Vous êtes sur la page 1sur 15

Downloaded from http://perspectivesinmedicine.cshlp.

org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise and the Skeletal Muscle Epigenome

Sean L. McGee and Ken R. Walder


Metabolic Research Unit, School of Medicine and Centre for Molecular and Medical Research, Deakin
University, Geelong, Victoria 3216, Australia
Correspondence: sean.mcgee@deakin.edu.au

An acute bout of exercise is sufficient to induce changes in skeletal muscle gene expression
that are ultimately responsible for the adaptive responses to exercise. Although much re-
search has described the intracellular signaling responses to exercise that are linked to
transcriptional regulation, the epigenetic mechanisms involved are only just emerging.
This review will provide an overview of epigenetic mechanisms and what is known in the
context of exercise. Additionally, we will explore potential interactions between metabolism
during exercise and epigenetic regulation, which serves as a framework for potential areas for
future research. Finally, we will consider emerging opportunities to pharmacologically ma-
nipulate epigenetic regulators and mechanisms to induce aspects of the skeletal muscle
exercise adaptive response for therapeutic intervention in various disease states.
www.perspectivesinmedicine.org

egular exercise evokes a number of adaptive cle can exert by increasing its contractile units,
R responses in skeletal muscle that contribute
to alterations in muscle function and physical
cross-sectional area, and the connective tissue
structures that assist in force transmission and
fitness. Indeed, skeletal muscle is a highly plastic muscle stabilization (Nader et al. 2014). Further-
tissue that can rapidly adapt to the energetic and more, strength exercise also increases the ability
mechanical demands placed on it through of skeletal muscle to produce energy through
repeated bouts of exercise. In response to anaerobic pathways (Egan and Zierath 2013).
endurance exercise, skeletal muscle increases its Many of these phenotypic adaptations are
capacity to produce energy through aerobic me- driven by transient alterations in the expression
tabolism by enhancing delivery of substrates of genes involved in various aspects of skeletal
and oxygen secondary to increasing capillary muscle function (Egan and Zierath 2013). The
density (Clausen and Trap-Jensen 1970). The expression of some genes increases during exer-
capacity for skeletal muscle substrate uptake is cise while others increase in the postexercise
also enhanced through a greater abundance of period. These transient changes in gene expres-
sarcolemmal glucose and fatty acid transporters sion result in persistent alterations in the levels
(Glatz et al. 2002). In parallel, the capacity to of the proteins that they encode, which ulti-
oxidize these substrates to produce ATP is en- mately produce a change in skeletal muscle phe-
hanced through increased mitochondrial size notype (Egan and Zierath 2013). A wealth of
and number (Holloszy 1967). In contrast, research has described many of the intracellular
strength exercise increases the power that a mus- signaling mechanisms that drive exercise-medi-

Editors: Juleen R. Zierath, Michael J. Joyner, and John A. Hawley


Additional Perspectives on The Biology of Exercise available at www.perspectivesinmedicine.org
Copyright # 2017 Cold Spring Harbor Laboratory Press; all rights reserved
Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

ated changes in gene expression, which include DNMTs are encoded in many genomes, from
activation of kinase cascades, transcription fac- bacteria to plants and mammals. The evolution-
tors, and transcriptional coregulators (Egan and ary conservation of these enzymes suggests that
Zierath 2013). It is well established that another DNA methylation provides a selective advantage
layer of biological regulation, termed epigenet- to the organism. In mammals, DNA methyla-
ics, also controls gene expression. Although tion typically occurs at CpG-rich regions of the
there is conjecture on the exact definition of genome, known as CpG islands, which are en-
the term (Henikoff and Greally 2016), epigenet- riched in the promoters and first exons of genes
ics is generally considered to describe heritable (Klose and Bird 2006). When the CpG island
changes in gene regulation that occur indepen- becomes methylated, the transcription of the
dently of changes in DNA sequence, but instead cognate genes will be blocked (Lister et al.
are caused by chemical modification of the var- 2009; Ziller et al. 2013). However, DNA methyl-
ious components of chromatin, which regulates ation also occurs outside CpG islands in mam-
the access of the transcriptional machinery to mals, and this also contributes the regulation of
DNA (Kouzarides 2007). gene transcription, including in skeletal muscle
This review will provide a brief overview of (Barres et al. 2009). DNA methylation is gener-
the most commonly studied epigenetic mecha- ally associated with a reduction in gene expres-
nisms before describing the nascent field of sion, thought to be mediated by affecting the
exercise epigenetics. We will also speculate on binding of transcriptional activators to promot-
potential interactions between exercise metab- er regions (Fig. 1) (Nakao 2001), although the
olism and epigenetic regulation before provid- precise mechanism involved is still under inves-
ing an assessment of the potential usage of tigation.
drugs that act by modulating epigenetic modi- DNA methylation patterns and the resultant
www.perspectivesinmedicine.org

fiers to induce aspects of the exercise adaptive effects on gene transcription are a critical part of
response in skeletal muscle. normal development programs, and these pro-
cesses are controlled by the precise regulation of
the DNMTs. Aberrant DNA methylation, often
OVERVIEW OF EPIGENETIC MECHANISMS
associated with altered levels or dysfunction of
Epigenetic modification of the fundamental the DNMTs, has been observed in a range of
components of chromatin, which includes the developmental diseases, such as neural tube de-
histone protein core and DNA, influences how fects (Chang et al. 2011), as well as adult dis-
surrounding genes are regulated. These modifi- eases, including a range of cancers (Robertson
cations can be broadly categorized into those 2005; Hamidi et al. 2015), but also in complex
affecting DNA, and those affecting histone pro- chronic diseases such as schizophrenia (Gavin
teins (Kouzarides 2007). The following section and Sharma 2010) and autism spectrum disor-
will provide an overview of the best-character- der (Jiang et al. 2004; Nagarajan et al. 2006).
ized epigenetic modifications and how they im- DNMT1 is regarded as a maintenance en-
pact on transcriptional regulation. zyme whose primary role is to preserve methyl-
ation patterns during DNA replication and mi-
tosis, which would otherwise alter methylation
DNA Methylation
patterns (Denis et al. 2011). DNMT3A and
DNA methylation involves the enzymatic addi- DNMT3B are the principal methyltransferases
tion of a methyl group to the fifth carbon of involved in establishing de novo DNA methyl-
a cytosine residue, yielding 5-methylcytosine ation patterns in somatic and differentiated cell
(Nakao 2001). This process is catalyzed by a types (Shen and Laird 2013; Bedi et al. 2014).
family of enzymes known as DNA methyltrans- Emerging evidence suggests that DNA methyl-
ferases (DNMTs) (Suzuki and Bird 2008), and ation patterns can be dynamically changed in
is regarded as one of the fundamental epigenet- response to various physiological challenges, in-
ic mechanisms to regulate gene transcription. cluding diet (Amaral et al. 2014; Silva-Martinez

