Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

J Physiol 599.

3 (2021) pp 803–817 803

SYMPOSIUM REVIEW

Exercise and mitochondrial health


Jonathan M. Memme1,2 , Avigail T. Erlich1,2 , Geetika Phukan1,2 and David A. Hood1,2
1
Muscle Health Research Centre, York University, Toronto, Ontario, Canada, M3J 1P3
2
School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada, M3J 1P3

Edited by: Scott Powers & Karyn Hamilton


The Journal of Physiology

Abstract Mitochondrial health is an important mediator of cellular function across a range of


tissues, and as a result contributes to whole-body vitality in health and disease. Our under-
standing of the regulation and function of these organelles is of great interest to scientists
and clinicians across many disciplines within our healthcare system. Skeletal muscle is a useful
model tissue for the study of mitochondrial adaptations because of its mass and contribution to
whole body metabolism. The remarkable plasticity of mitochondria allows them to adjust their

Jonathan M. Memme is a PhD candidate and recipient of the Natural


Sciences and Engineering Research Council of Canada (NSERC) Canada
Graduate Scholarship-Doctoral. His research explores the molecular
signals regulating muscle mitochondrial quality control with exercise and
disuse. Avigail T. Erlich is a PhD student and recipient of the Ontario
Graduate Scholarship, investigating mitochondrial turnover and
lysosomal biogenesis in muscle. Geetika Phukan is a postdoctoral fellow
with research focused on mitochondria in Alzheimer’s disease and muscle
chronic contractile activity. David A. Hood is a NSERC Canada Research
Chair in Cell Physiology, Professor in the School of Kinesiology and Health
Science, and the Director of the Muscle Health Research Centre at York
University. His research is devoted to studying mitochondrial biogenesis
and turnover in health and disease in mammalian skeletal and cardiac
muscles with exercise, disuse and ageing. All authors are members of the
Muscle Health Research Centre at York University.

This review was presented at the 2018 ACSM “Integrative Physiology of Exercise (IPE)” conference, which took place at Sheraton San Diego Hotel
and Marina, San Diego, California, 5–8 September 2018.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society DOI: 10.1113/JP278853
804 J. M. Memme and others J Physiol 599.3

volume, structure and capacity under conditions such as exercise, which is useful or improving
metabolic health in individuals with various diseases and/or advancing age. Mitochondria exist
within muscle as a functional reticulum which is maintained by dynamic processes of biogenesis
and fusion, and is balanced by opposing processes of fission and mitophagy. The sophisticated
coordination of these events is incompletely understood, but is imperative for organelle function
and essential for the maintenance of an interconnected organelle network that is finely tuned
to the metabolic needs of the cell. Further elucidation of the mechanisms of mitochondrial
turnover in muscle could offer potential therapeutic targets for the advancement of health and
longevity among our ageing populations. As well, investigating exercise modalities that are both
convenient and capable of inducing robust mitochondrial adaptations are useful in fostering
more widespread global adherence. To this point, exercise remains the most potent behavioural
therapeutic approach for the improvement of mitochondrial health, not only in muscle, but
potentially also in other tissues.

(Received 21 August 2019; accepted after revision 28 October 2019; first published online 1 November 2019)
Corresponding author David A. Hood: Muscle Health Research Centre, School of Kinesiology and Health Science, York
University, Toronto, ON, M3J 1P3, Canada. Email: dhood@yorku.ca

Abstract figure legend The mitochondrial life cycle is regulated by the finely tuned balance of biogenesis and fusion,
opposed to fission and mitophagy. At any moment these processes are in a state of flux and are induced and/or
suppressed depending on the status of the cell and the physiological conditions it is subjected to. A hallmark feature
of aged muscle is the reduction of both subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondrial fractions,
determined by a thinner SS mitochondria layer (yellow arrow) and fragmented IMF pool, as compared to their
younger counterparts. Additionally, aged muscle presents an accumulation of damaged mitochondria (green organelles).
Physical activity is a well-established inducer of mitochondrial remodelling that promotes increases in both SS layer
thickness as well as IMF interconnectivity. Additionally, aged muscle is capable of adapting to the exercise stimulus
which improves mitochondrial content and quality. Indications of a mitochondrial contribution to sarcopenia suggest
that, with regular exercise, mitochondrial health can be preserved well into old age and contribute to whole body
vitality.

Introduction to mitochondrial health synthesis, the sharing of metabolites, and Ca2+ handling
(Spinelli & Haigis, 2018). Conversely, where mitochondria
Mitochondria are unique organelles that are derived from are overabundant and/or dysfunctional, processes of
prokaryotic cells that long ago fused with a host cell. They fission and mitochondrial clearance (mitophagy) occur
are imperative for the provision of energy-rich ATP and to re-establish metabolic homeostasis and maintain
the regulation of cellular longevity across multiple organ mitochondrial health within the cells (Hood et al.
systems. Colloquially termed ‘the powerhouses of the 2019). At any moment these opposing processes are
cell’, this unofficial moniker is an underrepresentation in a state of flux, depending on the metabolic needs
of the variety of functions these mighty organelles of the tissue, to calibrate and promote an optimal
play. However, the fact that mitochondria are widely mitochondrial pool. When the cell is subjected to
recognizable beyond the science community is indicative conditions that perturb metabolic homeostasis, such as
of their relative importance as integral organelles in exercise or advancing age, this balance in mitochondrial
virtually all cells throughout the body. Moreover, the regulation can shift toward an increase in synthesis and
volume of mitochondrial research within the medical fusion, or fission and mitophagy, respectively (Mishra &
sciences community is steadily on the rise. Over the Chan, 2016). The classic mechanism of mitochondrial
past two decades mitochondrial-related publications fusion is achieved through the activity of the proteins
have continued to increase beyond the rate of all other mitofusin-1/2 (Mfn1/2) and optical atrophy protein
cellular organelles, possibly as a result of the recognition 1/2 (Opa1/2) which facilitate fusion of adjacent outer
of the convergence of essential signalling pathways and and inner membranes, respectively, thus establishing an
biological processes on mitochondria (Picard et al. expanded organelle network (Mishra & Chan, 2016).
2016). Conversely, mitochondrial fission is accomplished by the
Mitochondria are capable of adapting to altered joint action of proteins dynamin related protein 1 (Drp1),
metabolic demands. Through processes of biogenesis mitochondrial fission factor (Mff) and fission protein 1
and fusion, newly formed mitochondria are adjoined to (Fis1), the actions of which culminate in the constriction
neighbouring organelles to increase their capacity for ATP


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 805

and cleavage of organelle fragments from the network to Signalling involved in mitochondrial
allow for proper clearance (Losón et al. 2013; Toyama et al. biogenesis
2016).
Recent research has uncovered the complexity of As mammalian mitochondria are derived from symbiotic
mitochondrial morphology, exhibited by connections ancestors, they maintain their own individual 16.5-kb
through either electron dense intermitochondrial genome which works in conjunction with nuclear
junctions (IMJs), as well as membranous protrusions DNA for the expression of mitochondrial proteins
termed nanotunnels (Glancy et al. 2017; Vincent et al. (Calvo et al. 2016). Of the nearly 1200 proteins that
2017). It is proposed that IMJs allow for electrical make up mitochondria, mtDNA is responsible for
coupling of the mitochondrial pool, as well as the the transcription of just 13, albeit integral, electron
ability to rapidly segregate dysfunctional organelles transport chain (ETC) proteins, along with 2 rRNAs
from the reticulum, while simultaneously allowing for and 22 tRNAs (Anderson et al. 1981). Thus, the vast
either the repair or removal of the malfunctioning majority (>99%) of the remaining mitochondrial
organelle (Glancy et al. 2017). In contrast, nanotunnels proteins require transcription in the nucleus and import
promote the sharing of mtDNA, proteins, metabolites into their appropriate organellular compartments via
and other small molecules to non-adjacent mitochondria mitochondrial chaperones and protein import channels.
(Vincent et al. 2017). These small (40–200 nm diameter) This unique biological interaction, requiring the finely
double-membrane extensions appear to exist primarily tuned coordination of both mtDNA and nDNA genomes
to connect mitochondria in tissues with restricted to regulate organelle abundance, has added considerable
mitochondrial motility (Vincent et al. 2019, 2019). fascination, and complexity, to the understanding of
Nanotunnels may develop as a compensatory mechanism mitochondrial health, disease and turnover.
of mitochondrial crosstalk under stress conditions, when The nuclear transcription of mitochondrial proteins
more direct mitochondrial communication is not possible is undoubtedly regulated by many proteins, but none
(Vincent et al. 2019, 2019). is more prominent in the literature than the trans-
Mitochondrial morphology and function also differ criptional coactivator peroxisome proliferator activated
across tissues, in order to match the specialized receptor γ coactivator 1α (PGC-1α), considered the
metabolic demand of their respective cells, or the master regulator of mitochondrial biogenesis (Hood,
changing pathological or physiological conditions 2001; Handschin & Spiegelman, 2006; Scarpulla, 2011;
that they are subjected to (Fernández-Vizarra et al. Scarpulla et al. 2012). PGC-1α and its family members,
2011). Skeletal muscle provides an excellent tissue to PGC-1β and PGC-related co-activator (PRC), upregulate
investigate the various morphological configurations gene transcription by docking with transcription factors
of mitochondria within a cell. When viewed using (TFs) and additional proteins on DNA promoters to
electron microscopy (EM), mitochondria within muscle regulate nuclear genes encoding mitochondrial proteins
have distinct geographical subpopulations (Kayar et al. (NuGEMPs) (Puigserver et al. 1999; Scarpulla, 2011;
1988; Cogswell et al. 1993). Below the sarcolemmal Scarpulla et al. 2012). Acute exercise initiates a
membrane reside the subsarcolemmal (SS) mitochondria multitude of signals that converge on PGC-1α, with
which present the more classic rounded appearance. the most widely accepted signals being: (i) activation of
They specialize in providing ATP for nuclear gene calcium/calmodulin-dependent protein kinase (CaMK),
transcription and membrane transport (Kirkwood et al. through the increased intracellular Ca2+ concentration
1986; Ogata & Yamasaki, 1997). Interspersed throughout (Ojuka et al. 2003); (ii) activation of p38 mitogen-activated
the contractile myofibrillar protein network are the protein kinase (p38 MAPK), which is sensitive to multiple
intermyofibrillar (IMF) mitochondria, which are more stressors such as reactive oxygen species (ROS) (Puigserver
elongated and interconnected (Kirkwood et al. 1986; et al. 2001; Akimoto, 2005; Hood, 2009; Irrcher et al.
Ogata & Yamasaki, 1997). These mitochondria provide 2009; Ristow et al. 2009); and (iii) AMP-activated protein
ATP for muscle contraction and they are tightly connected kinase (AMPK) phosphorylation, activated by the hydro-
to the sarcoplasmic reticulum (SR). Thus, these may lysis of ATP by myosin ATPases and the resultant increase
additionally play a prominent role in Ca2+ signalling in AMP:ATP ratio (Winder et al. 2000; Irrcher et al.
(Ogata & Yamasaki, 1997; Boncompagni et al. 2009). 2009). Additionally, cAMP is another activator of PGC-1α
In response to whole-body exercise, skeletal muscle expression which may be induced through adrenergic
mitochondria increase their rates of ATP synthesis to signalling (Handschin et al. 2003; Fig. 1). Once activated,
match the metabolic demands of the cell. Coincidentally, PGC-1α can then interact with a variety of TFs, such
a myriad of signalling events converge to activate nuclear as nuclear respiratory factor (NRF)-1/2, which induce
and cytoplasmic proteins to orchestrate the initiation the expression of mitochondrial transcription factor A
of biogenesis, as well as mitophagy of a pre-existing, (Tfam). Tfam is subsequently imported into the organelle
unhealthy mitochondrial pool. and serves as the most important TF to upregulate the


