Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

BBA - Molecular Cell Research 1867 (2020) 118742

Contents lists available at ScienceDirect

BBA - Molecular Cell Research


journal homepage: www.elsevier.com/locate/bbamcr

Review

Role of damage and management in muscle hypertrophy: Different T


behaviors of muscle stem cells in regeneration and hypertrophy

So-ichiro Fukadaa, , Takayuki Akimotob, Athanassia Sotiropoulosc
a
Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
b
Faculty of Sport Sciences, Waseda University, Saitama, Japan
c
Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France

A R T I C LE I N FO A B S T R A C T

Keywords: Skeletal muscle is a dynamic tissue with two unique abilities; one is its excellent regenerative ability, due to the
Skeletal muscle activity of skeletal muscle–resident stem cells named muscle satellite cells (MuSCs); and the other is the
Muscle satellite cells adaptation of myofiber size in response to external stimulation, intrinsic factors, or physical activity, which is
Hypertrophy known as plasticity. Low physical activity and some disease conditions lead to the reduction of myofiber size,
Regeneration
called atrophy, whereas hypertrophy refers to the increase in myofiber size induced by high physical activity or
Damages
anabolic hormones/drugs. MuSCs are essential for generating new myofibers during regeneration and the in-
crease in new myonuclei during hypertrophy; however, there has been little investigation of the molecular
mechanisms underlying MuSC activation, proliferation, and differentiation during hypertrophy compared to
those of regeneration. One reason is that ‘degenerative damage’ to myofibers during muscle injury or upon
hypertrophy (especially overloaded muscle) is believed to trigger similar activation/proliferation of MuSCs.
However, evidence suggests that degenerative damage of myofibers is not necessary for MuSC activation/pro-
liferation during hypertrophy. When considering MuSC-based therapy for atrophy, including sarcopenia, it will
be indispensable to elucidate MuSC behaviors in muscles that exhibit non-degenerative damage, because de-
generated myofibers are not present in the atrophied muscles. In this review, we summarize recent findings
concerning the relationship between MuSCs and hypertrophy, and discuss what remains to be discovered to
inform the development and application of relevant treatments for muscle atrophy.

1. Introduction studies of regenerating muscle, the behaviors and molecular mechan-


isms underlying the activation and proliferation of MuSCs during hy-
Skeletal muscle is composed of terminally differentiated and mul- pertrophy remains to be elucidated.
tinucleated giant cells, called myofibers. In mammals, myofibers are It is assumed that MuSCs in overloaded muscle respond to damaged
postmitotic and cannot give rise to new myofibers. Instead, skeletal myofibers and repair them in a similar way to that observed during
muscle possesses mononuclear stem cells, or muscle satellite cells regeneration [6]. It is well documented that eccentric muscle contrac-
(MuSCs) [1], that are responsible for generating new myofibers [2,3]. tion damages skeletal muscles and the history of exercise-induced
Myofibers possess another ability, known as plasticity [4,5]. Myofibers muscle damage in humans has been summarized previously [7]. As
adjust their size in response to external stimulations, intrinsic factors, or described by Damas et al. (2018), the term muscle ‘damage’ is con-
physical activity. Bedridden people, patients with cancer cachexia or troversial and includes several physiological events, such as disturbance
sepsis, and the elderly exhibit a loss of muscle mass (i.e., muscle of the myofibril structure, decreased stability of the sarcolemma, and
atrophy), whereas resistance training and some kinds of drugs leads to lethal damage to myofibers. In this review, we use the term degen-
an increase in muscle mass and function (i.e., muscle hypertrophy). erative damage to refer to damages causing myofiber death. There is
Regarding hypertrophy, many studies indicate that proliferating MuSCs direct evidence demonstrating the existences of disrupted myofibril
contribute to an increase in muscle size through fusion to the growing structure, even in human studies [8], whereas such evidence regarding
myofiber and the addition of new myonuclei. However, compared to degenerative damage of myofibers in human muscle is rare. Yu et al.


Corresponding author at: Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita City, Osaka
565-0871, Japan.
E-mail address: fukada@phs.osaka-u.ac.jp (S.-i. Fukada).

https://doi.org/10.1016/j.bbamcr.2020.118742
Received 2 March 2020; Received in revised form 7 May 2020; Accepted 12 May 2020
Available online 14 May 2020
0167-4889/ © 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

(2002) reported that eccentric contraction did not cause degenerative the production of new myofibers during muscle regeneration [2,3].
damage to myofibers in the soleus or vastus lateralis muscles of male Although some investigations indicate that MuSCs exhibit multipotent
subjects (age: 18–39 years old) experiencing delayed onset muscle differentiation abilities (for example, MuSCs differentiate into fibro-
soreness (DOMS) [9]. On the other hand, Crameri et al. (2004) reported blastic cells and brown adipocytes in certain disease conditions, ge-
that one in eight male subjects (age: 25 ± 3 years) demonstrated signs netically modified mice, aged mice, and in vitro) [21–24], this ability,
of degenerative damage to myofibers in the vastus lateralis muscle after in normal MuSCs, is limited to the myogenic lineage [25,26]. Based on
a single bout of high intensity exercise [10]. Because a muscle biopsy previous knowledge, MuSCs are more primed for myogenic lineage by
only allows for the observation of a very small section of skeletal muscle retaining transcripts of the myogenic determination genes, MyoD and
[7], the direct assessment of the degenerative damage to myofibers in myogenic factor 5 (Myf5), without subsequent translation thereof
human muscle is difficult. Crameri et al. (2007) evidenced that artificial [27,28]. The expression of Myf5 is also reported in quiescent MuSCs
electrical stimulation induced degenerative damage to myofibers that [29]. In skeletal muscle, muscle-resident mesenchymal progenitors,
could be detected histologically, whereas voluntary eccentric contrac- also known as fibro-adipogenic progenitors (FAPs), are considered the
tion did not induce such damage [11]. Using an electrical stimulation major source of fibrosis and fat accumulation in both humans and mice
model of human muscle regeneration, Mackey and Kjaer (2017) were [22,25,26,30,31].
able to efficiently observe degenerative damage of myofibers [12]. In the past decade, molecular mechanisms underlying the quiescent
Hence, there is little direct evidence to confirm that exercise induces and undifferentiated state of MuSCs have been discovered [32–35]. The
degenerative damage to myofibers. canonical Notch signaling pathway is essential in sustaining the un-
Instead of histological studies, indirect assessments, including differentiated state of MuSCs via direct target genes, namely Hey1,
magnetic resonance imaging (MRI), have generally been used to detect HeyL, Hes1, collagen V, and mir-708 [36–42]. Intriguingly, collagen V
degenerative damage, but an MRI also reflects edema (swelling caused operates as a surrogate ligand for calcitonin receptor (CalcR), which
by excess fluid) in skeletal muscle. Increased levels of muscle proteins, represents another signaling pathway for maintaining MuSC quiescence
such as creatine kinase and myoglobin, in the blood are also widely [36,43–45]. Recently, we revealed that the CalcR-protein kinase A
used as markers of degenerative damage [13]. However, these markers (PKA) axis regulates Hippo pathway members, large tumor-suppressor
might not accurately reflect the death of myofibers because myofibers kinases 1/2 (Lats1/2), and Yes-associated protein 1 (Yap1) to sustain
possess repair systems, independent of MuSC fusion, that seal the da- quiescence in MuSCs [46]. The quiescent MuSC-specific microRNA,
maged sarcolemma [14,15]. In other words, it is possible that muscle mir-489, is encoded by intron 4 of the mouse Calcr gene and is also
proteins leaked from the damaged sarcolemma during the membrane important for maintaining quiescence by targeting the oncogene Dek
repair process. There is no doubt that exercise can induce degenerative [47].
damage of myofibers [16], but, to our knowledge, there is no direct Endothelial cells are thought to be important in maintaining MuSC
evidence disclosing that the degenerative damage of myofibers is re- quiescence because MuSCs are approximal to endothelial cells [48].
quired for MuSC activation/proliferation during hypertrophy. Darr and Verma et al. (2018) also exhibited that MuSCs and endothelial cells are
Schultz (1987) observed BrdU+ (proliferating) MuSCs in overloaded rat in close proximity and a positive relationship exists between the
muscle without degenerative damage to myofibers [17]. Blood flow quiescent state and distance between the latter and former. They also
restriction (BFR) alone, or in combination with exercise, results in evidenced that Notch ligand Dll4, expressed in endothelial cells, in-
muscle hypertrophy and strength gain. Beyond the effects of training, duces quiescence during MuSC self-renewal [49]. It is predicted that
the degenerative damage of myofibers and subsequent MuSC-depen- myofibers also supply Notch ligands to activate canonical Notch sig-
dent repair is one proposed mechanism. However, Loenneke et al. naling in MuSCs [34]. The relationship between myofiber- and en-
(2014) questioned whether the degenerative damage of myofibers is a dothelial cell–derived Notch ligands needs to be clarified to decipher
necessary effect of training, because available evidence does not sup- the role of Notch signaling in MuSC regulation. Myofibers play another
port the hypothesis that BFR in combination with low-intensity exercise role in MuSC maintenance by establishing direct cell contact with
increases the incidence of degenerative damage to myofibers [18]. MuSCs through N- and M-cadherin [50]. Additionally, myofiber-de-
Recently, we showed that MuSCs proliferate on living myofibers in rived Wnt4 is reported to maintain MuSCs quiescence by inhibiting
mouse models of tenotomy and overload [19]. These results imply that Yap1 transcription [51]. Moreover, Yap1 activity can also be regulated
the degenerative damage of myofibers is not necessary for MuSC acti- by cell-cell contact between myofibers and MuSCs, which suggests that
vation and proliferation in overloaded muscle. In addition, it is assumed MuSCs can employ at least three pathways to regulate Yap1 activity in
that the environments for the activation and proliferation of MuSCs in order to sustain quiescence (Fig. 1).
regenerating and overloaded muscle are completely different [19]. Mesenchymal progenitors are another cell type thought to
When considering appropriate therapy for diseases resulting in muscle
atrophy, the elucidation of the mechanisms underlying MuSC activa-
tion/proliferation in non-injured muscle is essential, because they do
not accompany the degenerative damage of myofibers. In this review,
the molecular mechanisms regulating MuSC quiescence in steady-state
conditions are updated and recent findings regarding the relationship
between MuSCs and hypertrophy, especially whether the degenerative
damage of myofibers is necessary for the activation/proliferation of
MuSCs, are summarized. Finally, we will discuss how to apply these
findings to the treatment of muscular atrophy.