2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise Epigenetics

Exercise metabolism ?
?
αHG
αKG succinate
fumurate
TET

Transcriptional Transcription initiation complex


repression DNA demethylation

Me Me Me Me

DNA methylation
Transcriptional
activation

DNMTs + SAM

Methionine

Figure 1. DNA methylation and the regulation of transcription. DNA methylation (Me), controlled by DNA
methyltransferases (DNMTs) using S-adenosylmethionine (SAM) as the methyl donor, prevents transcriptional
activators such as the transcriptional initiation complex from gaining access to gene regulatory regions. De-
methylation of DNA, initiated by the ten-eleven translocation (TET) enzymes, exposes DNA regulatory regions
to transcriptional activators. The TET enzymes are activated by the a-ketoglutarate (aKG) metabolites and
inhibited by metabolites with similar structures, including 2-hydroxyglutarate (2HG), succinate, and fumurate.
It is unclear whether exercise alters the profile of these metabolites in cellular compartments that might affect
TET activity.
www.perspectivesinmedicine.org

et al. 2016), exposure to toxins (Tryndyak et al. each of histone 2A, 2B, 3, and 4 (Li et al.
2016), and exercise (Voisin et al. 2015). These 2007). The double-stranded DNA helix wraps
changes are induced by altered activity of the around this stable core, which provides struc-
DNMTs in concert with changes in active de- tural stability to DNA and has also been hypoth-
methylation of DNA, which involves sequential esized to protect DNA from damaging agents
modification of the methylated cytosine by hy- and stimuli (Ljungman and Hanawalt 1992).
droxylation, deamination, and/or oxidation The physical relationship between DNA and
(Bhutani et al. 2011). The base is then replaced surrounding histone proteins determines how
by DNA repair mechanisms. These processes are surrounding genes are expressed, with a closed
mediated by the ten-eleven translocation chromatin formation favoring transcriptional
(TET), AID (activation-induced cytosine de- repression while an open chromatin structure
aminase)/APOBEC (apolipoprotein B mRNA favors transcriptional activation (Fig. 2) (Li
editing enzyme component 1) and the base ex- et al. 2007). Functionally, this is linked to the
cision repair (BER) enzymes, respectively (Bhu- ability of the transcriptional machinery, includ-
tani et al. 2011). Dynamic changes to DNA ing the basal transcription factors and RNA
methylation patterns in response to a range of polymerase, to gain access to gene regulatory
physiological stimuli are thought to be integral regions to initiate transcription (Kouzarides
to both physiological adaptation and disease 2007). A closed or condensed chromatin struc-
pathology. ture is the result of strong electrostatic interac-
tions between the negatively charged DNA
backbone and positively charged histone pro-
Histone Modifications
teins. Alterations in histone protein charge
Histone proteins form the nucleosome core, through posttranslational modifications are of-
which comprises an octamer of two copies ten sufficient to disrupt the interaction with

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876 3
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

Exercise-induced Excluded acetylated nucleosome


substrate flux a
a a
a a
HAT + Acetyl-CoA

Transcriptional Transcription initiation complex


repression
Histone acetylation

Histone deacetylation
Transcriptional
activation
HDACs

AMPK Lactate
βOHB

Exercise-induced
energy demand

Figure 2. Histone acetylation, transcription, and potential interactions with exercise metabolism. In their
deacetylated state, catalyzed by histone deacetylase (HDAC) enzymes, histones retain a close interaction with
DNA, resulting a compacted chromatin structure that is associated with transcriptional repression. Increased
histone acetyltransferase (HAT) activity and acetyl-CoA availability increase histone acetylation, resulting in a
loose chromatin structure that can result in transient nucleosome exclusion and exposure of regulatory DNA
www.perspectivesinmedicine.org

regions to transcriptional activators such as the transcriptional initiation complex. Inhibition of HDAC activity
has similar effects. Exercise metabolism could regulate this system through multiple mechanisms, including by
influencing acetyl-CoA availability and through regulation of factors that inhibit HDAC activity. These include
increased AMP-activated protein kinase (AMPK) activity and increased concentrations of the metabolites
lactate and b-hydroxybutyrate (bOHB).

DNA, resulting in an open chromatin confirma- favors an open chromatin confirmation and
tion (Kouzarides 2007). transcriptional activation. Indeed, K9 and K14
Although not exhaustive, characterized acetylation on H3 is associated with transcrip-
posttranslational histone modifications on a tional activation (Fig. 2). Histone acetylation
number of amino acid sites include phosphor- state is determined by the balance between
ylation, acetylation, methylation, ubiquitina- histone acetyltransferase (HAT) and histone de-
tion, sumoylation, and ADP-ribosylation (Ban- acetylase (HDAC) activities (Saha and Pahan
nister and Kouzarides 2011). A wide range of 2006). There are numerous HATand HDAC iso-
regulatory enzymes, the activity of which serves forms, each with different substrate specificities,
as a control nexus that links cellular signaling signaling pathway responsiveness and gene reg-
pathways with epigenetic control of gene ex- ulatory functions. These epigenetic modifiers
pression, governs these posttranslational his- are often recruited to specific nucleosome sites
tone modifications (Feinberg et al. 2016). One through interactions with DNA-bound tran-
of the best-characterized histone modifications scription factors, with various HDAC and
is lysine acetylation, particularly to histones 3 HAT isoforms having different affinities for dif-
(H3) and 4 (H4). Lysine amino acid (K) side- ferent transcription factors (Kouzarides 2007).
chains carry a positive charge when unmodi- This diversity allows control of histone acetyla-
fied; however, acetylation neutralizes this posi- tion at specific loci, ensuring specificity in the
tive charge, thereby disrupting the electrostatic gene expression response. Histone acetylation
interaction with DNA (Ettig et al. 2011). This can work in concert with other histone modifi-

4 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise Epigenetics

cations to provide precise control over the tran- muscle, which was associated with increased
scriptional response (Bannister and Kouzarides transcription of these genes. Interestingly, the
2011). For example, acetylation of K14 on H3 same exercise modality at a lower intensity
appears dependent on prior phosphorylation of (40% VO2 max) had no effect on DNA methyl-
serine 10 (Lo et al. 2000). Furthermore, H3 acet- ation, suggesting that exercise intensity may be a
ylation is often coupled with H3 methylation at critical factor in influencing DNA methylation.
K36, which appears to control transcriptional A recent study in mice has replicated some of
elongation (Wagner and Carpenter 2012). these findings, showing that exercise reduced
Such interactions have coined the term “the his- PGC1a promoter methylation, which was asso-
tone code” to describe broader regulatory mod- ciated with increased basal transcription of this
ifications controlling specific gene expression gene (Lochmann et al. 2015). Another study has
patterns. However, adding further complexity found that 120 min of cycling in healthy sub-
to this notion is the fact that subtle variation jects (60% VO2 max) increased methylation at
in modifications to the same histone site can the promoters of FABP3 and COX4L1, which
confer opposing effects on gene transcription. was associated with decreased expression of
For example, methylation of H3 at K9 has the these genes (Lane et al. 2015).
opposite effect to acetylation, inducing tran- These studies show that alterations in DNA
scriptional repression through inhibition of methylation can occur more rapidly than previ-
the p300 HAT (Stewart et al. 2005). These stud- ously thought and clearly indicate that DNA
ies highlight the complexity in the “histone methylation may be a crucial component of
code” concept and how it might control con- the immediate physiological response to an
text-specific gene expression responses. For a acute bout of exercise. The mechanism of alter-
full review on details of other histone post- ations in DNA methylation patterns in skeletal
www.perspectivesinmedicine.org