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
806 J. M. Memme and others J Physiol 599.3


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 807

transcription of mtDNA-derived proteins (Gordon et al. disruptions in mitochondrial protein import can arise
2001; Scarpulla, 2011). As such, this sequence of events as a result of nuclear/mitochondrial imbalances. When
plays an important role in matching the expression of the cell is under stress, such as during development
mitochondrially encoded genes to the changing expression or with exercise, protein import is accelerated and the
of the nuclear genome. This transcriptional coordination UPRmt is activated through the upregulation of HSP60
is important, as any imbalance between the expression and CPN10 to ensure that the accumulated unfolded
levels of either genome can lead to a disruption in proteins achieve their mature configuration (Memme
proteostasis within the organelle, and trigger a series of et al. 2016; Mesbah Moosavi & Hood, 2017; Oliveira
mitochondrial retrograde signals to the nucleus involving & Hood, 2018). Additionally, increased expression of
the mitochondrial unfolded protein response (UPRmt ) as mitochondrial proteases lon peptidase (LonP) and
an attempt to maintain a proper, functional proteome. caseinolytic mitochondrial matrix peptidase proteolytic
Further retrograde signalling can originate subunit (ClpP) degrade misfolded proteins to alleviate
post-transcriptionally at the level of protein import. their proteotoxic burden (Mottis et al. 2014).
Nuclear-encoded mitochondrial proteins contain The trigger that initiates the upregulation of these
a mitochondrial targeting sequence (MTS) that is chaperones may be ROS. ROS activate the stress
recognized by cytosolic chaperones. These chaperones are kinase, general control nonderepressible 2 (GCN2).
responsible for unfolding their cargo and delivering the Activated GCN2 phosphorylates the eukaryotic initiation
protein to the mitochondrial protein import machinery factor alpha (eIF2α), subsequently decreasing global
(PIM), comprising the translocases of the outer and inner protein synthesis, thus reducing the level of nascent
membranes (TOM and TIM, respectively) (Bolender et al. proteins that require import and assembly within
2008; Chacinska et al. 2009). A matrix-targeted protein the mitochondria (Shenton et al. 2006; Baker et al.
enters the mitochondria through the TOM complex 2012). While phosphorylated eIF2α decreases general
and then subsequently through the TIM complex, protein translation, it selectively increases translation of
before reaching its final destination within the organelle proteins containing an uORF in the 5ʹ UTR, such as
(Chacinska et al. 2009). In the matrix, the MTS is cleaved CCAAT/enhancer-binding protein (C/EBP) homologous
by the mitochondrial processing peptidase (MPP) and protein (CHOP), and the activating transcription factors
the protein is refolded to its mature conformation by 4 and 5 (ATF4 and ATF5) to upregulate the expression
mitochondrial chaperones, such as mtHSP70, HSP60 or of mitochondrial chaperones and proteases (Fiorese et al.
CPN10, and may be incorporated with other nuclear- 2016; Melber & Haynes, 2018). Proteolytic cleavage of
or mitochondrially encoded proteins to form functional misfolded proteins also generates peptide fragments that
complexes (Harbauer et al. 2014; Fig. 1). However, directly suppress organellular import capacity (Fig. 1).

Figure 1. Regulation of mitochondrial health with exercise and the influence of age
Exercise induces robust changes in mitochondrial content and quality that are beneficial for metabolic health. A,
motor unit recruitment of myofibres produces action potentials that induce Ca2+ release from the SR. Increased
cytosolic Ca2+ prompts myosin-actin interactions, crossbridge cycling, and the hydrolysis of ATP to produce muscle
force, as well as the generation of ATP and ROS from respiring mitochondria. B, consequently, these events activate
the signalling kinases that converge on PGC-1α, allowing its translocation to the nucleus where it coactivates TFs
to increase the expression of NuGEMPS, such as the mtDNA transcription factor, Tfam. C, newly expressed
nuclear-derived proteins are then imported into mitochondria through translocases of the outer, and then inner
membranes (TOM and TIM, respectively). D, the exercise stimulus increases protein levels within mitochondria,
which promotes the activation of the UPRmt . The cleavage of terminally misfolded protein aggregates in the matrix
and the release of their peptide fragments, block ATF5 mitochondrial entry, thus redirecting it to the nucleus to
upregulate transcription of mitochondrial chaperones and proteases and equip the organelle with an augmented
capacity for protein folding. E, mitochondrial fusion proteins Mfn1/2 along with Opa1 facilitate the fusion of the
outer and inner membranes, respectively, allowing for improved sharing of metabolites amongst neighbouring
organelles. F, regions of the mitochondrial network may become dysfunctional as they are incapable of matching
the metabolic needs of the tissue and require segregation from the reticulum in order to spare their adjacent
organelles from further impairment. These damaged organelles undergo fission, mediated by the interaction of
proteins Fis1 and Mff with Drp1 to constrict and remove the organelles, allowing for their clearance via mitophagy.
G, once separated, dysfunctional mitochondria with a low  accumulate PINK1 on their outer membrane. PINK1
recruits the E3-ligase Parkin, which subsequently ubiquitinates outer membrane proteins to flag the organelle for
removal via mitophagy. The adapter protein p62 binds to the ubiquitin on the tagged cargo, as well as to LC3-II
embedded in the phagophore membrane, and promotes the formation of the autophagosome, which fuses
with the lysosome to degrade the mitochondria and release the constituent amino acids for cellular recycling. H,
aged muscle displays attenuated PGC-1α signalling for mitochondrial biogenesis (red line), as well as increased
fission:fusion protein ratio (green line), thus promoting a fragmented network of organelles, as well as suppressed
mitophagy flux.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
808 J. M. Memme and others J Physiol 599.3