2. Muscle stem cells under steady-state conditions

MuSCs are mononuclear cells located beneath the basal lamina of


mature skeletal muscle fibers [1]. In steady-state conditions, MuSCs are
maintained in a quiescent and undifferentiated state. However, when Fig. 1. Molecular mechanisms underlying quiescence in MuSCs.
myofibers are damaged and degeneration occurs, MuSCs start to ex- Three putative molecular pathways regulate Yap1 activity in quiescent MuSCs.
press myoblast determination protein 1 (MyoD) and subsequently enter Fzd (Frizzled), Ror1/2, and Vangl1/2 are putative receptor and co-receptors for
the cell cycle [20]. There is no doubt that MuSCs are indispensable in Wnt4.

2
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

contribute to the maintenance of MuSCs in normal muscle tissue, be- swelling, especially in the early phase shortly after surgery.
cause the depletion of mesenchymal progenitors resulted in the loss of Although both surgical procedures are equally invasive and may be
MuSC pool in a mouse model [52]. Compared to cell-autonomous and attributed to the degenerative damage of myofibers, the SA model has a
other niche cells' regulation of MuSCs, the molecular mechanisms un- higher risk of degenerative damage compared to the tenotomy model
derlying the role of mesenchymal progenitors in the maintenance of (see Section 3.3). Both models are commonly used for the analyses of
MuSCs are unknown. The identification of these factors will lead to a MuSCs behaviors.
more in-depth understanding of MuSC quiescence, which will con-
tribute to the explanation of MuSC behaviors in regenerating and 3.1.2. Voluntary weightlifting exercise
overloaded muscle. The surgical procedures described above have a risk of causing
degeneration or inflammation in muscles. Klitgaard et al. (1988) de-
3. Muscle hypertrophy veloped a non-invasive, voluntary weightlifting rat model, which evi-
denced a significant increase in plantar flexor muscle weight and
Muscle hypertrophy, in response to intrinsic and extrinsic factors, is strength after 36 weeks of training [56]. Because this model requires
a characteristic of skeletal muscle plasticity that is even observed in constant human intervention, it has not been widely used in the re-
Drosophila (fruit flies) [53]. This means that hypertrophy is a capacity search field. Considering this shortfall, Cui et al. (2020) developed a
that is evolutionarily conserved in skeletal muscle, like its regenerative novel mouse model of voluntary weightlifting that does not involve
ability [54]. In order to identify the mechanisms underlying the in- human handling during the training [57]. In order to access food, mice
crease in myofiber size, many studies have focused on the roles of are required to push a weighted cage which leads to voluntary hindlimb
MuSCs in several kinds of animal models. However, whether the de- plantar flexion movement against shoulder-loaded resistance. Com-
generative damage of myofibers is necessary for MuSC activation/ pared to surgical models, the degree of muscle hypertrophy in the study
proliferation during muscle hypertrophy has not been sufficiently de- is modest, but highly consistent with that observed in humans
bated, and the molecular mechanisms regulating MuSC activation, (8%–12%) [58]. This type of model is suitable for the study of MuSCs
proliferation, and differentiation have not been identified. In the fol- behaviors, on a physiological level, in muscle hypertrophy during re-
lowing subsections, animal models used to investigate the role of sistance training.
MuSCs in muscle hypertrophy are introduced, and thereafter we discuss
whether degenerative damage to myofibers is required for MuSC acti- 3.1.3. Running equipment
vation and proliferation. Wheel running is an endurance training model that also leads to
muscle hypertrophy. Rodents are internally motivated to run sponta-
3.1. Animal models of muscle hypertrophy neously. In our analyses, the average running distance of a mouse is
6–10 km per night (unpublished data). A combined model, including
3.1.1. Surgical procedures increasing load and wheel running, was also developed to observe more
In rodent studies, synergistic ablation (SA) or tenotomy is widely remarkable adaptations of skeletal muscle [59,60]. The effect of vo-
used to induce muscle hypertrophy (Fig. 2). In both cases, reduced luntary wheel running alone on soleus wet mass was well observed, and
gastrocnemius/soleus muscle tension is often observed by targeting the the wet mass of additional muscles (plantaris, vastus lateralis, and ex-
plantaris muscle. The plantaris muscle receives all the tension following tensor carpi radialis longus and brevis) in rats increased in combination
surgery, resulting in overload hypertrophy. The difference between SA with load [61]. The plantaris and soleus muscles are both responsive to
and tenotomy is whether or not the synergistic muscles (gastrocnemius wheel running, but the soleus muscle does not undergo a shift in
and soleus muscles) are removed. In tenotomy, only the Achilles tendon myofiber type. The plantaris muscle, consisting mainly of fast type
is cut, whereas in SA, the gastrocnemius and soleus muscles are par- myofibers (30–40% Type IIa and 60–70% Type IIb + IIx), exhibits a
tially removed. The increase in plantaris muscle weight is attenuated shift toward slow-twitch oxidative myofibers (increased ratio of Type
2–3 weeks after tenotomy, relative to SA, but the muscle weight con- IIa and decreased ratio of Type IIb + IIx) following weighted wheel
tinues to increase for a longer period in the SA model [55]. Even for a running as well as tenotomy in mouse models [60]. Both the soleus and
short period, the ratio of muscle weight is also higher in the SA model plantaris muscles of mice and rats have been used extensively in the
than that of the tenotomy model. However, the increase in muscle analyses of MuSC behaviors in this model [62,63]. In mice, BrdU pulse
weight, or myofiber size, is considered to be a result of edema-induced experiments have detected the presence of BrdU+ cells between 3 and
muscle swelling, especially in the early phase of muscle hypertrophy, 14 days after the start of unweighted wheel running [63]. Basically,
which also occurs during human resistance training [13]. It is therefore many studies have indicated that degenerative damage to myofibers is
important to pay attention to the effects of edema-induced muscle the factor that induces MuSC proliferation [64].
Treadmill running is a forced exercise model for endurance training
in rodents. Because an eccentric contraction produces greater muscle
hypertrophy than a concentric contraction, downhill running is a
widely used method of eccentric training in both human and rodents.
Although muscles in humans and rodents differ due to varied gaits, one
advantage of the treadmill experiment is that it replicates the training
in humans, compared to the spontaneous wheel running model.
However, close attention must be paid to the negative effects of
treadmill running because alterations in neuroendocrine tissues and
immune responses, commonly associated with chronic stress, were
observed in male Sprague-Dawley rats [65]. In addition, many studies
indicated that downhill running injures the deep slow muscles of the
antigravity groups [66]. For example, Darr and Schultz (1987) showed
that downhill treadmill running induced myofiber degeneration that
was histologically characterized by the infiltration of mononuclear
cells, including macrophages, and necrotic myofibers in the rat soleus
Fig. 2. Two surgical models for inducing muscle hypertrophy in plantaris. muscle [17]. Moreover, the degenerative damage of myofibers was
Difference between the surgery of synergistic ablation and tenotomy. even observed in non-eccentric exercise training. McCormick and