translational modifications, their modifying en- muscle following a bout of exercise are un-
zymes, the “histone code” concept, and how known, but are likely to involve precise regula-
these modifications interact, readers are encour- tion of the DNMTs and the enzymes involved
aged to consult the following excellent reviews: in demethylation of DNA. Although little is
Bannister and Kouzarides (2011), Carone and known in this area, in vitro knockdown of
Rando (2012), and Rando (2012). DNMT3B was shown to prevent saturated fatty
The following sections will detail the emerg- acid-induced methylation of the promoter of
ing data describing the epigenetics of exer- PGC1a in a muscle cell line (Barres et al.
cise, which at present is principally described 2009), and TET1 was induced by hypoxia and
through alterations in DNA methylation and associated with the regulation of glycolytic gene
histone acetylation. expression in various cell lines (Tsai et al. 2014).
More research is required to understand the role
of DNA methylation and demethylation in the
SKELETAL MUSCLE DNA METHYLATION
adaptive response to acute exercise, but it ap-
AND REGULATING ENZYMES IN RESPONSE
pears clear that this system is likely to play a role.
TO EXERCISE
Of particular interest will be an understanding
There are a limited number of studies that have of whether such changes are retained over time,
examined the effects of exercise on DNA meth- and how rapidly they are lost before a further
ylation in skeletal muscle. The available data bout of exercise is required to have the same
suggest that acute exercise causes reduced meth- beneficial effect.
ylation in skeletal muscle in a gene-specific There is some evidence in the literature de-
manner. For example, Barres et al. (2012) scribing the effects of exercise training on DNA
showed that high-intensity exercise (80% VO2 methylation. Nitert et al. (2012) studied first-
max) caused an immediate reduction in the degree relatives of patients with T2D and found
methylation levels in the promoter regions of altered DNA methylation in skeletal muscle in
PGC1a, PDK4, and PPARd in human skeletal pathways, including MAPK, insulin, and calci-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876 5
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

um signaling, and in several genes known to sponse to 60 min of cycling in humans (McGee
have metabolic functions, including PRKAB1. et al. 2009). This study showed that H3 K36
After a 6-month exercise intervention in these acetylation was increased immediately after ex-
healthy but sedentary subjects, which consisted ercise, while there was no change in H3 K9/14
primarily of endurance exercise, decreased DNA acetylation. The physiological relevance of al-
methylation in skeletal muscle was observed for terations in global acetylation is not entirely
retinol metabolism and calcium signaling path- clear given that acetylation at specific gene
ways and a number of genes with known meta- loci are most closely associated with gene-spe-
bolic function such as MEF2A and NDUFC2 cific transcription. However, other studies have
(Nitert et al. 2012). The reduced DNA methyl- examined histone acetylation at specific gene
ation was associated with increased gene expres- loci in response to exercise. For example, re-
sion in many pathways, but particularly those peated bouts of swimming in rats increased
associated with mitochondrial function. Fur- H3 K9/14 acetylation at the GLUT4 promoter
thermore, Alibegovic et al. (2010) showed that in triceps muscle (Smith et al. 2008). Further
9 days of enforced bed rest tended to increase understanding of the role of chromatin remod-
DNA methylation in the promoter region of eling in the regulation of exercise adaptations
PPARGC1A, a key regulator of mitochondrial will require future studies to examine multiple
biogenesis and function, and that this was asso- histone modifications with approaches such as
ciated with down-regulation of many genes ChIP-seq. As these technologies are becoming
involved in mitochondrial function and partic- more accessible, so too are the opportunities
ularly oxidative phosphorylation. When the to decipher the molecular regulation of muscle
same subjects underwent 4 weeks of exer- adaptation.
cise retraining (supervised cycle ergometry at Histone modifications are controlled by a
www.perspectivesinmedicine.org

70% of VO2 max), DNA methylation in the diverse number of enzymes that are also regu-
PPARGC1A promoter tended to decrease (Ali- lated by various signaling mechanisms. These
begovic et al. 2010). include HATs and HDACs, histone methyltrans-
More research is required to understand ferases (HMTs) and demethylases (HDMs), and
the role of DNA methylation and demethyla- histone kinases and histone ligases (Bannister
tion in the adaptive response to exercise, but and Kouzarides 2011), to name a few. Many of
it appears clear that this system is likely to these chromatin remodeling enzymes exist as
play a prominent role. Of particular interest larger multipeptide transcriptional regulatory
will be an understanding of whether such complexes and control of their activity is regu-
changes are retained over time, and how rap- lated through complex cofactor exchanges, lo-
idly they are lost before a further bout of calization, and posttranslational modifications
exercise is required to have the same beneficial (Bannister and Kouzarides 2011). Just as his-
effect. tone acetylation has served as a starting point
for understanding how epigenetic modifica-
tions might influence skeletal muscle adapta-
SKELETAL MUSCLE HISTONE
tion to exercise, a number of studies have also
MODIFICATIONS AND REGULATING
examined the regulation of chromatin remod-
ENZYMES IN RESPONSE TO EXERCISE
eling enzymes that control histone acetylation
Just as histone acetylation is the most widely in response to exercise. For example, we have
studied histone modification in epigenetics, it previously found that 60 min of cycling exercise
is one of the few histone modifications that in humans reduces the nuclear abundance of
have been examined in the context of responses the class IIa HDACs in skeletal muscle (McGee
to exercise. We have previously examined skel- and Hargreaves 2004; McGee et al. 2009), which
etal muscle global H3 K9/14 and K36 acetyla- has been implicated in the regulation of muscle-
tion, which are associated with transcriptional specific genes via repression of the MEF2 tran-
initiation and elongation, respectively, in re- scription factor (McKinsey et al. 2000). This is

6 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise Epigenetics

consistent with phosphorylation-dependent ing mechanism involving multiple histone


nuclear export of the class IIa HDACs being a modifications to regulate transcriptional re-
major mechanism controlling class IIa HDAC sponses.
repressive activity. Once exported from the nu- Another link between exercise and epige-
cleus, MEF2 is derepressed, which activates netic regulation comes from the class III
MEF2-dependent transcription (McKinsey HDACs, termed sirtuins. Unlike the class I
et al. 2000). An interesting aspect of class IIa and II HDACs that are Zn2þ-dependent, the
HDAC biology is that they do not possess in- Sirtuin family are dependent on NADþ for
trinsic deacetylase activity themselves, but full activity, making this group of HDACs ex-
instead require a corepressor complex that in- quisitely sensitive to alterations in cellular me-
cludes N-Cor/SMRT and HDAC3 (Fischle tabolism and redox state (Chang and Guarente
et al. 2002). A reduction in HDAC activity and 2014). There are seven identified sirtuins, each
an increase in HAT activity has also been ob- with different subcellular localizations and sub-
served in the hippocampus of rats following a strate specificities (Houtkooper et al. 2012).
single session of treadmill exercise (Elsner et al. Once activated, the Sirtuins enhance a number
2011). of aspects of metabolism, including enhanced
Perhaps the most obvious link between ex- oxidative flux and capacity, by deacetylat-
ercise-regulated signal transduction and epige- ing both histone and nonhistone proteins
netic regulation comes from the kinases that are (Houtkooper et al. 2012). The effects of exercise
able to phosphorylate the class IIa HDACs, re- on various sirtuin isoforms are difficult to as-
sulting in class IIa HDAC nuclear export. We sess in activity assays, as allosteric activation by
found that the exercise-responsive AMP-acti- NADþ is rapidly lost. However, a number of
vated protein kinase (AMPK) is a class IIa sirtuin targets are deacetylated during exercise
www.perspectivesinmedicine.org