This inhibition of import promotes a mechanistic switch those involved in β-oxidation, the tricarboxylic acid (TCA)
towards mitochondrial clearance through mitophagy via cycle, and the electron transport chain, thus improving
stabilization of PTEN-induced putative kinase protein the capacity for energy provision to the exercising muscle
1 (PINK1) on the outer membrane of dysfunctional (Holloszy & Booth, 1976). In fact, a single bout of end-
organelles (see below). Thus, the protein import process is urance exercise is sufficient to induce structural changes
not only essential for the translocation of nuclear-derived in the mitochondrial network that promote augmented
proteins into the organelle, it also serves to match the function of the organelle network (Picard et al. 2013). With
health status of the organelle to the appropriate signalling prolonged endurance training, mitochondrial volume
response in order to maintain an optimal mitochondrial typically increases as much as 40–50% and this increase
pool. Altogether, the UPRmt within skeletal muscle has in content is paralleled by more modest improvements in
been shown to be important for the differentiation of respiration and oxidative capacity of the mitochondria on
C2C12 myoblasts into mature myotubes, as well as in a per organelle basis (Baldwin et al. 1972). Additionally,
the basal regulation of mitochondrial maintenance within mitochondrial adaptations to exercise vary across the
skeletal muscle in vivo (Mesbah Moosavi & Hood, 2017; different fibre types, depending on the initial organelle
Oliveira & Hood, 2018). content, as well as the degree of motor unit recruitment
during the training session. In humans, slow-twitch (type
The effect of various exercise training I) fibres contain the highest percentage of mitochondria,
followed by fast-twitch red (type IIa) fibres, and the whiter
modalities on mitochondria
type IIx fibres, respectively (Howald et al. 1985). With
It is now well recognized that exercise is a potent training, the mitochondrial content within any of these
stimulus to induce the signalling pathways described fibre types can be enhanced, indicating that mitochondrial
above, which ultimately produce robust phenotypic adaptations are not dependent on the myosin-based fibre
changes in the mitochondrial milieu and improve the type per se, but instead are based on the stimulus and the
quantity and quality of the organelle network, leading recruitment of that fibre (Lundby & Jacobs, 2016).
to greater muscle health. It was through John Holloszy’s Terjung and colleagues illustrated the important inter-
pioneering work that we first came to understand the play between intensity and duration in determining the
important training parameters that are required to achieve extent of mitochondrial adaptation in each fibre type
a particular level of adaptation, and how exercise training (Dudley et al. 1982). Type I fibres in particular are
promotes mitochondrial biogenesis (Holloszy, 1967). most easily recruited at exercise intensities as low as
Favourable mitochondrial adaptations in muscle can 40% of V̇O2 max and lower. As workload increases and
only be achieved if training is performed at a sufficient exceeds 40% of V̇O2 max, type IIa fibres are recruited, and
frequency, intensity and duration, for an adequate length only after the exercise intensity surpasses about 75% of
of time (Holloszy, 1967). Since the 1960s, decades’ worth V̇O2 max will type IIx fibres be engaged (Sale, 1987). Based
of research has demonstrated that exercise promotes on this principle – that motor units must be recruited
a robust increase in mitochondrial content, as well in order to adapt – training approaches have evolved
as improved oxidative phosphorylation and respiratory from the traditional long-distance endurance exercise to
capacity per mitochondrion (Burgomaster et al. 2008; alternative, time-saving modalities, such as sprint inter-
Jacobs & Lundby, 2013; Porter et al. 2015). Additionally, val training (SIT), high intensity interval training (HIIT)
chronic training reduces the production of ROS, indicative and even resistance exercise, which can lead to diverse
of an enhanced capacity for electron flow through the mitochondrial adaptations in muscle.
ETC (Holloway, 2017). Given that these adaptations are Resistance exercise is typically associated with muscle
favourable for improved muscle health, scientists have hypertrophy and improved force-generating capacity, as
continually strived to determine the precise signalling opposed to fatigue resistance and improved aerobic energy
pathways mediating mitochondrial quality control, which metabolism (Baar, 2006). Historically, the expansion of
includes the result of biogenesis and fusion, as opposed to muscle cell size via hypertrophy has been viewed as
fission and mitophagy. ‘diluting’ mitochondrial content in muscle and increasing
Since Hoppeler et al. (1973) described a correlation the diffusion distances (Lüthi et al. 1986). Indeed, a
between mitochondrial volume and V̇O2 peak , the recent review (Groennebaek & Vissing, 2017) of the
association of mitochondrial content within muscle and effect of resistance exercise has revealed the disparate
exercise capacity has been a focal point for fitness and results obtained on mitochondria. However, it now
exercise practitioners to harness modalities that improve appears that resistance exercise has the potential of
aerobic potential and performance. For many years, end- inducing tangible improvements in maximal coupled
urance training was considered the primary means of respiration measured in permeabilized myofibres, albeit
achieving mitochondrial adaptations. Endurance exercise without a parallel increase in mitochondrial gene
training increases total mitochondrial proteins including expression or mitochondrial mass (Porter et al. 2015;


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 809

Groennebaek & Vissing, 2017). These adaptations may the E3-ubiquitin ligase Parkin to mitochondria, which
be more pronounced in older individuals or in diseased mediates the ubiquitination of various outer membrane
muscle such as sporadic inclusion body myositis, where proteins such as Mfn2 and the voltage-dependent anion
mitochondrial gene expression and, subsequently, content channel (VDAC), tagging the organelle for degradation
is reduced in the basal state (Melov et al. 2007; Koo et al. (Geisler et al. 2010; Hood et al. 2011). The formation
2019). of the autophagic vesicle, the phagophore, begins once
Interval training modalities such as HIIT and SIT have microtubule-associated protein 1 light chain 3 (LC3)-I
gained considerable popularity of late, and are capable is lipidated into LC3-II with the aid of a collection
of eliciting a similar level of adaptation as traditional of autophagy-related gene products such as ATG7
endurance training while doing so in considerably less (Tanida et al. 2004). The adapter protein p62/SQSTM1
time, with a reduced exercise volume (MacInnis & Gibala, contains both ubiquitin and LC3-II interacting domains,
2017). These exercise conditions involve the recruitment and it serves as an anchor between the ubiquitinated
of all motor units, and this contributes to the detection mitochondrion and the double-membraned phagophore,
of mitochondrial adaptations in all muscle fibres within thus facilitating organelle encapsulation and the formation
a mixed fibre biopsy sample (MacInnis & Gibala, 2017). of the mature autophagosome (Geisler et al. 2010;
High intensity training bouts promote the activation of Matsuda et al. 2010; Kim & Hood, 2017; Chen et al.
the various signalling kinases that converge on PGC-1α to 2018a,b). The final stage of mitophagy requires the fusion
promote organelle synthesis, as with endurance exercise of the autophagosome with lysosomes for subsequent
(Gibala et al. 2009; Little et al. 2011). However, exercise breakdown by proteolytic enzymes within the lysosomal
at higher intensities generates more rapid ATP hydrolysis lumen (Vainshtein & Hood, 2016). Therefore, lysosomal
and greater Ca2+ release to generate force. This should health is extremely important for the maintenance of
lead to the enhanced activation of signalling kinases such mitochondrial quality in muscle.
as p38 MAPK, AMPK and CaMKII, and could account for The transcription factors that regulate lysosomal
the observations that repeated bouts of both HIIT and SIT biogenesis and function include the basic helix loop helix
produce as much as 25–35% increases in mitochondrial (bHLH)-leucine zipper transcription factors TFEB and
content in as few as 6–7 sessions (Gollnick et al. 1974; Egan TFE3. The overexpression of TFEB and TFE3 results in an
et al. 2010; MacInnis & Gibala, 2017). increased number of lysosomes and lysosomal enzymes
which serve to enhance catabolic activity (Settembre et al.
2013; Yang et al. 2018), while the depletion of these
Mitochondrial turnover and exercise
proteins reduces the expression of lysosomal genes. In
While the activation of the organelle biogenesis pathway addition to regulating lysosomal biogenesis, TFEB has also
leads to an increase in mitochondrial content as been shown to be involved in regulating mitochondrial
a result of training, it is also vital to eliminate biogenesis, and to coordinate the expression of genes
any mitochondrial segments that have outlived their involved in autophagosome biogenesis and lysosome
usefulness via mitophagy, in order to maintain or improve fusion, indicated by an increased clearance of lipid droplets
the quality of the mitochondrial pool (Ljubicic et al. 2009; and mitochondria with TFEB overexpression (Mansueto
Lira et al. 2013; Kim et al. 2018). Mitophagy occurs when et al. 2017).
double-membraned vesicles, autophagosomes, engulf Mitophagy is up-regulated by numerous cellular
damaged organelles that are flagged for degradation stresses, including the energetic imbalance brought about
by specialized proteins, when they exhibit a decreased by acute exercise (Vainshtein & Hood, 2016; Erlich
membrane potential and/or excessive increases in ROS et al. 2018; Chen et al. 2018b; Hood et al. 2019).
production (Wei et al. 2015; Kim & Hood, 2017; Kim During exercise, the AMP/ATP ratio increases, thereby
et al. 2018; Chen et al. 2018b; Triolo & Hood, 2019). activating AMPK and its downstream target Unc-51
The main mitophagy pathway that has been investigated like autophagy activating kinase 1 (ULK1). At the same
in the context of exercise involves PINK1 and Parkin time mTORC1, a known suppressor of the process, is
(Fig. 1), two proteins that are well recognized for inhibited (Kim et al. 2014; Tian et al. 2015; Laker et al.
their involvement, when mutated, in Parkinson’s disease 2017). This sequence of initial steps has been shown to
(Geisler et al. 2010; Matsuda et al. 2010; Lira et al. activate mitophagy, as evident 6 h post-exercise using
2013; Wei et al. 2015). Normally, PINK1 is imported the pMitoTimer reporter gene in vivo (Lira et al. 2013;
into mitochondria and degraded by resident proteases. Laker et al. 2017) Other studies have shown that acute
However, if the mitochondrial membrane potential exercise has a more rapid effect, by promoting Parkin
dissipates, PINK1 accumulates on the outer membrane localization to mitochondria as well as an increase in
as the protein import machinery loses its functionality mitophagy flux, measured via LC3-II, p62 and ubiquitin
(Geisler et al. 2010; Matsuda et al. 2010; Wei et al. 2015). immediately post-exercise (Vainshtein et al. 2015; Chen
The stabilization of PINK1 facilitates the recruitment of et al. 2018a,b). This appears to be regulated in part by


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
810 J. M. Memme and others J Physiol 599.3