3
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

Thomas (1992) demonstrated that 10 weeks of progressive treadmill myonuclei is required for hypertrophy. In a SA-induced model of
running induced MuSC proliferation with double the amount of de- muscle hypertrophy, both McCarthy et al. (2011) and Egner et al.
generative damage or repaired myofibers (defined morphologically (2016) commonly observed that MuSC-depleted mice showed no in-
using central nuclei or necrotic fibers) in the rat soleus muscle [67]. crease in the number of myonuclei during hypertrophy [78,79]. Goh
Umnova and Seene (1991) reported that the number of MuSCs in- et al. (2017) also detected that the number of myonuclei is reduced
creased by 2.5-fold in the quadriceps femoris of male rats with localized when MuSCs cannot fuse with myofibers due to a lack of myomaker, a
degenerative damage [68]. The frequency of the degenerative damage fusion protein [80]. Although a handful of studies have demonstrated
of myofibers depends on the intensity, duration, and frequency of ex- that other stem cells can contribute to myofiber regeneration under
ercise, and many studies that employed the treadmill model reported physiological conditions [81,82], there is no doubt that MuSCs are the
that MuSC activation/proliferation results from degenerative damage to most essential type of cell for supplying new myonuclei during over-
myofibers. Therefore, increased physical activity can induce degen- load-induced muscle hypertrophy. Whereas, McCarthy et al. (2011)
erative damage to myofibers. Except for the report by Darr and Schultz established a relatively short-term SA model and reported that myofi-
(1987) [17], there has been little discussion on the need for degen- bers lacking MuSCs grew similar to control myofibers, thereby con-
erative damage in MuSC activation/proliferation, whereas whether cluding that the addition of MuSC-derived myonuclei is not required for
damage, in the broadest sense of the word, is required for muscle hy- muscle hypertrophy [78]. On the contrary, after employing the same
pertrophy itself is a highly debated issue [69,70]. strategy, Egner et al. (2016) argued that MuSCs are critical for efficient
muscle hypertrophy [79]. Thus, whether MuSCs are required for muscle
3.1.4. Hormones and chemicals hypertrophy is a highly debated issue. Recently, independent studies
Androgens are a group of anabolic steroid hormone and generally have established different animal models, examining animals of dif-
refer to male sex hormones and substances with similar physiological ferent ages and for different time periods, and indicated that the loss of
effects. Natural male sex hormones include testosterone, dehy- MuSCs, or the impairment of their fusion capacity or proliferation, leads
drotestosterone (DHT), dehydroepiandrosterone (DHEA), and andros- to blunted muscle hypertrophy [19,55,80,83,84]. In the first 2–3 weeks
tenedione. It is well known that androgens have muscle strengthening following overload, the contribution of MuSCs might be minor or
effects. However, testosterone sensitivity not equally expressed in all blunted by some experimental conditions, including edema or the age
muscles. In rodents, the perineal muscles (the levator ani (LA) and of the mice [84]. However, in the late stages of overload, the efficiency
bulbocavernosus muscle) are very sensitive to testosterone [71]. Using of muscle hypertrophy can be blunted due to the myonuclear domain
myofiber-specific Cre mice (HSA-Cre), Chambon et al. (2010) reported being too large or the loss of MuSC paracrine factors [19,55,85]. Gen-
that the myofiber-androgen receptor (AR) transduces androgen-de- erally, both MuSCs-independent (e.g., mammalian target of rapamycin-
pendent postnatal fiber hypertrophy in the perineal muscles but not in (mTOR-) dependent protein synthesis) and MuSC-dependent (e.g.,
limb muscles. In addition, they also identified that myofiber-AR is es- myonuclear accretion) factors are required for efficient muscle hyper-
sential to generate maximum force of fast- and intermediate-twitch leg trophy (Fig. 3).
muscles by controlling the myofibrillar organization of androgen-in-
duced hypertrophic myofibers [72]. Regarding MuSCs, Sinha-Hikim
et al. (2003) indicated a correlation between the number of MuSCs and 3.2.2. Androgen
serum testosterone concentrations in humans who were administered Sinha-Hikim et al. (2002) indicated that testosterone-induced
weekly testosterone enanthate injections for 20 weeks [73]. Nnodim myofiber hypertrophy is associated with a dose-dependent increase in
(2001) reported that testosterone is responsible for denervation-in- myonuclear number [86], as well as an increased number of MuSCs
duced MuSC proliferation in the LA muscle because testosterone after testosterone administration in humans [73]. Using a mouse model,
treatment restored MuSC function in the denervated LA of castrated rats it was demonstrated that the administration of testosterone increased
[74]. Therefore, MuSCs are possible candidates for targeted therapy myonuclear number as a consequence of MuSC proliferation, because
with testosterone. An informative review regarding the regulation of such an increment was not observed in MuSC-depleted mice [87]. In
satellite cell function with androgens is summarized by Chen et al. fact, an increase in MuSC numbers was observed in the soleus (22%
(2005) [75]. When regarding MuSC quiescence, the mechanism indu- increase compared to the sham muscle) and plantaris muscle (29%) of
cing MuSC activation/proliferation can either be a consequence of
muscle hypertrophy, or the direct effect of androgens on quiescent
MuSCs to activate them. A study that investigated the effects of the
anabolic steroid, nandrolone decanoate, indicated that the use of this
steroid did not lead to myofiber damage in normal mice, although it
worsened symptoms of muscular dystrophy in mdx mice [76]. Thus,
MuSC activation/proliferation in an anabolic steroid model seems to be
independent of degenerative damage to myofibers.
The anabolic effects of beta2-agonists on skeletal muscle are also
well established and involve changes in protein kinetics which results in
muscle hypertrophy. Interestingly, the beta2-agonist, clenbuterol, in-
creased DNA and protein concentrations in denervation-induced muscle
atrophy, but had no impact on the DNA content in normal rat soleus
muscles [77], which suggests that the effects of clenbuterol on MuSCs
are limited to atrophied muscles.
In the following subsections, the proliferation of MuSCs in surgical
overload and testosterone models are discussed.

3.2. Roles of muscle stem cells in muscle hypertrophy Fig. 3. Loss of MuSC ability results in blunted muscle hypertrophy.
Both mTOR-dependent and MuSC-dependent mechanisms are critical for ef-
3.2.1. Surgical procedure fective muscle hypertrophy. The effect of MuSC might be obscured by some
In order to sustain the cytoplasm-to-myonucleus ratio (known as the experimental condition, but an increase in myonuclei is also important for ef-
myonuclear domain theory), the supply of new MuSC-derived ficient muscle hypertrophy.