HDAC kinase. Phosphorylation of HDAC5 by and there is evidence that sirtuin activity during
AMPK was also shown to increase H3 K9/14 exercise is controlled by AMPK (Canto et al.
acetylation and transcription of the GLUT4 2009). Knockout of various sirtuin isoforms
gene (McGee et al. 2008). The importance of in mice has had limited effects on metabolic
this signaling nexus to exercise adaptations adaptations to exercise (Menzies et al. 2013),
was highlighted by our identification of redun- possibly indicating that further redundancy in
dancy and compensation in class IIa regulation this adaptive response exists. Whether sirtuin
in the absence of AMPK signaling (McGee et al. activation during exercise contributes to adap-
2014). Indeed, a number of other exercise-re- tive responses through direct histone deacetyla-
sponsive kinases are also class IIa HDAC ki- tion or through deacetylation of nonhistone
nases, such as the calcium/calmodulin depen- proteins such as PGC-1a or Foxo1 (Canto
dent protein kinase II (CaMKII) (Rose and et al. 2009) also remains to be established. Al-
Hargreaves 2003; Backs et al. 2006). Through though no studies to our knowledge have
the use of the KN93 inhibitor, CaMKII has examined whether exercise modulates other
been implicated in the acetylation of H3 at the chromatin remodeling enzymes that control
GLUT4 promoter during swimming exercise in methylation, ubiquitination, etc., it would
rats (Smith et al. 2008). Another interesting link seem likely that there is a coordinated response
between these exercise-responsive kinases and to control exercise adaptions by regulating a
regulation of histone modifications is that number of chromatin remodeling enzymes. In-
many of these same kinases are also H3 kinases deed, just as the field of epigenetics is starting to
(McGee and Hargreaves 2008; Bungard et al. unravel aspects of this “histone code” and how
2010). This is particularly intriguing given it regulates specific transcriptional responses,
that phosphorylation of H3 at S10 is required the field of exercise physiology will also have
before acetylation of K9 and 14 can occur in a to design appropriate studies to unravel this
number of different cellular contexts (Cheung code and its potential contribution to skeletal
et al. 2000). This suggests a coordinated signal- muscle adaptations to exercise.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876 7
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

EXERCISE-MEDIATED EPIGENETIC et al. 2016; King-Himmelreich et al. 2016),


ALTERATIONS IN OTHER TISSUES further supporting the idea that circulating fac-
tors might be important. Interestingly, the ge-
Transcriptional adaptive responses to exercise nomic loci and genes regulated in blood cells by
occur in a number of tissues in addition to skel- these different exercise modalities are divergent.
etal muscle. For example, global gene expression This suggests that specific systemic signaling
alterations immediately following treadmill mechanisms are used to ensure an appropriate
running in mice are more profound in the liver physiological response to the particular exercise
than in skeletal muscle (Hoene et al. 2010). modality.
Therefore, epigenetic mechanisms must also
be engaged in nonmuscle tissues during exer-
cise. Intriguingly, a number of studies have ex- SKELETAL MUSCLE METABOLISM AND
EPIGENETIC INTERACTIONS AND THEIR
amined histone modifications in various brain
POTENTIAL ROLE IN EXERCISE ADAPTIVE
regions in response to exercise. Forced swim-
RESPONSES
ming induces H3 K14 acetylation and serine
10 phosphorylation in spatially distinct regions Although epigenetic mechanisms regulate the
of the dentate gyrus in a time dependent man- expression of metabolic gene networks in re-
ner (Chandramohan et al. 2008). In addition, sponse to exercise, there is emerging evidence
voluntary exercise in rats increases H3 acetyla- that metabolism can influence epigenetic pro-
tion at the brain-derived neurotrophic factor cesses by controlling the availability of sub-
(BDNF) promoter in the hippocampus (Go- strates used for epigenetic modifications and/
mez-Pinilla et al. 2011). These studies were or the activity of epigenetic modifiers. The ma-
among the first to associate changes in histone jor substrate for DNA methylation is S-adeno-
www.perspectivesinmedicine.org

modifications with alterations in exercise-in- sylmethionine (SAM), which is produced in the


duced gene expression. However, whether these methionine-recycling pathway (Sauter et al.
modifications are a result of exercise per se or a 2013). Changes in the cellular levels of SAM
psychological stress response to exercise remains result in altered levels of DNA methylation,
unclear. There is a dearth of information on how with concomitant effects on gene transcription
epigenetic mechanisms might regulate gene ex- (Ables et al. 2016). Ultimately, the methyl
pression programs in noncontracting tissues groups required for DNA methylation are de-
during exercise, although local alterations in en- rived from a range of dietary sources, and alter-
ergy balance, blood flow, and hypoxia are all ing the availability of dietary methyl donors
candidate stimuli. Evidence is emerging that cir- such as folate and selenium has been shown to
culating factors could be involved in systemic affect DNA methylation and gene expression,
regulation of epigenetic processes during exer- including improving diseases such as neural
cise. For example, exercise-conditioned plasma tube defects associated with DNA hypomethy-
reduced the nuclear abundance of DNMT3B in lation (Chango and Pogribny 2015). Although
isolated peripheral blood mononuclear cells there are numerous studies that have investigat-
(Horsburgh et al. 2015). Although the exact cir- ed the impact of increasing or decreasing die-
culating factors involved remain to be deter- tary methionine, which has impacts including
mined, a number of bona fide myokines that extending life span in a number of species fol-
are released from contracting muscle during lowing dietary methionine restriction (Ables
exercise have been identified (Whitham and et al. 2016), there is no information regarding
Febbraio 2016). How such circulating factors whether any of the phenotypic differences seen
might regulate epigenetic processes in response may be mediated by altered levels of DNA meth-
to exercise are only just emerging. However, ylation. Similarly, the effects of exercise on the
other studies have noted altered DNA methyla- methionine-recycling pathway are currently un-
tion in response to both endurance and resis- known. Although ATP is required for the con-
tance exercise in various blood cells (Denham version of SAM to methionine (Sauter et al.

8 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise Epigenetics

2013), it is difficult to conceive that the ATP In addition to substrate availability directly
demand during exercise could acutely compro- controlling epigenetic modifications, a number
mise methionine availability and methylation of metabolites are known to regulate the activity
reactions. In contrast, the activity of the de- of histone modifiers. Indeed, lactate has been
methylating TET enzymes is increased by the observed to inhibit HDACs in the context of
tricarboxylic acid (TCA) cycle intermediate me- cancer (Latham et al. 2012). It is unknown
tabolite a-ketoglutarate, whereas accumulation whether lactate accumulation during intense
of metabolites with similar structural features, exercise contributes to the increased local his-
such as succinate, fumurate, and 2-hydroxyglu- tone acetylation that has been observed during
tarate inhibit TETactivity (Figueroa et al. 2010). endurance-based exercise. Given that lactate
Although the effect of exercise on the abun- concentrations of 10 – 50 mM are required to
dance of these metabolites is unclear, particu- reduce total HDAC activity 50%– 80%
larly in cellular compartments relevant to TET (Latham et al. 2012), it appears unlikely that
regulation, this could suggest that metabolite lactate is a key regulator of HDAC activity dur-
fluxes are critical for coordinating skeletal mus- ing predominantly aerobic endurance exercise.
cle DNA-methylation responses to exercise. However, during intense supramaximal exer-
The availability of the two-carbon metabo- cise, muscle lactate concentrations can reach
lite acetyl-CoA can also be rate limiting for acet- 60 mM (Jacobs et al. 1983). This suggests
ylation reactions (Galdieri and Vancura 2012). that lactate production during intense exercise
Acetyl-CoA is principally derived from pyruvate could acutely reduce HDAC activity and con-
oxidation and through b-oxidation of fatty ac- tribute to epigenetic regulation in addition to
ids, both of which occur in the mitochondria. canonical signaling pathways that control epi-
However, acetyl-CoA is unable to diffuse across genetic modifiers, which are activated during
www.perspectivesinmedicine.org

