men. J Gerontol A Biol Sci Med Sci. 2016.
239. Xu T, Zhou Q, Che L, et al. Circulating miR-21, miR-378, and miR-940
increase in response to an acute exhaustive exercise in chronic heart
failure patients. Oncotarget. 2016;7:12414-12425.

You might also like