4
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

6-month-old control mice harboring MuSCs, but was not the case in increased myofiber size or degenerative damage. Alternatively, the two
MuSC-depleted mice three weeks after testosterone administration. events are unrelated, and the signaling pathways that regulate both
However, the increase in myofiber cross-sectional area was not affected active and quiescent states stimulate MuSC activation and proliferation
by the depletion of MuSCs at least three weeks after testosterone and occur independently of myofiber growth. Questions remain re-
treatment [87]. Like the overload model, a longer period of study or garding how exercise and overload induce MuSC proliferation and fu-
muscle force measurement is required to conclude whether MuSCs are sion independently of hypertrophy. We discuss this point in Section 4.1.
necessary for testosterone-induced muscle hypertrophy. In addition, the
role of testosterone might be age-dependent, because it was reported 4. Comparison of muscle stem cell behaviors between muscle
that SA-induced muscle hypertrophy is dependent on MuSC accretion in hypertrophy and regeneration
young mice (below 4 months old), which is not the case in mature mice
(over 4 months old) [84]. During muscle regeneration, MuSCs proliferate beneath the retained
basal lamina, called ‘ghost fibers’ [97]. We have previously evidenced
3.3. Degenerative damage to myofibers and increased myofiber size are not that reduced myogenic regeneration occurs where ghost fibers are di-
necessary for muscle stem cell activation/proliferation minished (caused by injecting a fig protease, ficin, in combination with
cardiotoxin), which indicates that the retained basal lamina is critical
As described above, exercise-induced muscle hypertrophy is fre- for MuSCs behaviors during muscle regeneration [98]. On the other
quently accompanied by degenerative damage to myofibers in animal hand, it is assumed that MuSCs proliferate between the basal lamina
models, and such injuries may also be observed in animal models of and plasma membranes of myofibers in muscles experiencing hyper-
overload. Therefore, it is commonly believed that degenerative damage trophy and minimal degeneration (tenotomy model). This means the
to myofibers induces MuSC activation/proliferation in these models of environments for MuSC activation/proliferation and the kinetics of
muscle hypertrophy. Thus, whether degenerative damage is necessary MuSC behavior are completely different in regenerating and overloaded
for MuSC activation/proliferation has not been debated to a great ex- muscle (Fig. 4) [19]. In regenerating muscle, a stepwise process of
tent. However, if overload-induced muscle hypertrophy accompanies MuSC activation, proliferation, and differentiation proceeds, whereas
severe and extensive degenerative damage to myofibers, MuSC-de- MuSC proliferation and differentiation occur simultaneously, for rela-
pleted mice should exhibit more severe phenotypes because MuSCs are tively long periods, in tenotomy-overloaded muscle [19]. In this sec-
indispensable to the repair of damaged myofibers. This means that the tion, the behaviors of MuSCs are compared between regenerating and
area of degenerative damage is limited in the overload models. In fact, overloaded muscle (Fig. 4).
Peterson group evidenced that approximately 30% of myofibers stained
positive for embryonic myosin heavy chain (eMyHC; detects newly 4.1. Activation
generated myofibers) 14 days after SA [78], whereas 0.1–1.4% or
0.2–18.4% of myofibers were positive for eMyHC 10 days after te- Several models, including injury by freezing and barium chloride,
notomy in 2- or 4-month-old C57BL/6 mice, respectively [84]. Ad- notexin, and cardiotoxin injection, are used to observe muscle re-
ditionally, we previously implemented a tenotomy model to induce generation. Detailed analyses of multiple injury models are summarized
hypertrophy and demonstrated increased MuSC numbers with little by Hardy et al. (2016) [99]. Perhaps MuSCs start to express MyoD
degenerative damage to myofibers. Furthermore, almost all analyzed quickly in all regenerating muscles, and subsequently enter the cell
myofibers contained Ki67+ MuSCs [19]. Generally, the majority of cycle. In our previously established cardiotoxin model of muscle re-
MuSCs become activated and proliferate in overloaded live myofibers, generation, we observed that MuSCs first start to express MyoD, and at
regardless of the extent of degenerative damage, which means that least 50% of MuSCs are Ki67+ one day after injury [100]. In re-
degenerative damage to myofibers does not seem to be necessary for generating muscle, the components of the MuSC niche, including
MuSC activation and proliferation. myofibers, disappear. Therefore, it is conceivable that the loss of the
It is also important to distinguish whether muscle hypertrophy al- MuSC niche leads to reduced quiescence signaling, which is partially
ways follows an increase in myonuclear number because this will reveal responsible for the activation of MuSCs. In contrast, the MuSC niche is
the relevance between increased myofiber size and MuSC quiescence. relatively, or completely, retained in overloaded muscle. In our ana-
Inducible activation of Akt (also known as protein kinase B; PKB) sti- lyses, MuSCs start to express Ki67 two days after tenotomy [19], in-
mulates myofiber hypertrophy without an increase in myonuclear dicating that MuSC activation occurs later in overloaded muscle than in
number [88]. Importantly, the force generated by skeletal muscle was regenerating muscle. The factor that induces MuSC activation in over-
also increased by Akt activation. Furthermore, new MuSC-derived loaded muscle is unknown, but some potential mechanisms have been
myonuclei were not observed in JunB-induced muscle hypertrophy or identified. One possibility is that MuSCs perceive mechanical cues in a
myostatin-null mice [89,90]. These results suggest that signaling direct manner. Another possibility is that non-MuSCs, including myo-
pathways in myofiber growth, and increased myofiber size and strength fibers, sense the mechanical stress and subsequently release the acti-
do not affect MuSC quiescence and mobilization in the growing myo- vation factor which acts on MuSCs or disrupts their quiescent state.
fibers. Overload disrupts the extracellular matrix (ECM) and might initiate
Conversely, a considerable amount of data indicates that MuSC MuSC activation by releasing cytokines that were retained in the ECM
behaviors, independent of myofiber hypertrophy, are affected by ex- structure, or by reducing the amount of ligands available for quiescence
ercise or mechanical load [91]. In a human study using muscle biopsies signaling. In fact, the remodeling or disruption of the ECM (caused by
of endurance-trained cyclists, an increase in myonuclear number, in the the increased expression of some kinds of matrix metalloprotease
absence of hypertrophy or injury, was observed in response to an in- (MMP), tissue inhibitor of metalloproteinase (TIMP), and collagens)
tensified training load [91–93]. Similarly, a rat treadmill running and was observed three days after SA in rats [101]. However, because this
unweighted wheel running model reported an increase in the number of model includes the degenerative damage of myofibers, it is unclear to
MuSC-derived myonuclei, a lower incidence of histological disruption, what extent overload alone alters ECM remodeling without the added
and unchanged myofiber diameter [62,94]. It is also noteworthy that effects of degenerative damage to myofibers.
MuSC-dependent myonuclear accretion occurred in the absence of hy- Even if myofibers evade cell death, the effect of a damaged sarco-
pertrophy in aged mice subjected to SA surgery [95]. Moreover, a lemma on MuSC activation cannot be ignored. In a murine cancer ca-
human study in the elderly reported that MuSC proliferation and chexia model, the structures of the sarcolemma and ECM were per-
muscle hypertrophy occur independently of one another [96]. There- turbed by circulating cachectic factors, which led to MuSC activation
fore, MuSC activation and proliferation do not seem consequential of [102]. In addition, an increase in the myonuclear number was detected

5
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

Fig. 4. Behaviors of muscle stem cell in regenerating or overloaded muscle.


The upper cartoon summarizes of MuSC behaviors and time course during regeneration processes. The lower cartoon shows the kinetics and behaviors of MuSCs in
overloaded muscle.