phospholipid membranes and mitochondrial supramaximal exercise (Gibala et al. 2009). Ke-
citrate is exported from the TCA cycle via the tone bodies such as bOHB can also inhibit
citrate transporter to the cytosol and the nucle- HDAC activity (Shimazu et al. 2013). Ketone
us. Nucleocytosolic citrate can then be convert- bodies are principally generated in the liver in
ed to back to acetyl-CoA by ATP-citrate lyase response to elevated rates of b-oxidation and
(ACL) and used in acetylation reactions of his- plasma bOHB increases during prolonged en-
tone and nonhistone proteins (Choudhary et al. durance exercise (Bordin et al. 1992). Circulat-
2014). This pathway appears to be particularly ing bOHB is taken up by a number of tissues,
important for linking metabolic flux with met- particularly the brain, where it can be converted
abolic gene expression. Evidence for this comes back to acetyl-CoA for oxidation. Whether in-
from experiments in which increases in the ex- creased systemic bOHB contributes to exercise-
pression of genes such as GLUT4, HK2, PFK-1, mediated transcriptional regulation in non-
and LDH in response to increasing glucose con- muscle tissues, through direct regulation of
centrations are abrogated with ACL knockdown HDACs or through generation of acetyl-CoA
(Wellen et al. 2009). At present, it is unknown for acetylation reactions, remains to be deter-
how the energetic demands of exercise might mined. However, altered gene expression is a
influence histone acetylation secondary to in- well-characterized response to exercise in non-
creased TCA cycle flux. Given that exercise contracting tissues (Hoene and Weigert 2010;
increases skeletal muscle acetyl-CoA levels via Catoire et al. 2012) and is thought to contribute
increased substrate catabolism (Constantin- to the many whole body positive health effects
Teodosiu et al. 1991), it is tempting to speculate of exercise.
that an increased acetyl-CoA pool would con- Collectively, these studies indicate that al-
tribute to increased skeletal muscle histone tered metabolism during exercise could have a
acetylation in concert with altered activity/ profound impact on the epigenome (Figs. 1 and
function of HDACs and HATs to enhance the 2). Future studies will be required to mechanis-
transcription of specific genes. tically determine whether this is the case.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876 9
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

EPIGENETIC THERAPIES TO INDUCE cerns about off-target effects, the increase in his-
EXERCISE ADAPTATIONS tone acetylation in other tissues during exercise,
such as in the brain (Chandramohan et al. 2008;
As the epigenetics of exercise becomes better Gomez-Pinilla et al. 2011), indicates that some
understood, opportunities are presented to ex- of these effects might not be considered off-tar-
perimentally manipulate aspects of the exercise get. Furthermore, many positive benefits might
adaptive response. Clearly, this has many poten- be derived from systemic HDAC inhibition. For
tial therapeutic implications for managing example, HDAC inhibitors have been used to
chronic diseases underpinned by inactivity, prevent pathological cardiac hypertrophy
which include obesity, type 2 diabetes, and car- (Kong et al. 2006), psychiatric diseases (Meylan
diovascular and liver diseases. et al. 2016), and cancer (Falkenberg and John-
Following from our observations that the stone 2014) in preclinical and clinical studies.
repressive actions of the class IIa HDACs are re- Indeed, the wide-ranging positive benefits of
duced by exercise (McGee and Hargreaves 2004; HDAC inhibition are consistent with these en-
McGee et al. 2009), as signified by disruption of zymes as important regulators of the exercise
the class II HDAC corepressor complex and their adaptive response in a number of tissues.
nuclear export, we have recently examined These studies highlight the value in under-
whether manipulating class IIa HDAC activity standing the epigenetic processes regulating
could induce exercise adaptations in skeletal skeletal muscle adaptations to exercise. As the
muscle. Genetic manipulation of HDAC4 and field continues to progress and defines the roles
5, so that they could not form a corepressor com- of other epigenetic modifiers such as HATs,
plex, increased MEF2 transcriptional activity, HMTs, and HMDs and ubiquitin ligases in the
the expression of a program of exercise-respon- exercise adaptive response, there will be more
www.perspectivesinmedicine.org

sive metabolic genes and fatty acid oxidation opportunities for therapeutic intervention in
(Gaur et al. 2016). We next identified an existing chronic diseases driven by inactivity. Given the
HDAC inhibitor, termed scriptaid, which was huge burden imparted by such diseases on
also able to disrupt the class IIa HDAC corepres- modern societies, such research should be a pri-
sor complex. Acute administration of scriptaid ority for the field.
to mice increased the expression of exercise-
responsive metabolic genes in skeletal muscle,
CONCLUDING REMARKS
whereas chronic administration increased exer-
cise performance, whole-body energy ex- Epigenetic regulation of gene expression is a
penditure, and lipid oxidation and reduced major control point for gene expression re-
plasma glucose and lipids (Gaur et al. 2016). A sponses across all organisms in response to en-
number of other studies using various HDAC vironmental stimuli. Methylation of DNA and
inhibitors have reported similar enhancements posttranslational modification of histones are
in various aspects of muscle and whole-body the major epigenetic control mechanisms that
metabolism, including insulin sensitivity (Tan have been characterized to date under experi-
et al. 2015) and enhanced oxidative capacity mental conditions. The interaction between
(Galmozzi et al. 2013). The exact mechanism DNA methylation and the various histone mod-
of action of some of these broad-spectrum ifications at specific gene loci is only just begin-
HDAC inhibitors remains to be determined, ning to be understood; however, it is expected
but could include inhibition of HDAC3, the that these complex spatially and temporally de-
class I HDAC that provides deacetylase activity pendent interactions will become clearer as the
to the class IIa HDAC corepressor complex technology to probe these interactions becomes
(Fischle et al. 2002; Hudson et al. 2015). System- more accessible.
ic administration of these inhibitors could have At present, very little is known about the
compound-specific effects in tissues other than epigenetics that contribute to transcriptional
skeletal muscle. Although this could raise con- responses and phenotypic adaptations to exer-