PGC-1α, since the exercise-induced increase in mitophagy tissue, and has extended implications on mobility, the
signalling and flux were not apparent in PGC-1α knockout risk of falls, and the onset of type 2 diabetes and obesity
animals (Vainshtein et al. 2015). Acute exercise is also (López-Otı́n et al. 2013; Carter et al. 2015). Mitochondria
capable of enhancing the activation and localization of are culpable organelles in the progression of sarcopenia as
TFEB to the nucleus, possibly due to transient Ca2+ they are important regulators of a variety of factors that
fluxes and subsequent calcineurin activation, which would contribute to the etiology of the condition, such as ATP
dephosphorylate TFEB and mediate its translocation provision, oxidative stress, proteostasis, and apoptosis, as
(Erlich et al. 2018). Therefore, exercise acts as a stimulus well as inflammation and Ca2+ handling (Heber et al.
for the induction of both PGC-1α and TFEB, which 1996; Rapizzi et al. 2002; Kujoth et al. 2005; Schaap et al.
act in concert to regulate both organelle biogenesis, as 2006). The natural process of ageing along with prolonged
well as mitophagy (i.e. turnover). Thus, these proteins sedentarism promotes impairments in mitochondrial
are critically involved in maintaining the health of the integrity, further contributing to the progressive nature
mitochondrial pool within skeletal muscle. of sarcopenia. Aged muscle can exhibit both fragmented
In response to repeated bouts of exercise in the form of mitochondria, and/or atypically enlarged organelles when
endurance training, a number of studies have documented viewed under EM (Iqbal et al. 2013; Leduc-Gaudet et al.
increases in autophagy and mitophagy markers within 2015). Both SS and IMF fractions appear visibly thinner
muscle (Lira et al. 2013; Ju et al. 2016). Chen et al. and smaller within aged muscle compared to young
(2018b) provided a direct measurement of mitophagy flux counterparts, and many studies have reported decreased
utilizing colchicine treatment following a 6 week training mitochondrial content with age (Short et al. 2003, 2005;
protocol. They found enhanced Parkin localization to the Chabi et al. 2008; Ljubicic et al. 2009; Iqbal et al. 2013).
mitochondria, as well as an increase in Parkin expression in This reduction is also paralleled by impaired protein
muscle of trained mice. Despite this, the exercise-induced synthesis and respiration, and increased uncoupling
increase in mitophagy flux was reduced following training of O2 consumption to ATP synthesis contributing to
(Lira et al. 2013; Chen et al. 2018b; Hood et al. 2019). reduced maximal ATP reduction rate (MAPR) (Boffoli
Other studies showed that mitophagy flux was reduced et al. 1994; Rooyackers et al. 1996; Marcinek et al.
in the basal state following a period of chronic contra- 2005). The reasons for this decline at the cellular level
ctile activity (CCA), an alternative model of endurance include reductions in the transcription of numerous genes
training (Carter et al. 2018a; Leduc-Gaudet et al. 2019). encoding mitochondrial enzymes, which are paralleled at
It is apparent from these studies, summarized elsewhere the protein level (Baraibar et al. 2013; Liu et al. 2013;
(Guan et al. 2019), that the improvement in mitochondrial Carter et al. 2015). For example, at both the transcript
function evident with these types of exercise reduced the and protein level, proteins involved in organelle fission
necessity for mitophagy signalling. In addition, chronic and fusion are suppressed in muscle with age (Ibebunjo
exercise results in significant elevations in TFEB protein et al. 2013). Additionally, there is an apparent shift in
as well as lysosomal markers Cathepsin D, mucolipin the balance of these factors towards enhanced fission,
(MCOLN1), and lysosomal associated membrane proteins which agrees with the morphometric data suggesting a
(LAMP) 1 and 2 (Kim & Hood, 2017; Mansueto et al. fragmented network (Iqbal et al. 2013). As PGC-1α is
2017; Kim et al. 2018). These results support the idea considered the master regulator of mitochondrial protein
that chronic exercise induces the expression of lysosomes expression, it is perhaps unsurprising that it, also, is
and autophagy-related genes which ensure that muscle diminished at both the mRNA and protein level with
is primed to clear dysfunctional organelles, including age, and this decreased expression probably contributes
mitochondria, if their degradation is required. Given the to the reductions in mitochondrial volume that are
similar increases in mitochondrial content and function observed (Conley et al. 2007; Chabi et al. 2008; Koltai
observed in HIIT and SIT as compared to traditional et al. 2012; Fig. 1). PGC-1α levels are reduced in ageing
endurance training, it is reasonable to consider that muscle as a consequence of decreased transcription of
mitochondrial turnover would be induced to a similar the PGC-1α gene, possibly accompanied by enhanced
extent in these alternative training modalities. However, promoter methylation, and age-associated alterations
little research has explored these relationships, and future in the balance of regulatory transcription factors
work in this area is warranted. which determine PGC-1α transcription (Carter et al.
2018b).
Is mitochondrial health maintained with Another potential cause of mitochondrial dysfunction
that contributes to sarcopenia is the advancement of
age?
mtDNA mutations that occurs with progressing age. In
A hallmark feature of ageing is the progressive loss of fact, there is evidence to suggest that, where fibres harbour
muscle mass, termed sarcopenia, which develops from mtDNA mutations in excess of 80%, there is a greater
structural and molecular changes occurring within the indication of fibre breakage and atrophy (Wanagat et al.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 811

2001; Bua et al. 2006). These mutations would give rise to vitality. Individuals that have maintained high physical
decrements in overall ETC function, which could in turn activity throughout life have preserved mitochondrial
contribute, in part, to the progressive atrophy observed in content and function (Lanza et al. 2008; Joseph et al.
adults after 50 years of age (Carter et al. 2015). However, 2012). In addition, those who take to exercise at advanced
since other investigators have described no link between age are also capable of restoring mitochondrial content
mitochondrial COX deficiency and fibre atrophy (Rowan and function to more closely resemble that of their
et al. 2011), further work in this area seems to be required. younger counterparts, when training programmes are
A central issue regarding mitochondrial health in ageing matched for relative intensity (Short et al. 2003; Carter
muscle is whether the apparent decline in function and et al. 2015). Thus, exercise remains a reliable and
content is a result of increasing levels of physical inactivity, non-pharmacological strategy for the maintenance of
or a consequence of the ageing process per se. Previous metabolic health into the late stages of life.
reviews have provided a more complete analysis of this
issue (Hood et al. 2016). What is clear is that regular
Mitochondrial health and the brain
exercise can ameliorate at least a portion of the decline in
organelle health with age. However, this is accomplished While this review has focused on mitochondrial health in
with reduced sensitivity to the exercise stimulus, as skeletal muscle, the benefits of exercise are widespread,
signalling kinase activation is attenuated in muscle with affecting other organ systems. The effects of exercise
age, as is the ability to remove dysfunctional mitochondria on mitochondrial health in other tissues has been
via exercise-induced mitophagy (Ljubicic & Hood, 2009; sparsely studied, but the recognition that muscle secretes
Chen et al. 2018a). Resistance exercise may offer some myokines, or exerkines, during exercise, suggests that
favourable advantages. Despite the reduced stimulus for the health benefits of exercise may include muscle
mitochondrial biogenesis that this mode of exercise creates activity-mediated benefits to other tissues, such as the
(see discussion above), it does stimulate muscle fibre brain. The brain consumes about 20% of the body’s total
hypertrophy, and it prompts the recruitment and fusion of oxygen consumption, and it is almost entirely dependent
satellite cells, which do not exhibit the mtDNA mutations on mitochondrial energy production, as neurons have
that are found in mature muscle fibres (Taivassalo, 1999; limited glycolytic activity. Much of this energy is utilized
Kowald & Kirkwood, 2014; Carter et al. 2015). by neurons to establish membrane potentials, synthesize,
Aberrant mitophagy is also observed in sarcopenic secrete and recycle neurotransmitters, as well as maintain
tissue (Fig. 1), which both exacerbates the dysfunction cellular Ca2+ homeostasis for effective neuronal signal
of the reticulum contributing to advanced atrophy and transmission. Mitochondria are also major producers
also explains the formation of abnormally enlarged of ROS in the brain, and ROS-induced toxicity is an
mitochondria when viewed under electron micro- established cause of pathologies observed in neuro-
scope. However, the question of whether the increased degenerative diseases such as Alzheimer’s disease (AD),
rates of mitophagy that occur in aged tissue are pro- Parkinson’s disease (PD), and multiple sclerosis (MS)
or maladaptive remains, since mitochondria remain (Park et al. 2018). Deficits in cellular bioenergetics,
dysfunctional despite measured elevations in mitophagy along with an oxidized redox environment, are the
flux (O’Leary et al. 2013; Carter et al. 2018a; Chen et al. major contributors to an age-related decline in neuro-
2018a). A final consequence of inadequate organelle nal health, as evidenced by the dysfunctional TCA
recycling is the overproduction of ROS leading to the cycle, compromised electron transport and altered
subsequent activation of apoptosis. Indeed, in aged tissue mitochondrial morphology evident in post-mortem
ROS are elevated along with reduced Ca2+ retention. tissues of AD patients (Radak et al. 2016).
This promotes the mitochondrial release of pro-apoptotic Exercise has long been associated with the wellbeing
factors and subsequent DNA fragmentation, ultimately of neuronal health, and it is routinely prescribed as
promoting regional atrophy along the myofibre (Bua a therapeutic and preventative strategy in dementia
et al. 2002; Chabi et al. 2008; Gouspillou et al. 2014). patients. Exercise also alters brain structure in neuro-
Fortunately, exercise can serve to reduce the accumulation degenerative disease patients, mediating increases in
of mitochondrial ROS and attenuate apoptotic cell death angiogenesis, hippocampal neurogenesis, synaptic
in muscle (Ljubicic et al. 2009; Di Meo et al. 2019). Indeed, plasticity, and decreases in age-related brain atrophy
endurance training is capable of increasing the expression (Bernardo et al. 2016). Coincident with this are
levels of a majority of the downregulated proteins that are exercise-induced alterations in mitochondrial function
involved in energy metabolism, and of restoring the ATP and biogenesis in neurons. Voluntary exercise induced
synthesis capability of aged muscle (Lanza et al. 2008). increased uncoupling protein (UCP) 2 mRNA expression
Indeed, lifelong training, or even the adoption of physical and mitochondrial oxygen consumption in mouse
activity behaviours later in life, are established therapies hippocampus, along with a concomitant increase in
to improve mitochondrial health in order to preserve dendritic spines and mitochondrial number in wild-type,