in the absence of muscle injury in patients with rheumatoid arthritis cells, whereas the frequency of asymmetric division was 4.3% 3 days
(RA), a chronic inflammatory disease [103]. These results suggest that after injection [106]. Moreover, most MuSCs had proliferated on
damage to the structure of the sarcolemma or ECM triggers MuSC ac- myotubes and nascent myofibers 5 days after notexin injection.
tivation, even in healthy muscle hypertrophy. It could also be specu- Therefore, one can speculate that the presence of myofibers affects cell
lated that inflammation, including edema, directly induces MuSC acti- division. In muscle hypertrophy, MuSCs proliferate on existing myofi-
vation or indirectly disrupts MuSC quiescence by perturbing the bers, suggesting that asymmetric division frequently occurs to generate
structure of the sarcolemma or ECM. In order to determine the Pax7+MyoD− cells and myogenin+ differentiated cells in growing
minimum factors required for inducing MuSCs activation in hyper- myofibers.
trophy, the identification of factors, or genes, that activate MuSCs and In vitro, there are many cytokines that promote MuSC proliferation.
induce their proliferation in sedentary mice would be a challenging, yet Hepatocyte growth factor (HGF), basic fibroblast growth factor (bFGF),
fruitful experiment. and insulin-like growth factor (IGF) are candidates for MuSC pro-
liferation in muscle regeneration [109–111], but there is no direct
evidence indicating the physiological importance of these cytokines for
4.2. Proliferation MuSC proliferation in vivo. Unexpectedly, the depletion of the HGF
receptor, c-Met, did not affect MuSC proliferation in regenerating
Two to three days after injury, MyoD+ MuSCs proliferate vigorously muscle [112]. It could be speculated that the redundant roles of other
and begin to express myogenin to promote MuSC fusion. We previously cytokines might obscure the effect of the loss of HGF signaling in re-
reported that the majority of MuSCs proliferated in the absence of generating muscle. On the other hand, Zhu et al. (2016) indicated that
MyoD protein expression in overloaded muscle [19]. Notably, gene- signal transducer and activator of transcription 3 (Stat3) is physiolo-
expression analyses indicated that Hey1 expression was decreased in gically critical for MuSC proliferation in regenerating muscle [113].
proliferating MuSCs to the same extent as that observed in the injury Therefore, the cytokines regulating Stat3 activation (e.g., the inter-
model, whereas HeyL expression was sustained in MuSCs in overloaded leukin-6 (IL-6) family) might be essential for MuSC proliferation. Ev-
muscle. Besides HeyL, Notch target genes are highly expressed in erything considered, it could be predicted that several cytokines, in-
MuSCs in overloaded muscle compared to that of regenerating muscle cluding the activator of Stat3, play redundant roles in MuSC
[19]. Some studies have evidenced that the number of MyoD+ cells proliferation during regeneration.
increased in response to eccentric contraction in humans and a rat SA In overloaded muscle, IL-6 deficiency abrogated MuSC proliferation
model [104,105]. However, compared to Pax7+ cells, the number of and myonuclear accretion through decreased Stat3 activation, which
MyoD+ cells was minimal (< 10% or 30% of Pax7+ cells) [104], and indicates the importance of IL-6 in MuSCs behavior in overloaded
MyoD binding activity was not increased following exercise [105]. muscle [114]. Guerci et al. (2012) also found that myofiber-specific
Single cell RNA-sequence analyses will elucidate the fate of pro- depletion of serum response factor (SRF), a transcription factor, blunts
liferating MyoD− MuSCs, and more detailed analyses are required to overload-induced hypertrophy and reduces MuSC proliferation due to
conclude the existence of this MuSC population in overloaded human decreased IL-6 levels [115]. Interestingly, SRF in myofibers induces IL-6
muscle. expression in response to overload, which leads to MuSC proliferation
To study the proliferation of stem cells, including MuSCs, two [115]. These results suggest that SRF plays a role in the translation of
models, namely symmetric and asymmetric cell division, have been mechanical stimuli into paracrine signals and regulates MuSC pro-
proposed. In regenerating muscles, MuSCs proliferate either symme- liferation through IL-6. However, the molecular mechanism underlying
trically or asymmetrically both in vivo and in vitro [106–108]. Re- the transduction of mechanical stimuli in order to activate SRF in
cently, Evano et al. (2020) indicated that 5 days after notexin (a kind of overloaded muscle is still unknown. Whereas SRF activity is reduced in
snake toxin that differs from cardiotoxin) injection, about a quarter of atrophied muscle due to the altered subcellular localization of the
cell divisions were asymmetric and gave rise to Pax7+ and myogenin+

6
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

transcriptional co-activator called myocardin-related transcription 4.4. Macrophages and infiltration


factor (Mrtf) [116]. Interestingly, disuse atrophy induced the nuclear
accumulation of G-protein, which reduces SRF activity by inhibiting the During the regeneration processes, masses of inflammatory cells
nuclear accumulation of Mrtf [116]. Because Mrtf is involved in cardiac invade degenerated myofibers [131]. Within 12 h of muscle injury,
hypertrophy [117], it could be speculated that overload-dependent neutrophils infiltrate, and then major immune cells are switched to
hypertrophy in muscles also requires the nuclear location of Mrtf, macrophages [131]. The importance of inflammatory cells, especially
which is controlled by mechanical cue-induced actin dynamics for the macrophages, is well established [132–134]. The depletion of macro-
regulation of SRF transcriptional activity. In line with this hypothesis, phages results in delayed or impaired regeneration [133,134]. One
mechanical strain applied to myoblasts in culture was sufficient to in- important function of macrophages is the removal of degenerated
duce rapid actin polymerization and nuclear accumulation of Mrtf myofibers and debris. Like remodeling in other tissues, the contribu-
[118]. Another mechanism that bridges the gap between SRF activity tions of different macrophage subtypes are spatially and temporally
and mechanical cues is the activity of Yap/Taz, two molecules that are coordinated. In the early stages, pro-inflammatory (M1) macrophages,
well known mechanosensors [119]. In mammary epithelial and breast which secrete pro-inflammatory cytokines, play an essential role by
cancer cells, SRF interacts directly with Yap1 [120], suggesting that removing the debris. M2 macrophages, which exhibit an anti-in-
mechanical stimulation of the Yap1-SRF axis is important for overload- flammatory phenotype, support the myogenic differentiation processes,
induced hypertrophy in muscles. Collectively, growth factors important and macrophage skewing from the M1 toward the M2 phenotype is
in MuSC proliferation might be limited in overloaded muscle, and IL-6 important during muscle regeneration [135]. Supposing that hyper-
family-mediated Stat3 activation might be a common mechanism for trophy does not accompany the death of myofibers, macrophages re-
MuSC proliferation in both regenerating and overloaded muscle. sponsible for debris removal will not be needed. This means that,
However, the cellular source and expression mechanisms of cytokines compared to regenerating muscle, a limited subtype of macrophages
common to MuSC proliferation in overloaded muscle are likely to differ might contribute to muscle hypertrophy. Little is known regarding the
from those of regenerating muscle. contribution of macrophages to hypertrophy in overloaded muscle,
Another potential factor for MuSC proliferation is Silent mating type whereas the contribution of macrophages to the recovery from muscle
information regulation 2 homolog 1 (Sirt1)-dependent factors in myo- atrophy and the correlation between the number of macrophages and
fibers. Sirt1 is a sensitive modulator of metabolic processes, and reg- increased myofiber size in human-endurance exercise training, but not
ulates fat and glucose metabolism [121]. An increase in the expression resistance training, has been reported [136,137].
level of Sirt1 in mRNA and proteins was observed 2 weeks after SA In a murine study, treatment with the specific COX-2 inhibitor, NS-
surgery in a rat model [122]. In sedentary mice, the overexpression of 398, blunted the increase in mass and protein content without affecting
Sirt1 in adult myofibers accompanied an increase in myonuclear the Akt pathway in overloaded muscles compared to that of control
number while the MuSC pool size was sustained [123], suggesting that mice [138], suggesting that the inflammatory response is also necessary
MuSCs are self-renewed and supplied myonuclei by the increased ex- for muscle hypertrophy. However, this treatment might also affect
pression of Sirt1. These results also suggest that myofiber-derived fac- MuSC fusion processes because COX-2 regulates IL-4 expression in
tors promote proliferation of MuSCs regardless of degenerative damage overloaded myofibers [115]. Further study of macrophage depletion
to myofibers. will reveal the importance of macrophages in MuSC behaviors in
muscle hypertrophy.