10 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise Epigenetics

cise. Early studies have established roles for Alibegovic AC, Sonne MP, Hojbjerre L, Bork-Jensen J, Ja-
cobsen S, Nilsson E, Faerch K, Hiscock N, Mortensen B,
acute reductions in DNA methylation and in- Friedrichsen M, et al. 2010. Insulin resistance induced by
creases in histone acetylation as contributors to physical inactivity is associated with multiple transcrip-
this response. Additional studies also implicate tional changes in skeletal muscle in young men. Am J
regulation of the class IIa HDACs as being im- Physiol Endocrinol Metab 299: E752– E763.
Amaral CL, Crisma AR, Masi LN, Martins AR, Hirabara SM,
portant in exercise adaptations. Another emerg- Curi R. 2014. DNA methylation changes induced by a
ing area of research will be how alterations in high-fat diet and fish oil supplementation in the skeletal
muscle metabolism impact on epigenetics and muscle of mice. J Nutrigenet Nutrigenomics 7: 314 –326.
exercise adaptations, given that a number of Backs J, Song K, Bezprozvannaya S, Chang S, Olson EN.
2006. CaM kinase II selectively signals to histone deace-
metabolites are known to be rate limiting for tylase 4 during cardiomyocyte hypertrophy. J Clin Invest
epigenetic modification reactions or directly 116: 1853– 1864.
regulate the activity of epigenetic modifiers. Fi- Bannister AJ, Kouzarides T. 2011. Regulation of chromatin
nally, an exciting area for future research will be by histone modifications. Cell Res 21: 381 –395.
whether our understanding of the epigenetics of Barres R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, Krook
A, Zierath JR. 2009. Non-CpG methylation of the PGC-
exercise can be leveraged to provide therapeutic 1a promoter through DNMT3B controls mitochondrial
targets and ultimately therapies for diseases un- density. Cell Metab 10: 189– 198.
derpinned by inactivity, such as obesity, type 2 Barres R, Yan J, Egan B, Treebak JT, Rasmussen M, Fritz T,
diabetes, and cardiovascular diseases. Future Caidahl K, Krook A, O’Gorman DJ, Zierath JR. 2012.
Acute exercise remodels promoter methylation in human
studies should systematically examine the ef- skeletal muscle. Cell Metab 15: 405 –411.
fects of exercise modality, intensity, and dura- Bedi U, Mishra VK, Wasilewski D, Scheel C, Johnsen SA.
tion when examining interactions between 2014. Epigenetic plasticity: A central regulator of epithe-
lial-to-mesenchymal transition in cancer. Oncotarget 5:
epigenetic mechanisms to provide an exercise- 2016–2029.
epigenetic roadmap. Incorporating new omics
www.perspectivesinmedicine.org

Bhutani N, Burns DM, Blau HM. 2011. DNA demethylation


technologies, such as unbiased proteomics en- dynamics. Cell 146: 866–872.
compassing posttranslational modifications, to Bordin D, Bottecchia D, Bettini V, Aragno R, Sartorelli L.
existing epigenetic pipelines will make this a 1992. Effect of middle-intensity exercise on carnitine and
b-hydroxybutyrate plasmatic concentration in men and
realistic prospect that could have wide-ranging women. J Sports Med Phys Fitness 32: 394–399.
implications for understanding the health ben- Bungard D, Fuerth BJ, Zeng PY, Faubert B, Maas NL, Viollet
efits of exercise that could be harnessed for B, Carling D, Thompson CB, Jones RG, Berger SL. 2010.
medicine. Signaling kinase AMPK activates stress-promoted tran-
scription via histone H2B phosphorylation. Science 329:
1201–1205.
Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L,
ACKNOWLEDGMENTS Milne JC, Elliott PJ, Puigserver P, Auwerx J. 2009. AMPK
regulates energy expenditure by modulating NADþ me-
The authors thank Prof. Mark Hargreaves (The tabolism and SIRT1 activity. Nature 458: 1056–1060.
University of Melbourne) and members of their Carone BR, Rando OJ. 2012. Rewriting the epigenome. Cell
laboratories for ongoing discussions on the top- 149: 1422– 1423.
ic of this review. The authors are supported by Catoire M, Mensink M, Boekschoten MV, Hangelbroek R,
Muller M, Schrauwen P, Kersten S. 2012. Pronounced
grants from the National Health and Medical effects of acute endurance exercise on gene expression
Research Council (NHMRC), the Diabetes Aus- in resting and exercising human skeletal muscle. PLoS
tralia Research Program (DARP), the National ONE 7: e51066.
Breast Cancer Foundation (NBCF), and the Chandramohan Y, Droste SK, Arthur JS, Reul JM. 2008. The
forced swimming-induced behavioural immobility re-
Centre for Molecular and Medical Research sponse involves histone H3 phospho-acetylation and
(C-MMR) at Deakin University. c-Fos induction in dentate gyrus granule neurons via
activation of the N-methyl-D-aspartate/extracellular
signal-regulated kinase/mitogen- and stress-activated
kinase signalling pathway. Eur J Neurosci 27: 2701–2713.
REFERENCES
Chang HC, Guarente L. 2014. SIRT1 and other sirtuins in
Ables GP, Hens JR, Nichenametla SN. 2016. Methionine metabolism. Trends Endocrinol Metab 25: 138–145.
restriction beyond life-span extension. Ann NY Acad Sci Chang H, Zhang T, Zhang Z, Bao R, Fu C, Wang Z, Bao Y, Li
1363: 68–79. Y, Wu L, Zheng X, et al. 2011. Tissue-specific distribution

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876 11
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

of aberrant DNA methylation associated with maternal Galmozzi A, Mitro N, Ferrari A, Gers E, Gilardi F, Godio C,
low-folate status in human neural tube defects. J Nutr Cermenati G, Gualerzi A, Donetti E, Rotili D, et al. 2013.
Biochem 22: 1172–1177. Inhibition of class I histone deacetylases unveils a mito-
Chango A, Pogribny IP. 2015. Considering maternal dietary chondrial signature and enhances oxidative metabolism
modulators for epigenetic regulation and programming in skeletal muscle and adipose tissue. Diabetes 62: 732–
of the fetal epigenome. Nutrients 7: 2748– 2770. 742.
Cheung P, Tanner KG, Cheung WL, Sassone-Corsi P, Denu Gaur V, Connor T, Sanigorski A, Martin SD, Bruce CR,
JM, Allis CD. 2000. Synergistic coupling of histone H3 Henstridge DC, Bond ST, McEwen KA, Kerr-Bayles L,
phosphorylation and acetylation in response to epider- Ashton TD, et al. 2016. Disruption of the class IIa
mal growth factor stimulation. Mol Cell 5: 905– 915. HDAC corepressor complex increases energy expenditure
Choudhary C, Weinert BT, Nishida Y, Verdin E, Mann M. and lipid oxidation. Cell Rep 16: 2802– 2810.
2014. The growing landscape of lysine acetylation links Gavin DP, Sharma RP. 2010. Histone modifications, DNA
metabolism and cell signalling. Nat Rev Mol Cell Biol 15: methylation, and schizophrenia. Neurosci Biobehav Rev
536–550. 34: 882– 888.
Clausen JP, Trap-Jensen J. 1970. Effects of training on the Gibala MJ, McGee SL, Garnham AP, Howlett KF, Snow RJ,
distribution of cardiac output in patients with coronary Hargreaves M. 2009. Brief intense interval exercise acti-
artery disease. Circulation 42: 611– 624. vates AMPK and p38 MAPK signaling and increases the
Constantin-Teodosiu D, Carlin JI, Cederblad G, Harris RC, expression of PGC-1a in human skeletal muscle. J Appl
Hultman E. 1991. Acetyl group accumulation and pyru- Physiol 106: 929–934.
vate dehydrogenase activity in human muscle during in- Glatz JF, Bonen A, Luiken JJ. 2002. Exercise and insulin
cremental exercise. Acta Physiol Scand 143: 367–372. increase muscle fatty acid uptake by recruiting putative
Denham J, Marques FZ, Bruns EL, O’Brien BJ, Charchar FJ. fatty acid transporters to the sarcolemma. Curr Opin Clin
2016. Epigenetic changes in leukocytes after 8 weeks of Nutr Metab Care 5: 365–370.
resistance exercise training. Eur J Appl Physiol 116: 1245– Gomez-Pinilla F, Zhuang Y, Feng J, Ying Z, Fan G. 2011.
1253. Exercise impacts brain-derived neurotrophic factor plas-
Denis H, Ndlovu MN, Fuks F. 2011. Regulation of mamma- ticity by engaging mechanisms of epigenetic regulation.
lian DNA methyltransferases: A route to new mecha- Eur J Neurosci 33: 383– 390.
nisms. EMBO Rep 12: 647 –656. Hamidi T, Singh AK, Chen T. 2015. Genetic alterations of
www.perspectivesinmedicine.org