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
812 J. M. Memme and others J Physiol 599.3

but not in UCP2 knockout mice (Dietrich et al. 2008). variety of signalling pathways and biological processes that
Likewise, exercise promotes improved mitochondrial impact on cellular health, beyond ATP provision. Skeletal
function through enhanced complex I activity in the muscle is of significant interest in that it comprises a large
brains of aged mice (Gusdon et al. 2017). Chronic exercise proportion of body mass, it is highly metabolic, and its
also increased the levels of various mitochondrial TCA mitochondrial content is malleable to external stimuli.
enzymes and ETC activity in neurons, as well as the Thus, muscle represents an ideal tissue for the study of
synthesis and secretion of various neurotrophins, such as mitochondria in health and disease.
brain derived neurotrophic factor (BDNF) (Navarro et al. Scientists are now beginning to appreciate the molecular
2004; Ding et al. 2006; Marques-Aleixo et al. 2015). BDNF mechanisms by which mitochondrial content, function
signalling is linked to mitochondrial health because it and dynamics are maintained within the cell. Regulation
improves the respiratory coupling efficiency of synaptic of the finely tuned balance between biogenesis and
mitochondria, it upregulates antioxidant enzymes, and mitophagy, the maintenance of a healthy network of
it mediates PGC-1α-induced mitochondrial biogenesis organelles, and their ability to interconnect where
(Marosi & Mattson, 2014). Such adaptations induced by appropriate in order to enhance the efficiency of the
chronic exercise can reduce harmful redox aberrations functional reticulum, are areas of important future study.
observed in AD patients. In addition, a greater understanding of the redundancies
How might exercise modulate mitochondrial health in in the nuclear-derived signalling leading to mitochondrial
the brain (or in other tissues)? When muscle contra- turnover, as well as the retrograde signalling from
cts it releases a combination of peptides, lipids, mRNA, the organelle to the nucleus, also warrant considerable
microRNAs and mtDNA, collectively termed myokines research effort. Elucidation of these signalling networks
(Safdar et al. 2016; Trovato et al. 2019). These are trans- can help identify pharmacological targets that would be
ported via exosomes, small extracellular vesicles that can useful adjuncts to enhance mitochondria in the pre-
be secreted into the extracellular environment and can sence of an exercise stimulus. This would be of inter-
be taken up by distant cells via endocytosis (Feng et al. est to scientists and health care professionals, given the
2010; Whitham et al. 2018). The finding that intra- decrements in mitochondria observed with age, as well
cellular Ca2+ can influence vesicular secretion garnered as the multitude of secondary diseases that may arise
interest in skeletal muscles, because Ca2+ release by SR which depend on mitochondrial function, such as type
signals muscle contraction and leads to changes in gene 2 diabetes and obesity. Thus, exercise remains our best
expression (Safdar et al. 2016). During contractile activity, behavioural medicine for improving mitochondrial health
the secretome contains hundreds of myokines, which may and individual longevity.
contribute to adaptations in various organs in a paracrine From an applied perspective, while endurance
or endocrine fashion (Giudice & Taylor, 2017; Hoffmann exercise training has long been appreciated to enhance
& Weigert, 2017). Some of these include members of skeletal muscle aerobic capacity through augmented
the interleukin (IL) family of proteins, BDNF, fibroblast mitochondrial quality control, content and function,
growth factor 21 (FGF-21), irisin, and vascular endothelial alternative exercise training modalities are gaining traction
growth factor (VEGF), and these can potentially mediate as viable interventions capable of inducing similar
signalling between muscle and various organs, including improvements in mitochondria, while condensing a
the brain, and other tissues, thereby transmitting the similar workload into a shorter period, and thus achieving
beneficial effects of exercise to these tissues (Pedersen & a level of time efficiency per exercise bout. Further work
Febbraio, 2012; Covington et al. 2016; Lu et al. 2016; Safdar exploring different training programmes that improve the
et al. 2016; Gomes et al. 2017; Kim & Song, 2017). Physical efficiency of the stimulus and foster an increase in public
activity has long been established to have a beneficial role adoption present an attractive area of study as well.
in preventing neurodegenerative diseases and improving
the cognitive abilities of affected people (Vecchio et al.
2018). Investigating the role of exercise-induced myokines References
in influencing mitochondrial health in other tissues, such
as the brain, is worthy of further study. Akimoto T (2005). Exercise stimulates Pgc-1 transcription in
skeletal muscle through activation of the p38 MAPK
pathway. J Biol Chem 280, 19587–19593.
Conclusion and future perspectives Anderson S, Bankier AT, Barrell BG, de Bruijn MH, Coulson
AR, Drouin J, Eperon IC, Nierlich DP, Roe BA, Sanger F,
The role of mitochondria in contributing to overall health Schreier PH, Smith AJ, Staden R & Young IG (1981).
and vitality is continually coming into sharper focus, Sequence and organization of the human mitochondrial
evidenced by the increasing volume of mitochondrial genome. Nature 290, 457–465.
research that is being conducted within the medical science Baar K (2006). Training for endurance and strength. Med Sci
community. Indeed, these organelles are at the nexus of a Sport Exerc 38, 1939–1944.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 813

Baker BM, Nargund AM, Sun T & Haynes CM (2012). Chacinska A, Koehler CM, Milenkovic D, Lithgow T & Pfanner
Protective coupling of mitochondrial function and protein N (2009). Importing mitochondrial proteins: machineries
synthesis via the eIF2α kinase GCN-2. PLoS Genet 8, and mechanisms. Cell 138, 628–644.
e1002760. Chen CC, Erlich AT, Crilly MJ & Hood DA (2018a). Parkin is
Baldwin KM, Klinkerfuss GH, Terjung RL, Molé PA & Holloszy required for exercise-induced mitophagy in muscle: impact
JO (1972). Respiratory capacity of white, red, and of aging. Am J Physiol - Endocrinol Metab 315, E404–E415.
intermediate muscle: adaptative response to exercise. Am J Chen CC, Erlich AT & Hood DA (2018b). Role of Parkin and
Physiol 222, 373–378. endurance training on mitochondrial turnover in skeletal
Baraibar MA, Gueugneau M, Duguez S, Butler-Browne G, muscle. Skelet Muscle 8, 1–14.
Bechet D & Friguet B (2013). Expression and modification Cogswell AM, Stevens RJ & Hood DA (1993). Properties of
proteomics during skeletal muscle ageing. Biogerontology 14, skeletal muscle mitochondria from subsarcolemmal and
339–352. intermyofibrillar isolated regions. Am J Physiol Cell Physiol
Bernardo TC, Marques-Aleixo I, Beleza J, Oliveira PJ, Ascensão Physiol 264, C383–C389.
A & Magalhães J (2016). Physical exercise and brain Conley KE, Marcinek DJ & Villarin J (2007). Mitochondrial
mitochondrial fitness: the possible role against Alzheimer’s dysfunction and age. Curr Opin Clin Nutr Metab Care 10,
disease. Brain Pathol 26, 648–663. 688–692.
Boffoli D, Scacco SC, Vergari R, Solarino G, Santacroce G & Covington JD, Tam CS, Bajpeyi S, Galgani JE, Noland RC,
Papa S (1994). Decline with age of the respiratory chain Smith SR & Redman LM (2016). Myokine expression in
activity in human skeletal muscle. Biochim Biophys Acta muscle and myotubes in response to exercise stimulation.
1226, 73–82. Med Sci Sport Exerc 48, 384–390.
Bolender N, Sickmann A, Wagner R, Meisinger C & Pfanner N Dietrich MO, Andrews ZB & Horvath TL (2008).
(2008). Multiple pathways for sorting mitochondrial Exercise-induced synaptogenesis in the hippocampus is
precursor proteins. EMBO Rep 9, 42–49. dependent on UCP2-regulated mitochondrial adaptation.
Boncompagni S, Rossi AE, Micaroni M, Beznoussenko G V, J Neurosci 28, 10766–10771.
Polishchuk RS, Dirksen RT & Protasi F (2009). Di Meo S, Napolitano G & Venditti P (2019). Mediators of
Mitochondria are linked to calcium stores in striated muscle physical activity protection against ROS-linked skeletal
by developmentally regulated tethering structures. Mol Biol muscle damage. Int J Mol Sci 20, 3024.
Cell 20, 1058–1067. Ding Q, Vaynman S, Souda P, Whitelegge JP & Gomez-Pinilla
Bua EA, Johnson J, Herbst A, Delong B, McKenzie D, Salamat S F (2006). Exercise affects energy metabolism and neural
& Aiken JM (2006). Mitochondrial DNA – deletion plasticity-related proteins in the hippocampus as revealed by
mutations accumulate intracellularly to detrimental levels in proteomic analysis. Eur J Neurosci 24, 1265–1276.
aged human skeletal muscle fibers. Am J Hum Genet 79, Dudley GA, Abraham WM & Terjung RL (1982). Influence of
469–480. exercise intensity and duration on biochemical adaptations
Bua EA, McKiernan SH, Wanagat J, McKenzie D & Aiken JM in skeletal muscle. J Appl Physiol 53, 844–850.
(2002). Mitochondrial abnormalities are more frequent in Egan B, Carson BP, Garcia-Roves PM, Chibalin AV, Sarsfield
muscles undergoing sarcopenia. J Appl Physiol 92, FM, Barron N, McCaffrey N, Moyna NM, Zierath JR &
2617–2624. O’Gorman DJ (2010). Exercise intensity-dependent
Burgomaster KA, Howarth KR, Phillips SM, Rakobowchuk M, regulation of peroxisome proliferator-activated receptor
Macdonald MJ, Mcgee SL & Gibala MJ (2008). Similar coactivator-1 mRNA abundance is associated with
metabolic adaptations during exercise after low volume differential activation of upstream signalling kinases in
sprint interval and traditional endurance training in human skeletal muscle. J Physiol 588, 1779–1790.
humans. J Physiol 5861, 151–160. Erlich AT, Brownlee DM, Beyfuss K & Hood DA (2018).
Calvo SE, Clauser KR & Mootha VK (2016). MitoCarta2.0: an Exercise induces TFEB expression and activity in skeletal
updated inventory of mammalian mitochondrial proteins. muscle in a PGC-1α-dependent manner. Am J Physiol Cell
Nucleic Acids Res 44, D1251–D1257. Physiol 314, C62–C72.
Carter HN, Chen CCW & Hood DA (2015). Mitochondria, Feng D, Zhao W-L, Ye Y-Y, Bai X-C, Liu R-Q, Chang L-F,
muscle health, and exercise with advancing age. Physiology Zhou Q & Sui S-F (2010). Cellular internalization of
30, 208–223. exosomes occurs through phagocytosis. Traffic 11, 675–687.
Carter HN, Kim Y, Erlich AT, Zarrin-khat D & Hood DA Fernández-Vizarra E, Enrı́quez JA, Pérez-Martos A, Montoya
(2018a). Autophagy and mitophagy flux in young and aged J & Fernández-Silva P (2011). Tissue-specific differences in
skeletal muscle following chronic contractile activity. mitochondrial activity and biogenesis. Mitochondrion 11,
J Physiol 596, 3567–3584. 207–213.
Carter HN, Pauly M, Tryon LD & Hood DA (2018b). Fiorese CJ, Schulz AM, Lin YF, Rosin N, Pellegrino MW &
Effect of contractile activity on PGC-1α transcription in Haynes CM (2016). The transcription factor ATF5 mediates
young and aged skeletal muscle. J Appl Physiol 124, a mammalian mitochondrial UPR. Curr Biol 26,
1605–1615. 2037–2043.
Chabi B, Ljubicic V, Menzies KJ, Huang JH, Saleem A & Hood Geisler S, Holmström KM, Skujat D, Fiesel FC, Rothfuss OC,
DA (2008). Mitochondrial function and apoptotic Kahle PJ & Springer W (2010). PINK1/Parkin-mediated
susceptibility in aging skeletal muscle. Aging Cell 7, mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat
2–12. Cell Biol 12, 119–131.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
814 J. M. Memme and others J Physiol 599.3