4.3. Fusion 5. What remains to be discovered and possible applications in the


treatment of muscular disorders
In the regeneration processes, mononuclear cells first fuse with each
other, and then with the multinuclear myotubes. The fusion proteins, Muscular disorders can simply be classified into two categories:
myomaker and myomixer/myomerger/minion, are involved in the fu- diseases with degenerative damage to myofibers, such as muscular
sion processes. For myoblast-myoblast fusion, myomaker is required dystrophy, and diseases without such degenerative damage, such as
bilaterally (i.e., on both fusing cells) and myomixer/myomerger/ muscle atrophy. Muscular dystrophy patients exhibit the progressive
minion is required unilaterally (i.e., on only one cell) [124–129]. In loss of myofibers due to reduced regenerative ability, which is one
overloaded muscle, mononuclear cell-myofiber fusion occurs and potential factor for explaining the differences in pathological outcomes
MuSC-expressed myomaker is critical for this type of fusion [80], observed even when using animal models with the same kind of mu-
however it is not clear whether myomaker is also required on the tation. For example, there is a dramatic difference among mouse strains
myofiber membrane [128]. Different fusion mechanisms might exist [139] as C57BL/6 mice exhibit excellent muscle regenerative ability,
between myoblast-myoblast fusion and MuSC-myofiber fusion. whereas DBA/2 mice demonstrated a progressive loss of muscle weight
SRF, in the MuSCs, has been identified as a master regulator of in a model of Duchenne Muscular Dystrophy (DMD) [140–143]. These
MuSC fusion (in both fusion partners) and is required for the efficient results suggest that the augmentation of muscle regenerative ability
fusion of MuSCs to growing myofibers (in overloaded muscle) and with could provide a treatment option for muscular dystrophies. In this case,
each other in order to form newly regenerated myofibers [83]. Inter- the investigation of muscle regeneration processes, especially the
leukin 4 (IL-4) is reported to promote the fusion of myoblasts to nascent identification of potent growth-promoting factors for MuSCs, could lead
myotubes [130]. Guerci et al. (2012) evidenced that IL-4, derived from to the development of ‘regenerative medicine’ for treating muscular
myofibers, is critical for the fusion of MuSCs to myofibers in overloaded diseases accompanied by degenerative damage to myofibers.
muscle [115]. Collectively, these results imply that the critical cell fu- Considering the treatment of muscle atrophy, promoting muscle
sion mechanisms of regenerating and overloaded muscles are relatively regeneration is not an adequate strategy because cycles of myofiber
similar. degeneration and regeneration are not observed. Although the loss of
It is important to note, in a murine model of injury, the myonuclei in MuSCs did not affect myonuclear numbers during aging, changes in
regenerated myofibers were centrally located, but new MuSC-derived myonuclear shape and decreased numbers of myonuclei in the extensor
myonuclei in overloaded muscle are observed in peripheral locations digitorum longus (EDL) or tibialis anterior (TA) muscle, but not in the
[19,78,80]. It would be interesting to investigate the functional dif- plantaris muscle, in response to aging have been reported [144–146].
ference between central and peripheral nuclei derived from MuSCs. These results imply that MuSC myonuclear turnover does not occur
frequently in sedentary mice. The question remains whether myo-
nuclear number reduction is responsible for age-associated muscle

7
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

atrophy, especially in humans [147]. 7. Conclusion


When the quality and function is reduced in old myonuclei or
atrophied myofibers, the supplementation of new MuSC-derived myo- The importance of MuSCs in repairing degenerated myofibers is
nuclei is a potential therapeutic strategy for muscle atrophy. Given that unquestionable, but whether they are required for muscle hypertrophy
MuSC behaviors and regulatory mechanisms in overloaded muscle is debatable. In our opinion, the supplementation of new MuSC-derived
differ from those in regenerating muscle, we should learn from the myonuclei is critical for efficient muscle hypertrophy, and MuSC acti-
molecular mechanisms underlying MuSC activation/proliferation/fu- vation/proliferation is independent of mTOR/Akt signaling in myofi-
sion in overloaded muscle in order to accomplish ‘myonuclear replen- bers [88,115]. Mouse models (e.g., a beta-adrenergic agonist, Akt-tg
ishment therapy’ using MuSCs. Accordingly, Taylor-Weiner et al. mice, JunB, and myostatin-null) have evidenced muscle hypertrophy
(2020) used an in vitro culture system and showed that myonuclear without MuSC accretion which indicates that muscle hypertrophy is not
proteins, including transcriptional factors, can propagate to different always accompanied by increased myonuclear number. Therefore,
myonuclei in the same myofiber [148]. Furthermore, they evidenced MuSC activation/proliferation does not occur as a consequence of an
that muscle hypertrophy increased the transport of high molecular increase in myofiber size, whereas the abovementioned results indicate
weight nuclear proteins, while atrophy restricted the transport of that active signaling pathways exist and induce MuSC activation/pro-
smaller nuclear proteins. If propagation occurs similarly in vivo, studies liferation in overloaded uninjured muscle. Perhaps, given that MuSCs
on the individual myonuclear level (myonucleus analyses) are required demonstrate different behaviors and kinetics in overloaded and re-
to understand alterations in myonuclear functions and the effect of generating muscle, overloaded muscle may require overload-specific
‘diseased myonuclei’ on other myonuclei during disease progression. In molecules and events.
order to consider ‘myonuclear replenishment therapy’ using MuSCs, it is Finally, our conclusion is that degenerative damage to myofibers is
also necessary to investigate whether MuSC-derived myonuclei affect not necessary for both MuSC behaviors and hypertrophy in overloaded
‘diseased myonuclei’ or vice versa. muscle.
Using genetically MuSC-depleted sedentary mice, it was demon-
strated that the role of MuSCs in the onset of sarcopenia is limited Declaration of competing interest
[145,146]. This suggests that the loss of MuSCs might not be re-
sponsible for the onset and progression of sarcopenia. However, Eng- The authors declare that they have no known competing financial
lund et al. (2020) indicated that lifelong physical activity induces the interests or personal relationships that could have appeared to influ-
loss of MuSCs which blunts myofiber hypertrophy [149]. Furthermore, ence the work reported in this paper.
the inhibitory roles of MuSCs and MuSC-derived myonuclei in the de-
generation of the neuromuscular junction during aging have also been Acknowledgements
reported [150]. The extent to which decreased MuSC pool size and
function are responsible for the onset and progression of human sar- We thank Katherine Ono for proofreading the paper. This study is
copenia is still unknown. However, if we can control MuSC behaviors supported by JSPS KAKENHI grant, Grant-in-Aid for Scientific Research
and renew myonuclei in atrophic muscles, regardless of the contribu- (B) (19H04000 to S. F.) and Intramural Research Grant for Neurological
tion of MuSCs to human sarcopenia, MuSCs might be a great potential and Psychiatric Disorders of NCNP (28-6 to S. F.).
target for the treatment of muscular atrophy, including sarcopenia, as
well as muscular dystrophy. References