Egan B, Zierath JR. 2013. Exercise metabolism and the mo- DNA methylation machinery in human diseases. Epige-
lecular regulation of skeletal muscle adaptation. Cell nomics 7: 247–265.
Metab 17: 162–184. Henikoff S, Greally JM. 2016. Epigenetics, cellular memory
Elsner VR, Lovatel GA, Bertoldi K, Vanzella C, Santos FM, and gene regulation. Curr Biol 26: R644– R648.
Spindler C, de Almeida EF, Nardin P, Siqueira IR. 2011. Hoene M, Weigert C. 2010. The stress response of the liver to
Effect of different exercise protocols on histone acetyl- physical exercise. Exerc Immunol Rev 16: 163– 183.
transferases and histone deacetylases activities in rat hip- Hoene M, Franken H, Fritsche L, Lehmann R, Pohl AK,
pocampus. Neuroscience 192: 580–587. Haring HU, Zell A, Schleicher ED, Weigert C. 2010. Ac-
Ettig R, Kepper N, Stehr R, Wedemann G, Rippe K. 2011. tivation of the mitogen-activated protein kinase (MAPK)
Dissecting DNA-histone interactions in the nucleosome signalling pathway in the liver of mice is related to plasma
by molecular dynamics simulations of DNA unwrapping. glucose levels after acute exercise. Diabetologia 53: 1131–
Biophys J 101: 1999– 2008. 1141.
Falkenberg KJ, Johnstone RW. 2014. Histone deacetylases Holloszy JO. 1967. Biochemical adaptations in muscle. Ef-
and their inhibitors in cancer, neurological diseases and fects of exercise on mitochondrial oxygen uptake and
immune disorders. Nat Rev Drug Discov 13: 673– 691. respiratory enzyme activity in skeletal muscle. J Biol
Feinberg AP, Koldobskiy MA, Gondor A. 2016. Epigenetic Chem 242: 2278– 2282.
modulators, modifiers and mediators in cancer aetiology Horsburgh S, Todryk S, Toms C, Moran CN, Ansley L. 2015.
and progression. Nat Rev Genet 17: 284– 299. Exercise-conditioned plasma attenuates nuclear concen-
Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih trations of DNA methyltransferase 3B in human periph-
A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, et eral blood mononuclear cells. Physiol Rep 3: e12621.
al. 2010. Leukemic IDH1 and IDH2 mutations result in a Houtkooper RH, Pirinen E, Auwerx J. 2012. Sirtuins as
hypermethylation phenotype, disrupt TET2 function, regulators of metabolism and healthspan. Nat Rev Mol
and impair hematopoietic differentiation. Cancer Cell Cell Biol 13: 225– 238.
18: 553 –567. Hudson GM, Watson PJ, Fairall L, Jamieson AG, Schwabe
Fischle W, Dequiedt F, Hendzel MJ, Guenther MG, Lazar JW. 2015. Insights into the recruitment of class IIa histone
MA, Voelter W, Verdin E. 2002. Enzymatic activity asso- deacetylases (HDACs) to the SMRT/NCoR transcrip-
ciated with class II HDACs is dependent on a multipro- tional repression complex. J Biol Chem 290: 18237–
tein complex containing HDAC3 and SMRT/N-CoR. 18244.
Mol Cell 9: 45– 57. Jacobs I, Tesch PA, Bar-Or O, Karlsson J, Dotan R. 1983.
Galdieri L, Vancura A. 2012. Acetyl-CoA carboxylase regu- Lactate in human skeletal muscle after 10 and 30 s of
lates global histone acetylation. J Biol Chem 287: 23865– supramaximal exercise. J Appl Physiol Respir Environ Ex-
23876. erc Physiol 55: 365 –367.

12 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise Epigenetics

Jiang YH, Sahoo T, Michaelis RC, Bercovich D, Bressler J, McGee SL, Swinton C, Morrison S, Gaur V, Campbell DE,
Kashork CD, Liu Q, Shaffer LG, Schroer RJ, Stockton Jorgensen SB, Kemp BE, Baar K, Steinberg GR, Har-
DW, et al. 2004. A mixed epigenetic/genetic model for greaves M. 2014. Compensatory regulation of HDAC5
oligogenic inheritance of autism with a limited role for in muscle maintains metabolic adaptive responses and
UBE3A. Am J Med Genet A 131: 1 –10. metabolism in response to energetic stress. FASEB J 8:
King-Himmelreich TS, Schramm S, Wolters MC, Schmetzer 3384–3395.
J, Moser CV, Knothe C, Resch E, Peil J, Geisslinger G, McKinsey TA, Zhang CL, Lu J, Olson EN. 2000. Signal-de-
Niederberger E. 2016. The impact of endurance exercise pendent nuclear export of a histone deacetylase regulates
on global and AMPK gene-specific DNA methylation. muscle differentiation. Nature 408: 106– 111.
Biochem Biophys Res Commun 474: 284– 290. Menzies KJ, Singh K, Saleem A, Hood DA. 2013. Sirtuin 1-
Klose RJ, Bird AP. 2006. Genomic DNA methylation: The mediated effects of exercise and resveratrol on mitochon-
mark and its mediators. Trends Biochem Sci 31: 89–97. drial biogenesis. J Biol Chem 288: 6968–6979.
Kong Y, Tannous P, Lu G, Berenji K, Rothermel BA, Olson Meylan EM, Halfon O, Magistretti PJ, Cardinaux JR. 2016.
EN, Hill JA. 2006. Suppression of class I and II histone The HDAC inhibitor SAHA improves depressive-like be-
deacetylases blunts pressure-overload cardiac hypertro- havior of CRTC1-deficient mice: Possible relevance for
phy. Circulation 113: 2579– 2588. treatment-resistant depression. Neuropharmacology 107:
111 –121.
Kouzarides T. 2007. Chromatin modifications and their
function. Cell 128: 693–705. Nader GA, von Walden F, Liu C, Lindvall J, Gutmann L,
Pistilli EE, Gordon PM. 2014. Resistance exercise training
Lane SC, Camera DM, Lassiter DG, Areta JL, Bird SR, Yeo modulates acute gene expression during human skeletal
WK, Jeacocke NA, Krook A, Zierath JR, Burke LM, et al. muscle hypertrophy. J Appl Physiol 116: 693 –702.
2015. Effects of sleeping with reduced carbohydrate avail-
ability on acute training responses. J Appl Physiol 119: Nagarajan RP, Hogart AR, Gwye Y, Martin MR, LaSalle JM.
2006. Reduced MeCP2 expression is frequent in autism
643–655.
frontal cortex and correlates with aberrant MECP2 pro-
Latham T, Mackay L, Sproul D, Karim M, Culley J, Harrison moter methylation. Epigenetics 1: e1– e11.
DJ, Hayward L, Langridge-Smith P, Gilbert N, Ramsa-
Nakao M. 2001. Epigenetics: Interaction of DNA methyla-
hoye BH. 2012. Lactate, a product of glycolytic metabo-
tion and chromatin. Gene 278: 25– 31.
lism, inhibits histone deacetylase activity and promotes
changes in gene expression. Nucleic Acids Res 40: 4794– Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E,
www.perspectivesinmedicine.org