Gibala MJ, McGee SL, Garnham AP, Howlett KF, Snow RJ & Holloszy JO (1967). Biochemical adaptations in muscle. J Biol
Hargreaves M (2009). Brief intense interval exercise activates Chem 242, 2278–2282.
AMPK and p38 MAPK signaling and increases the Holloszy JO & Booth FW (1976). Biochemical adaptations to
expression of PGC-1α in human skeletal muscle. J Appl endurance exercise in muscle. Annu Rev Physiol 38,
Physiol 106, 929–934. 273–291.
Giudice J & Taylor JM (2017). Muscle as a paracrine and Holloway GP (2017). Nutrition and training influences on the
endocrine organ. Curr Opin Pharmacol 34, 49–55. regulation of mitochondrial adenosine diphosphate
Glancy B, Hartnell LM, Combs CA, Fenmou A, Sun J, Murphy sensitivity and bioenergetics. Sport Med 47, S13–S21.
E, Subramaniam S & Balaban RS (2017). Power grid Hood DA (2001). Invited Review: contractile activity-induced
protection of the muscle mitochondrial reticulum. Cell Rep mitochondrial biogenesis in skeletal muscle. J Appl Physiol
19, 487–496. 90, 1137–1157.
Gollnick PD, Piehl K & Saltin B (1974). Selective glycogen Hood DA (2009). Mechanisms of exercise-induced
depletion pattern in human muscle fibres after exercise of mitochondrial biogenesis in skeletal muscle. Appl Physiol
varying intensity and at varying pedalling rates. J Physiol Nutr Metab 34, 465–472.
241, 45–57. Hood DA, Memme JM, Oliveira AN & Triolo M (2019).
Gomes JLP, Fernandes T, Soci UPR, Silveira AC, Barretti DLM, Maintenance of skeletal muscle mitochondria in health,
Negrão CE & Oliveira EM (2017). Obesity downregulates exercise, and aging. Annu Rev Physiol 81, 19–41.
MicroRNA-126 inducing capillary rarefaction in skeletal Hood DA, Tryon LD, Carter HN, Kim Y & Chen CCW (2016).
muscle: effects of aerobic exercise training. Oxid Med Cell Unravelling the mechanisms regulating muscle
Longev 2017, 2415246. mitochondrial biogenesis. Biochem J 473,
Gordon JW, Rungi AA, Inagaki H & Hood DA (2001). Effects 2295–2314.
of contractile activity on mitochondrial transcription factor Hood DA, Uguccioni G, Vainshtein A & D’souza D (2011).
A expression in skeletal muscle. J Appl Physiol 90, 389–396. Mechanisms of exercise-induced mitochondrial biogenesis
Gouspillou G, Sgarioto N, Kapchinsky S, Purves-Smith F, in skeletal muscle: implications for health and disease.
Norris B, Pion CH, Barbat-Artigas S, Lemieux F, Taivassalo Compr Physiol 1, 1119–1134.
T, Morais JA, Aubertin-Leheudre M & Hepple RT (2014). Hoppeler H, Lüthi P, Claassen H, Weibel ER & Howald H
Increased sensitivity to mitochondrial permeability (1973). The ultrastructure of the normal human skeletal
transition and myonuclear translocation of endonuclease G muscle. Pflügers Arch 344, 217–232.
in atrophied muscle of physically active older humans. Howald H, Hoppeler H, Claassen H, Mathieu O & Straub R
FASEB J 28, 1621–1633. (1985). Influences of endurance training on the
Groennebaek T & Vissing K (2017). Impact of resistance ultrastructural composition of the different muscle fiber
training on skeletal muscle mitochondrial biogenesis, types in humans. Pflügers Arch 403, 369–376.
content, and function. Front Physiol 8, 713. Ibebunjo C, Chick JM, Kendall T, Eash JK, Li C, Zhang Y,
Guan Y, Drake JC & Yan Z (2019). Exercise-induced mitophagy Vickers C, Wu Z, Clarke BA, Shi J, Cruz J, Fournier B,
in skeletal muscle and heart. Exerc Sport Sci Rev 47, 151–156. Brachat S, Gutzwiller S, Ma Q, Markovits J, Broome M,
Gusdon AM, Callio J, Distefano G, O’Doherty RM, Goodpaster Steinkrauss M, Skuba E, Galarneau JR, Gygi SP & Glass DJ
BH, Coen PM & Chu CT (2017). Exercise increases (2013). Genomic and proteomic profiling reveals reduced
mitochondrial complex I activity and DRP1 expression in mitochondrial function and disruption of the
the brains of aged mice. Exp Gerontol 90, 1–13. neuromuscular junction driving rat sarcopenia. Mol Cell Biol
Handschin C, Rhee J, Lin J, Tarr PT & Spiegelman BM (2003). 33, 194–212.
An autoregulatory loop controls peroxisome Iqbal S, Ostojic O, Singh K, Joseph A-M & Hood DA (2013).
proliferator-activated receptor gamma coactivator 1alpha Expression of mitochondrial fission and fusion regulatory
expression in muscle. Proc Natl Acad Sci U S A 100, proteins in skeletal muscle during chronic use and disuse.
7111–7116. Muscle Nerve 48, 963–970.
Handschin C & Spiegelman BM (2006). Peroxisome Irrcher I, Ljubicic V & Hood DA (2009). Interactions between
proliferator-activated receptor γ coactivator 1 coactivators, ROS and AMP kinase activity in the regulation of
energy homeostasis, and metabolism. Endocr Rev 27, PGC-1alpha transcription in skeletal muscle cells. Am J
728–735. Physiol Cell Physiol 296, C116–C123.
Harbauer AB, Zahedi RP, Sickmann A, Pfanner N & Meisinger Jacobs RA & Lundby C (2013). Mitochondria express enhanced
C (2014). The protein import machinery of mitochondria-a quality as well as quantity in association with aerobic fitness
regulatory hub in metabolism, stress, and disease. Cell Metab across recreationally active individuals up to elite athletes.
19, 357–372. J Appl Physiol 114, 344–350.
Heber D, Ingles S, Ashley JM, Maxwell MH, Lyons RF & Joseph A-M, Adhihetty PJ, Buford TW, Wohlgemuth SE, Lees
Elashoff RM (1996). Clinical detection of sarcopenic obesity HA, Nguyen LM-D, Aranda JM, Sandesara BD, Pahor M,
by bioelectrical impedance analysis. Am J Clin Nutr 64, Manini TM, Marzetti E & Leeuwenburgh C (2012). The
472S–477S. impact of aging on mitochondrial function and biogenesis
Hoffmann C & Weigert C (2017). Skeletal muscle as an pathways in skeletal muscle of sedentary high- and
endocrine organ: the role of myokines in exercise low-functioning elderly individuals. Aging Cell 11,
adaptations. Cold Spring Harb Perspect Med 7, a029793. 801–809.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 815