6. Future directions [1] A. Mauro, Satellite cell of skeletal muscle fibers, J. Biophys. Biochem. Cytol. 9
(1961) 493–495.
Notably, MuSCs are maintained in randomly contracting or relaxing [2] C. Lepper, T.A. Partridge, C.M. Fan, An absolute requirement for Pax7-positive
satellite cells in acute injury-induced skeletal muscle regeneration, Development
myofibers, meaning that MuSCs might always perceive these move- 138 (17) (2011) 3639–3646.
ments evoked by myofibers. Although MuSC quiescence should be [3] R. Sambasivan, R. Yao, A. Kissenpfennig, L. Van Wittenberghe, A. Paldi,
sustained in daily life, both overload and strength exercise induce MuSC B. Gayraud-Morel, H. Guenou, B. Malissen, S. Tajbakhsh, A. Galy, Pax7-expressing
satellite cells are indispensable for adult skeletal muscle regeneration,
proliferation, meaning that MuSCs have a physiological contraction Development 138 (17) (2011) 3647–3656.
threshold in order to avoid accidental activation/proliferation. The [4] S.I. Fukada, The roles of muscle stem cells in muscle injury, atrophy and hyper-
molecules and pathways that regulate the impact of daily myofiber trophy, J. Biochem. 163 (5) (2018) 353–358.
[5] D.L. Allen, R.R. Roy, V.R. Edgerton, Myonuclear domains in muscle adaptation
contractions, and which molecules disrupt the quiescent state are still
and disease, Muscle Nerve 22 (10) (1999) 1350–1360.
unknown. Such investigations will help us to identify the mechanisms [6] C.B. Ebbeling, P.M. Clarkson, Exercise-induced muscle damage and adaptation,
underlying MuSC activation/proliferation in overloaded muscle, which Sports Med. 7 (4) (1989) 207–234.
[7] P.M. Clarkson, M.J. Hubal, Exercise-induced muscle damage in humans, Am J
may lead to MuSC-based therapy for atrophied or aging muscle.
Phys Med Rehabil 81 (11 Suppl) (2002) S52–S69.
The theory of muscle memory is also a fascinating issue in MuSC [8] J.G. Yu, L. Carlsson, L.E. Thornell, Evidence for myofibril remodeling as opposed
biology. There are signs that previously trained muscles reacquire some to myofibril damage in human muscles with DOMS: an ultrastructural and im-
parameters at an accelerated rate if they are resubmitted to exercise, munoelectron microscopic study, Histochem. Cell Biol. 121 (3) (2004) 219–227.
[9] J.G. Yu, C. Malm, L.E. Thornell, Eccentric contractions leading to DOMS do not
even after prolonged detraining [151–153]. Bruusgaard et al. (2010) cause loss of desmin nor fibre necrosis in human muscle, Histochem. Cell Biol. 118
indicated that SA surgery lead to an increase in the myonuclear number (1) (2002) 29–34.
and was subsequently sustained in denervation-induced atrophied [10] R.M. Crameri, H. Langberg, P. Magnusson, C.H. Jensen, H.D. Schroder,
J.L. Olesen, C. Suetta, B. Teisner, M. Kjaer, Changes in satellite cells in human
myofibers, which supports the muscle memory requirement as myo- skeletal muscle after a single bout of high intensity exercise, J. Physiol. 558 (Pt 1)
nuclei are able to sustain certain parameters [154]. However, Dungan (2004) 333–340.
et al. (2019) indicated the number of myonuclei gained during training [11] R.M. Crameri, P. Aagaard, K. Qvortrup, H. Langberg, J. Olesen, M. Kjaer, Myofibre
damage in human skeletal muscle: effects of electrical stimulation versus volun-
(a combination of load with wheel running) was lost after detraining tary contraction, J. Physiol. 583 (Pt 1) (2007) 365–380.
[60]. Thus, the muscle memory requirement regarding sustained [12] A.L. Mackey, M. Kjaer, The breaking and making of healthy adult human skeletal
amount of myonuclei is highly debated [155]. The physiological and muscle in vivo, Skelet. Muscle 7 (1) (2017) 24.
[13] F. Damas, C.A. Libardi, C. Ugrinowitsch, The development of skeletal muscle
memory roles of myonuclei in daily life should be elucidated in the near hypertrophy through resistance training: the role of muscle damage and muscle
future as this may provide us with a new sense of appreciation re- protein synthesis, Eur. J. Appl. Physiol. 118 (3) (2018) 485–500.
garding the value of MuSCs in the development of new treatments for [14] D. Bansal, K. Miyake, S.S. Vogel, S. Groh, C.C. Chen, R. Williamson, P.L. McNeil,
K.P. Campbell, Defective membrane repair in dysferlin-deficient muscular
muscular disease.