4803. Yang BT, Lang S, Parikh H, Wessman Y, et al. 2012. Impact


of an exercise intervention on DNA methylation in skel-
Li B, Carey M, Workman JL. 2007. The role of chromatin etal muscle from first-degree relatives of patients with
during transcription. Cell 128: 707– 719. type 2 diabetes. Diabetes 61: 3322– 3332.
Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Rando OJ. 2012. Combinatorial complexity in chromatin
Tonti-Filippini J, Nery JR, Lee L, Ye Z, Ngo QM, et al. structure and function: Revisiting the histone code.
2009. Human DNA methylomes at base resolution show Curr Opin Genet Dev 22: 148– 155.
widespread epigenomic differences. Nature 462: 315 –
Robertson KD. 2005. DNA methylation and human disease.
322.
Nat Rev Genet 6: 597 –610.
Ljungman M, Hanawalt PC. 1992. Efficient protection
Rose AJ, Hargreaves M. 2003. Exercise increases Ca2þ-cal-
against oxidative DNA damage in chromatin. Mol Carci-
modulin-dependent protein kinase II activity in human
nog 5: 264– 269. skeletal muscle. J Physiol 553: 303 –309.
Lo WS, Trievel RC, Rojas JR, Duggan L, Hsu JY, Allis CD, Saha RN, Pahan K. 2006. HATs and HDACs in neurodegen-
Marmorstein R, Berger SL. 2000. Phosphorylation of ser- eration: A tale of disconcerted acetylation homeostasis.
ine 10 in histone H3 is functionally linked in vitro and in Cell Death Differ 13: 539– 550.
vivo to Gcn5-mediated acetylation at lysine 14. Mol Cell
Sauter M, Moffatt B, Saechao MC, Hell R, Wirtz M. 2013.
5: 917 –926.
Methionine salvage and S-adenosylmethionine: Essential
Lochmann TL, Thomas RR, Bennett JP Jr., Taylor SM. 2015. links between sulfur, ethylene and polyamine biosynthe-
Epigenetic modifications of the PGC-1a promoter dur- sis. Biochem J 451: 145– 154.
ing exercise induced expression in mice. PLoS ONE 10:
Shen H, Laird PW. 2013. Interplay between the cancer ge-
e0129647.
nome and epigenome. Cell 153: 38–55.
McGee SL, Hargreaves M. 2004. Exercise and myocyte en-
Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le
hancer factor 2 regulation in human skeletal muscle. Di- Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD,
abetes 53: 1208– 1214. et al. 2013. Suppression of oxidative stress by b-hydroxy-
McGee SL, Hargreaves M. 2008. AMPK and transcriptional butyrate, an endogenous histone deacetylase inhibitor.
regulation. Front Biosci 13: 3022–3033. Science 339: 211–214.
McGee SL, van Denderen BJ, Howlett KF, Mollica J, Schert- Silva-Martinez GA, Rodriguez-Rios D, Alvarado-Caudillo
zer JD, Kemp BE, Hargreaves M. 2008. AMP-activated Y, Vaquero A, Esteller M, Carmona FJ, Moran S, Nielsen
protein kinase regulates GLUT4 transcription by phos- FC, Wickstrom-Lindholm M, Wrobel K, et al. 2016. Ar-
phorylating histone deacetylase 5. Diabetes 57: 860 –867. achidonic and oleic acid exert distinct effects on the DNA
McGee SL, Fairlie E, Garnham AP, Hargreaves M. 2009. methylome. Epigenetics 11: 321– 334.
Exercise-induced histone modifications in human skele- Smith JA, Kohn TA, Chetty AK, Ojuka EO. 2008. CaMK
tal muscle. J Physiol 587: 5951–5958. activation during exercise is required for histone hyper-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876 13
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

S.L. McGee and K.R. Walder

acetylation and MEF2A binding at the MEF2 site on the hypoxia-induced epithelial-mesenchymal transition by
Glut4 gene. Am J Physiol Endocrinol Metab 295: E698– acting as a co-activator. Genome Biol 15: 513.
E704. Voisin S, Eynon N, Yan X, Bishop DJ. 2015. Exercise training
Stewart MD, Li J, Wong J. 2005. Relationship between his- and DNA methylation in humans. Acta Physiol (Oxf )
tone H3 lysine 9 methylation, transcription repression, 213: 39–59.
and heterochromatin protein 1 recruitment. Mol Cell Biol
Wagner EJ, Carpenter PB. 2012. Understanding the lan-
25: 2525–2538.
guage of Lys36 methylation at histone H3. Nat Rev Mol
Suzuki MM, Bird A. 2008. DNA methylation landscapes: Cell Biol 13: 115– 126.
Provocative insights from epigenomics. Nat Rev Genet
9: 465 –476. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross
JR, Thompson CB. 2009. ATP-citrate lyase links cellular
Tan HW, Sim AY, Huang SL, Leng Y, Long YC. 2015. HC
metabolism to histone acetylation. Science 324: 1076–
toxin (a HDAC inhibitor) enhances IRS1-Akt signalling
and metabolism in mouse myotubes. J Mol Endocrinol 55: 1080.
197–207. Whitham M, Febbraio MA. 2016. The ever-expanding my-
Tryndyak V, de Conti A, Doerge DR, Olson GR, Beland FA, okinome: Discovery challenges and therapeutic implica-
Pogribny IP. 2016. Furan-induced transcriptomic and tions. Nat Rev Drug Discov 15: 719–729.
gene-specific DNA methylation changes in the livers of Ziller MJ, Gu H, Muller F, Donaghey J, Tsai LT, Kohl-
Fischer 344 rats in a 2-year carcinogenicity study. Arch bacher O, De Jager PL, Rosen ED, Bennett DA, Bern-
Toxicol doi: 10.1007/s00204-016-1786-8. stein BE, et al. 2013. Charting a dynamic DNA meth-
Tsai YP, Chen HF, Chen SY, Cheng WC, Wang HW, Shen ZJ, ylation landscape of the human genome. Nature 500:
Song C, Teng SC, He C, Wu KJ. 2014. TET1 regulates 477 –481.
www.perspectivesinmedicine.org

14 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a029876
Downloaded from http://perspectivesinmedicine.cshlp.org/ at MOUNT SINAI SCH OFMED on March 21, 2017 - Published by Cold
Spring Harbor Laboratory Press

Exercise and the Skeletal Muscle Epigenome


Sean L. McGee and Ken R. Walder

Cold Spring Harb Perspect Med published online March 20, 2017

Subject Collection The Biology of Exercise

Exercise and the Skeletal Muscle Epigenome Autophagy-Dependent Beneficial Effects of


Sean L. McGee and Ken R. Walder Exercise
Jens Frey Halling and Henriette Pilegaard
Role of Nuclear Receptors in Exercise-Induced
Muscle Adaptations
Barbara Kupr, Svenia Schnyder and Christoph
Handschin

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved

Vous aimerez peut-être aussi