Ju JS, Jeon SI, Park JY, Lee JY, Lee SC, Cho KJ & Jeong JM Lira VA, Okutsu M, Zhang M, Greene NP, Laker RC, Breen DS,
(2016). Autophagy plays a role in skeletal muscle Hoehn KL & Yan Z (2013). Autophagy is required for
mitochondrial biogenesis in an endurance exercise-trained exercise training-induced skeletal muscle adaptation and
condition. J Physiol Sci 66, 417–430. improvement of physical performance. FASEB J 27,
Kayar SR, Hoppeler H, Mermod L & Weibel ER (1988). 4184–4193.
Mitochondrial size and shape in equine skeletal muscle: a Little JP, Safdar A, Bishop D, Tarnopolsky MA & Gibala MJ
three-dimensional reconstruction study. Anat Rec 222, (2011). An acute bout of high-intensity interval training
333–339. increases the nuclear abundance of PGC-1α and activates
Kim H-J & Song W (2017). Resistance training increases mitochondrial biogenesis in human skeletal muscle. Am J
fibroblast growth factor-21 and irisin levels in the skeletal Physiol Regul Integr Comp Physiol 300, R1303–R1310.
muscle of Zucker diabetic fatty rats. J Exerc Nutr Biochem 21, Liu D, Sartor MA, Nader GA, Pistilli EE, Tanton L, Lilly C,
50–54. Gutmann L, IglayReger HB, Visich PS, Hoffman EP &
Kim J, Kundu M, Viollet B & Guan K-L (2014). AMPK and Gordon PM (2013). Microarray analysis reveals novel
mTOR regualte autophagy through direct phosphorylation features of the muscle aging process in men and women. J
of Ulk1. Nat Cell Biol 13, 132–141. Gerontol A Biol Sci Med Sci 68, 1035–1044.
Kim Y & Hood DA (2017). Regulation of the autophagy system Ljubicic V & Hood DA (2009). Diminished
during chronic contractile activity-induced muscle contraction-induced intracellular signaling towards
adaptations. Physiol Rep 5, e13307. mitochondrial biogenesis in aged skeletal muscle. Aging Cell
Kim Y, Triolo M, Erlich AT & Hood DA (2018). Regulation of 8, 394–404.
autophagic and mitophagic flux during chronic contractile Ljubicic V, Joseph A-M, Adhihetty PJ, Huang JH, Saleem A,
activity-induced muscle adaptations. Pflügers Arch 471, Uguccioni G & Hood DA (2009). Molecular basis for an
431–440. attenuated mitochondrial adaptive plasticity in aged skeletal
Kirkwood SP, Munn EA & Brooks GA (1986). Mitochondrial muscle. Aging (Albany NY) 1, 818–830.
reticulum in limb skeletal muscle. Am J Physiol Cell Physiol López-Otı́n C, Blasco MA, Partridge L, Serrano M & Kroemer
251, C395–C402. G (2013). The hallmarks of aging. Cell 153, 1194–1217.
Koltai E, Hart N, Taylor AW, Goto S, Ngo JK, Davies KJA & Losón OC, Song Z, Chen H & Chan DC (2013). Fis1, Mff,
Radak Z (2012). Age-associated declines in mitochondrial MiD49, and MiD51 mediate Drp1 recruitment in
biogenesis and protein quality control factors are minimized mitochondrial fission. Mol Biol Cell 24, 659–667.
by exercise training. Am J Physiol Integr Comp Physiol 303, Lu Y, Li H, Shen S-W, Shen Z-H, Xu M, Yang C-J, Li F, Feng
R127–R134. Y-B, Yun J-T, Wang L & Qi H-J (2016). Swimming exercise
Koo J-H, Kang E-B & Cho J-Y (2019). Resistance exercise increases serum irisin level and reduces body fat mass in
improves mitochondrial quality control in a rat model of high-fat-diet fed Wistar rats. Lipids Health Dis 15, 93.
sporadic inclusion body myositis. Gerontology 65, 240–252. Lundby C & Jacobs RA (2016). Adaptations of skeletal muscle
Kowald A & Kirkwood TBL (2014). Transcription could be the mitochondria to exercise training. Exp Physiol 101, 17–22.
key to the selection advantage of mitochondrial deletion Lüthi J, Howald H, Claassen H, Rösler K, Vock P & Hoppeler H
mutants in aging. Proc Natl Acad Sci U S A 111, (1986). Structural changes in skeletal muscle tissue with
2972–2977. heavy-resistance exercise. Int J Sports Med 07, 123–127.
Kujoth GC, Hiona A, Pugh TD, Someya S, Panzer K, MacInnis MJ & Gibala MJ (2017). Physiological adaptations to
Wohlgemuth SE, Hofer T, Seo AY, Sullivan R, Jobling WA, interval training and the role of exercise intensity. J Physiol
Morrow JD, Van Remmen H, Sedivy JM, Yamasoba T, 595, 2915–2930.
Tanokura M, Weindruch R, Leeuwenburgh C & Prolla TA Mansueto G, Armani A, Viscomi C, Zeviani M, Sandri M,
(2005). Mitochondrial DNA mutations, oxidative stress, and Ballabio A, Mansueto G, Armani A, Viscomi C, Orsi LD,
apoptosis in mammalian aging. Science 309, 481–484. Cegli R De, Polishchuk E V, Lamperti C, Di Meo I,
Laker RC, Drake JC, Wilson RJ, Lira VA, Lewellen BM, Ryall Romanello V, Marchet S, Saha PK & Zong H (2017).
KA, Fisher CC, Zhang M, Saucerman JJ, Goodyear LJ, Transcription factor EB controls metabolic flexibility during
Kundu M & Yan Z (2017). Ampk phosphorylation of Ulk1 is exercise article transcription factor EB controls metabolic
required for targeting of mitochondria to lysosomes in flexibility during exercise. Cell Metab 25, 182–196.
exercise-induced mitophagy. Nat Commun 8, 548. Marcinek DJ, Schenkman KA, Ciesielski WA, Lee D & Conley
Lanza IR, Short DK, Short KR, Raghavakaimal S, Basu R, Joyner KE (2005). Reduced mitochondrial coupling in vivo alters
MJ, McConnell JP & Nair KS (2008). Endurance exercise as a cellular energetics in aged mouse skeletal muscle. J Physiol
countermeasure for aging. Diabetes 57, 2933–2942. 569, 467–473.
Leduc-Gaudet J-P, Picard M, Pelletier FS-J, Sgarioto N, Auger Marosi K & Mattson MP (2014). BDNF mediates adaptive
M-J, Vallée J, Robitaille R, St-Pierre DH & Gouspillou G brain and body responses to energetic challenges. Trends
(2015). Mitochondrial morphology is altered in atrophied Endocrinol Metab 25, 89–98.
skeletal muscle of aged mice. Oncotarget 6, 17923–17937. Marques-Aleixo I, Santos-Alves E, Balça MM, Rizo-Roca D,
Leduc-Gaudet J-P, Reynaud O, Hussain SN & Gouspillou G Moreira PI, Oliveira PJ, Magalhães J & Ascensão A (2015).
(2019). Parkin overexpression protects from aging-related Physical exercise improves brain cortex and cerebellum
loss of muscle mass and strength. J Physiol 597, mitochondrial bioenergetics and alters apoptotic, dynamic
1975–1991. and auto(mito)phagy markers. Neuroscience 301, 480–495.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
816 J. M. Memme and others J Physiol 599.3

Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, Porter C, Reidy PT, Bhattarai N, Sidossis LS & Rasmussen BB
Sou Y-S, Saiki S, Kawajiri S, Sato F, Kimura M, Komatsu M, (2015). Resistance exercise training alters mitochondrial
Hattori N & Tanaka K (2010). PINK1 stabilized by function in human skeletal muscle. Hum Skelet Muscle Med
mitochondrial depolarization recruits Parkin to damaged Sci Sport Exerc 47, 1922–1931.
mitochondria and activates latent Parkin for mitophagy. Puigserver P, Adelmant G, Wu Z, Fan M, Xu J, O’malley B &
J Cell Biol 189, 211–221. Spiegelman BM (1999). Activation of PPAR coactivator-1
Melber A & Haynes CM (2018). UPR mt regulation and through transcription factor docking. Science 286,
output: a stress response mediated by mitochondrial-nuclear 1368–1371.
communication. Cell Res 28, 281–295. Puigserver P, Rhee J, Lin J, Wu Z, Yoon JC, Zhang CY, Krauss S,
Melov S, Tarnopolsky MA, Beckman K, Felkey K & Hubbard A Mootha VK, Lowell BB & Spiegelman BM (2001).
(2007). Resistance exercise reverses aging in human skeletal Cytokine stimulation of energy expenditure through p38
muscle. PLoS One 2, e465. MAP kinase activation of PPARγ coactivator-1. Mol Cell 8,
Memme JM, Oliveira AN & Hood DA (2016). Chronology of 971–982.
UPR activation in skeletal muscle adaptations to chronic Radak Z, Suzuki K, Higuchi M, Balogh L, Boldogh I & Koltai E
contractile activity. Am J Physiol Cell Physiol 310, (2016). Physical exercise, reactive oxygen species and
C1024–C1036. neuroprotection. Free Radic Biol Med 98, 187–196.
Mesbah Moosavi ZS & Hood DA (2017). The unfolded Rapizzi E, Pinton P, Szabadkai G, Wieckowski MR,
protein response in relation to mitochondrial biogenesis in Vandecasteele G, Baird G, Tuft RA, Fogarty KE & Rizzuto R
skeletal muscle cells. Am J Physiol Cell Physiol 312, (2002). Recombinant expression of the voltage-dependent
C583–C594. anion channel enhances the transfer of Ca2+ microdomains
Mishra P & Chan DC (2016). Metabolic regulation of to mitochondria. J Cell Biol 159, 613–624.
mitochondrial dynamics. J Cell Biol 212, 379–387. Ristow M, Zarse K, Oberbach A, Klöting N, Birringer M,
Mottis A, Jovaisaite V & Auwerx J (2014). The mitochondrial Kiehntopf M, Stumvoll M, Kahn CR & Blüher M (2009).
unfolded protein response in mammalian physiology. Antioxidants prevent health-promoting effects of physical
Mamm genome 25, 424–433. exercise in humans. Proc Natl Acad Sci U S A 106,
Navarro A, Gomez C, López-Cepero JM & Boveris A (2004). 8665–8670.
Beneficial effects of moderate exercise on mice aging: Rooyackers OE, Adey DB, Ades PA & Nair KS (1996). Effect of
survival, behavior, oxidative stress, and mitochondrial age on in vivo rates of mitochondrial protein synthesis in
electron transfer. Am J Physiol Regul Integr Comp Physiol human skeletal muscle. Proc Natl Acad Sci U S A 93,
286, R505–R511. 15364–15369.
O’Leary MFN, Vainshtein A, Iqbal S, Ostojic O & Hood DA Rowan SL, Purves-Smith FM, Solbak NM & Hepple RT (2011).
(2013). Adaptive plasticity of autophagic proteins to Accumulation of severely atrophic myofibers marks the
denervation in aging skeletal muscle. Am J Physiol Cell acceleration of sarcopenia in slow and fast twitch muscles.
Physiol 304, C422–C430. Exp Gerontol 46, 660–669.
Ogata T & Yamasaki Y (1997). Ultra-high-resolution scanning Safdar A, Saleem A & Tarnopolsky MA (2016). The potential of
electron microscopy of mitochondria and sarcoplasmic endurance exercise-derived exosomes to treat metabolic
reticulum arrangement in human red, white, and diseases. Nat Rev Endocrinol 12, 504–517.
intermediate muscle fibers. Anat Rec 248, 214–223. Sale DG (1987). Influence of exercise and training on motor
Ojuka EO, Jones TE, Han D-H, Chen M & Holloszy JO unit activation. Exerc Sport Sci Rev 15, 95–151.
(2003). Raising Ca2+ in L6 myotubes mimics effects of Scarpulla RC (2011). Metabolic control of mitochondrial
exercise on mitochondrial biogenesis in muscle. FASEB J 17, biogenesis through the PGC-1 family regulatory network.
675–681. Biochim Biophys Acta 1813, 1269–1278.
Oliveira AN & Hood DA (2018). Effect of Tim23 knockdown in Scarpulla RC, Vega RB & Kelly DP (2012). Transcriptional
vivo on mitochondrial protein import and retrograde integration of mitochondrial biogenesis. Trends Endocrinol
signaling to the UPRmt in muscle. Am J Physiol Cell Physiol Metab 23, 459–466.
315, C516–C526. Schaap LA, Pluijm SMF, Deeg DJH & Visser M (2006).
Park J-S, Davis RL & Sue CM (2018). Mitochondrial Inflammatory markers and loss of muscle mass (sarcopenia)
dysfunction in Parkinson’s disease: new mechanistic insights and strength. Am J Med 119, 526.e9–526.e17.
and therapeutic perspectives. Curr Neurol Neurosci Rep 18, Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F,
21. Visvikis O, Huynh T, Carissimo A, Klisch TJ, Wollenberg
Pedersen BK & Febbraio MA (2012). Muscles, exercise and AC, Di Bernardo D, Irazoqui JE & Ballabio A (2013). TFEB
obesity: skeletal muscle as a secretory organ. Nat Rev controls cellular lipid metabolism through a
Endocrinol 8, 457–465. starvation-induced autoregulatory loop. Nat Cell Biol 15,
Picard M, Gentil BJ, McManus MJ, White K, St. Louis K, 647–658.
Gartside SE, Wallace DC & Turnbull DM (2013). Acute Shenton D, Smirnova JB, Selley JN, Carroll K, Hubbard SJ,
exercise remodels mitochondrial membrane interactions in Pavitt GD, Ashe MP & Grant CM (2006). Global
mouse skeletal muscle. J Appl Physiol 115, 1562–1571. translational responses to oxidative stress impact upon
Picard M, Wallace DC & Burelle Y (2016). The rise of multiple levels of protein synthesis. J Biol Chem 281,
mitochondria in medicine. Mitochondrion 30, 105–116. 29011–29021.


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society
J Physiol 599.3 Exercise and mitochondrial health 817

Short KR, Bigelow ML, Kahl J, Singh R, Coenen-Schimke J, Wei H, Liu L & Chen Q (2015). Selective removal of
Raghavakaimal S & Nair KS (2005). Decline in skeletal mitochondria via mitophagy: distinct pathways for different
muscle mitochondrial function with aging in humans. Proc mitochondrial stresses. Biochim Biophys Acta 1853,
Natl Acad Sci U S A 102, 5618–5623. 2784–2790.
Short KR, Vittone JL, Bigelow ML, Proctor DN, Rizza RA, Whitham M, Parker BL, Friedrichsen M, Hingst JR, Hjorth M,
Coenen-Schimke JM & Nair KS (2003). Impact of aerobic Hughes WE, Egan CL, Cron L, Watt KI, Kuchel RP,
exercise training on age-related changes in insulin sensitivity Jayasooriah N, Estevez E, Petzold T, Suter CM, Gregorevic P,
and muscle oxidative capacity. Diabetes 52, 1888–1896. Kiens B, Richter EA, James DE, Wojtaszewski JFP &
Spinelli JB & Haigis MC (2018). The multifaceted Febbraio MA (2018). Extracellular vesicles provide a means
contributions of mitochondria to cellular metabolism. Nat for tissue crosstalk during exercise. Cell Metab 27,
Cell Biol 20, 745–754. 237–251.
Taivassalo T (1999). Gene shifting: a novel therapy for Winder WW, Holmes BF, Rubink DS, Jensen EB, Chen M &
mitochondrial myopathy. Hum Mol Genet 8, 1047–1052. Holloszy JO (2000). Activation of AMP-activated protein
Tanida I, Ueno T & Kominami E (2004). LC3 conjugation kinase increases mitochondrial enzymes in skeletal muscle.
system in mammalian autophagy. Int J Biochem Cell Biol 36, J Appl Physiol 88, 2219–2226.
2503–2518. Yang M, Liu E, Tang L, Lei Y, Sun X, Hu J, Dong H, Yang SM,
Tian W, Li W, Chen Y, Yan Z, Huang X, Zhuang H, Zhong W, Gao M & Tang B (2018). Emerging roles and regulation of
Chen Y, Wu W, Lin C, Chen H, Hou X, Zhang L, Sui S, Zhao MiT/TFE transcriptional factors. Cell Commun Signal 16,
B, Hu Z, Li L & Feng D (2015). Phosphorylation of ULK1 by 1–11.
AMPK regulates translocation of ULK1 to mitochondria and
mitophagy. FEBS Lett 589, 1847–1854.
Toyama EQ, Herzig S, Courchet J, Lewis TL, Losón OC,
Hellberg K, Young NP, Chen H, Polleux F, Chan DC & Shaw
RJ (2016). Metabolism. AMP-activated protein kinase Additional information
mediates mitochondrial fission in response to energy stress.
Competing interests
Science 351, 275–281.
Triolo M & Hood DA (2019). Mitochondrial breakdown in The authors have no competing interests to declare.
skeletal muscle and the emerging role of the lysosomes. Arch
Biochem Biophys 661, 66–73.
Trovato E, Di Felice V & Barone R (2019). Extracellular vesicles:
delivery vehicles of myokines. Front Physiol 10, 522. Author contributions
Vainshtein A & Hood DA (2016). The regulation of autophagy All authors contributed to writing the manuscript. J.M.M.
during exercise in skeletal muscle. J Appl Physiol 120, and D.A.H. conceived of the review, identified and inter-
664–673. preted relevant studies for inclusion, and critically revised the
Vainshtein A, Tryon LD, Pauly M & Hood DA (2015). Role of manuscript. All authors approved of the final manuscript and
PGC-1α during acute exercise-induced autophagy and agreed to be accountable for all aspects of the work. All persons
mitophagy in skeletal muscle. Am J Physiol Cell Physiol 308,
designated as authors qualify for authorship, and all those who
C710–C719.
qualify for authorship are listed.
Vecchio LM, Meng Y, Xhima K, Lipsman N, Hamani C &
Aubert I (2018). The neuroprotective effects of exercise:
maintaining a healthy brain throughout aging. Brain Plast 4,
17–52. Funding
Vincent AE, Turnbull DM, Eisner V, Hajnóczky G & Picard M
(2017). Mitochondrial nanotunnels. Trends Cell Biol 27, Our laboratory is funded by an operating grant from the Natural
787–799. Sciences and Engineering Research Council of Canada (NSERC;
Vincent AE, White K, Davey T, Philips J, Ogden RT, Lawess C, grant number 38462) to D.A.H. and D.A.H. is a Tier 1 NSERC
Warren C, Hall MG, Ng YS, Falkous G, Holden T, Deehan D, Canada Research Chair in Cell Physiology. J.M.M. is funded by
Taylor RW, Turnbull DM & Picard M (2019). Quantitative a NSERC CGS-D.
3D mapping of the human skeletal muscle mitochondrial
network. Cell Rep 26, 996–1009.e4.
Wanagat J, Cao Z, Pathare P & Aiken JM (2001). Mitochondrial Keywords
DNA deletion mutations colocalize with segmental electron
transport system abnormalities, muscle fiber atrophy, fiber ageing, exercise training, lysosomal biogenesis, mitochondrial
splitting, and oxidative damage in sarcopenia. FASEB J 15, biogenesis, mitochondrial quality control, mitophagy, skeletal
322–332. muscle, UPRmt


C 2019 The Authors. The Journal of Physiology 
C 2019 The Physiological Society

You might also like