8
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

dystrophy, Nature 423 (6936) (2003) 168–172. [41] S. Fujimaki, D. Seko, Y. Kitajima, K. Yoshioka, Y. Tsuchiya, S. Masuda, Y. Ono,
[15] C. Cai, H. Masumiya, N. Weisleder, N. Matsuda, M. Nishi, M. Hwang, J.K. Ko, Notch1 and Notch2 coordinately regulate stem cell function in the quiescent and
P. Lin, A. Thornton, X. Zhao, Z. Pan, S. Komazaki, M. Brotto, H. Takeshima, J. Ma, activated states of muscle satellite cells, Stem Cells 36 (2) (2018) 278–285.
MG53 nucleates assembly of cell membrane repair machinery, Nat. Cell Biol. 11 [42] M.B. Baghdadi, J. Firmino, K. Soni, B. Evano, D. Di Girolamo, P. Mourikis, D.
(1) (2009) 56–64. Castel, S. Tajbakhsh, Notch-induced miR-708 antagonizes satellite cell migration
[16] K. Nosaka, P.M. Clarkson, Muscle damage following repeated bouts of high force and maintains quiescence, Cell Stem Cell 23(6) (2018) 859–868 e5.
eccentric exercise, Med. Sci. Sports Exerc. 27 (9) (1995) 1263–1269. [43] M. Yamaguchi, Y. Watanabe, T. Ohtani, A. Uezumi, N. Mikami, M. Nakamura,
[17] K.C. Darr, E. Schultz, Exercise-induced satellite cell activation in growing and T. Sato, M. Ikawa, M. Hoshino, K. Tsuchida, Y. Miyagoe-Suzuki, K. Tsujikawa,
mature skeletal muscle, J Appl Physiol (1985) 63(5) (1987) 1816–21. S. Takeda, H. Yamamoto, S. Fukada, Calcitonin receptor signaling inhibits muscle
[18] J.P. Loenneke, R.S. Thiebaud, T. Abe, Does blood flow restriction result in skeletal stem cells from escaping the quiescent state and the niche, Cell Rep. 13 (2) (2015)
muscle damage? A critical review of available evidence, Scandinavian Journal of 302–314.
Medicine & Science in Sports 24 (6) (2014) e415–e422. [44] S. Fukada, A. Uezumi, M. Ikemoto, S. Masuda, M. Segawa, N. Tanimura,
[19] S. Fukuda, A. Kaneshige, T. Kaji, Y.T. Noguchi, Y. Takemoto, L. Zhang, H. Yamamoto, Y. Miyagoe-Suzuki, S. Takeda, Molecular signature of quiescent
K. Tsujikawa, H. Kokubo, A. Uezumi, K. Maehara, A. Harada, Y. Ohkawa, satellite cells in adult skeletal muscle, Stem Cells 25 (10) (2007) 2448–2459.
S.I. Fukada, Sustained expression of HeyL is critical for the proliferation of muscle [45] M. Quarta, J.O. Brett, R. DiMarco, A. De Morree, S.C. Boutet, R. Chacon,
stem cells in overloaded muscle, Elife 8 (2019) e48284. M.C. Gibbons, V.A. Garcia, J. Su, J.B. Shrager, S. Heilshorn, T.A. Rando, An ar-
[20] P.S. Zammit, J.P. Golding, Y. Nagata, V. Hudon, T.A. Partridge, J.R. Beauchamp, tificial niche preserves the quiescence of muscle stem cells and enhances their
Muscle satellite cells adopt divergent fates: a mechanism for self-renewal? J. Cell therapeutic efficacy, Nat. Biotechnol. 34 (7) (2016) 752–759.
Biol. 166 (3) (2004) 347–357. [46] L. Zhang, Y.T. Noguchi, H. Nakayama, T. Kaji, K. Tsujikawa, M. Ikemoto-Uezumi,
[21] H. Yin, A. Pasut, V.D. Soleimani, C.F. Bentzinger, G. Antoun, S. Thorn, P. Seale, A. Uezumi, Y. Okada, T. Doi, S. Watanabe, T. Braun, Y. Fujio, S.I. Fukada, The
P. Fernando, W. van Ijcken, F. Grosveld, R.A. Dekemp, R. Boushel, M.E. Harper, CalcR-PKA-Yap1 axis is critical for maintaining quiescence in muscle stem cells,
M.A. Rudnicki, MicroRNA-133 controls brown adipose determination in skeletal Cell Reports 29(8) (2019) 2154–2163 e5.
muscle satellite cells by targeting Prdm16, Cell Metab. 17 (2) (2013) 210–224. [47] T.H. Cheung, N.L. Quach, G.W. Charville, L. Liu, L. Park, A. Edalati, B. Yoo,
[22] P. Pessina, Y. Kharraz, M. Jardi, S. Fukada, A.L. Serrano, E. Perdiguero, P. Munoz- P. Hoang, T.A. Rando, Maintenance of muscle stem-cell quiescence by microRNA-
Canoves, Fibrogenic cell plasticity blunts tissue regeneration and aggravates 489, Nature 482 (7386) (2012) 524–528.
muscular dystrophy, Stem Cell Reports 4 (6) (2015) 1046–1060. [48] C. Christov, F. Chretien, R. Abou-Khalil, G. Bassez, G. Vallet, F.J. Authier,
[23] A.S. Brack, M.J. Conboy, S. Roy, M. Lee, C.J. Kuo, C. Keller, T.A. Rando, Increased Y. Bassaglia, V. Shinin, S. Tajbakhsh, B. Chazaud, R.K. Gherardi, Muscle satellite
Wnt signaling during aging alters muscle stem cell fate and increases fibrosis, cells and endothelial cells: close neighbors and privileged partners, Mol. Biol. Cell
Science 317 (5839) (2007) 807–810. 18 (4) (2007) 1397–1409.
[24] A. Asakura, M. Komaki, M. Rudnicki, Muscle satellite cells are multipotential stem [49] M. Verma, Y. Asakura, B.S.R. Murakonda, T. Pengo, C. Latroche, B. Chazaud, L.K.
cells that exhibit myogenic, osteogenic, and adipogenic differentiation, McLoon, A. Asakura, Muscle satellite cell cross-talk with a vascular niche main-
Differentiation 68 (4–5) (2001) 245–253. tains quiescence via VEGF and Notch signaling, Cell Stem Cell 23(4) (2018)
[25] A. Uezumi, S. Fukada, N. Yamamoto, S. Takeda, K. Tsuchida, Mesenchymal pro- 530–543 e9.
genitors distinct from satellite cells contribute to ectopic fat cell formation in [50] A.J. Goel, M.K. Rieder, H.H. Arnold, G.L. Radice, R.S. Krauss, Niche cadherins
skeletal muscle, Nat. Cell Biol. 12 (2) (2010) 143–152. control the quiescence-to-activation transition in muscle stem cells, Cell Rep. 21
[26] A.W. Joe, L. Yi, A. Natarajan, F. Le Grand, L. So, J. Wang, M.A. Rudnicki, (8) (2017) 2236–2250.
F.M. Rossi, Muscle injury activates resident fibro/adipogenic progenitors that fa- [51] S. Eliazer, J.M. Muncie, J. Christensen, X. Sun, R.S. D'Urso, V.M. Weaver, A.S.
cilitate myogenesis, Nat. Cell Biol. 12 (2) (2010) 153–163. Brack, Wnt4 from the niche controls the mechano-properties and quiescent state of
[27] V. Zismanov, V. Chichkov, V. Colangelo, S. Jamet, S. Wang, A. Syme, muscle stem cells, Cell Stem Cell 25(5) (2019) 654–665.e4.
A.E. Koromilas, C. Crist, Phosphorylation of eIF2alpha is a translational control [52] M.N. Wosczyna, C.T. Konishi, E.E. Perez Carbajal, T.T. Wang, R.A. Walsh, Q. Gan,
mechanism regulating muscle stem cell quiescence and self-renewal, Cell Stem M.W. Wagner, T.A. Rando, Mesenchymal stromal cells are required for re-
Cell 18 (1) (2016) 79–90. generation and homeostatic maintenance of skeletal muscle, Cell Reports 27(7)
[28] A. de Morree, C.T.J. van Velthoven, Q. Gan, J.S. Salvi, J.D.D. Klein, I. Akimenko, (2019) 2029–2035 e5.
M. Quarta, S. Biressi, T.A. Rando, Staufen1 inhibits MyoD translation to actively [53] F. Demontis, N. Perrimon, Integration of insulin receptor/Foxo signaling and dMyc
maintain muscle stem cell quiescence, Proc. Natl. Acad. Sci. U. S. A. 114 (43) activity during muscle growth regulates body size in Drosophila, Development 136
(2017) E8996–E9005. (6) (2009) 983–993.
[29] B. Gayraud-Morel, F. Chretien, A. Jory, R. Sambasivan, E. Negroni, P. Flamant, [54] M.B. Baghdadi, S. Tajbakhsh, Regulation and phylogeny of skeletal muscle re-
G. Soubigou, J.Y. Coppee, J. Di Santo, A. Cumano, V. Mouly, S. Tajbakhsh, Myf5 generation, Dev. Biol. 433 (2) (2017) 299-209.
haploinsufficiency reveals distinct cell fate potentials for adult skeletal muscle [55] C.S. Fry, J.D. Lee, J.R. Jackson, T.J. Kirby, S.A. Stasko, H. Liu, E.E. Dupont-
stem cells, J. Cell Sci. 125 (Pt 7) (2012) 1738–1749. Versteegden, J.J. McCarthy, C.A. Peterson, Regulation of the muscle fiber micro-
[30] A. Uezumi, S. Fukada, N. Yamamoto, M. Ikemoto-Uezumi, M. Nakatani, M. Morita, environment by activated satellite cells during hypertrophy, FASEB J. 28 (4)
A. Yamaguchi, H. Yamada, I. Nishino, Y. Hamada, K. Tsuchida, Identification and (2014) 1654–1665.
characterization of PDGFRalpha(+) mesenchymal progenitors in human skeletal [56] H. Klitgaard, A model for quantitative strength training of hindlimb muscles of the
muscle, Cell Death Dis. 5 (2014) e1186. rat, J Appl Physiol (1985) 64(4) (1988) 1740–5.
[31] A. Uezumi, T. Ito, D. Morikawa, N. Shimizu, T. Yoneda, M. Segawa, M. Yamaguchi, [57] D. Cui, J.C. Drake, R.J. Wilson, R.J. Shute, B. Lewellen, M. Zhang, H. Zhao,
R. Ogawa, M.M. Matev, Y. Miyagoe-Suzuki, S. Takeda, K. Tsujikawa, K. Tsuchida, O.L. Sabik, S. Onengut, S.S. Berr, S.S. Rich, C.R. Farber, Z. Yan, A novel voluntary
H. Yamamoto, S. Fukada, Fibrosis and adipogenesis originate from a common weightlifting model in mice promotes muscle adaptation and insulin sensitivity
mesenchymal progenitor in skeletal muscle, J. Cell Sci. 124 (Pt 21) (2011) with simultaneous enhancement of autophagy and mTOR pathway, FASEB J.
3654–3664. (2020), https://doi.org/10.1096/fj.201903055R.
[32] T.H. Cheung, T.A. Rando, Molecular regulation of stem cell quiescence, Nat. Rev. [58] A.J. Blazevich, D. Cannavan, D.R. Coleman, S. Horne, Influence of concentric and
Mol. Cell Biol. 14 (6) (2013) 329–340. eccentric resistance training on architectural adaptation in human quadriceps
[33] S. Fukada, Y. Ma, T. Ohtani, Y. Watanabe, S. Murakami, M. Yamaguchi, Isolation, muscles, J Appl Physiol (1985) 103(5) (2007) 1565–75.
characterization, and molecular regulation of muscle stem cells, Front. Physiol. 4 [59] A. Ishihara, R.R. Roy, Y. Ohira, Y. Ibata, V.R. Edgerton, Hypertrophy of rat
(2013) 317. plantaris muscle fibers after voluntary running with increasing loads, J Appl
[34] B. Evano, S. Tajbakhsh, Skeletal muscle stem cells in comfort and stress, NPJ Physiol (1985) 84(6) (1998) 2183–9.
Regen Med 3 (2018) 24. [60] C.M. Dungan, K.A. Murach, K.K. Frick, S.R. Jones, S.E. Crow, D.A. Englund,
[35] H. Yin, F. Price, M.A. Rudnicki, Satellite cells and the muscle stem cell niche, I.J. Vechetti Jr., V.C. Figueiredo, B.M. Levitan, J. Satin, J.J. McCarthy,
Physiol. Rev. 93 (1) (2013) 23–67. C.A. Peterson, Elevated myonuclear density during skeletal muscle hypertrophy in
[36] M.B. Baghdadi, D. Castel, L. Machado, S.I. Fukada, D.E. Birk, F. Relaix, response to training is reversed during detraining, Am J Physiol Cell Physiol 316
S. Tajbakhsh, P. Mourikis, Reciprocal signalling by Notch-collagen V-CALCR re- (5) (2019) C649–C654.
tains muscle stem cells in their niche, Nature 557 (2018) 714–718. [61] K. Legerlotz, B. Elliott, B. Guillemin, H.K. Smith, Voluntary resistance running
[37] Y.T. Noguchi, M. Nakamura, N. Hino, J. Nogami, S. Tsuji, T. Sato, L. Zhang, K. wheel activity pattern and skeletal muscle growth in rats, Exp. Physiol. 93 (6)
Tsujikawa, T. Tanaka, K. Izawa, Y. Okada, T. Doi, H. Kokubo, A. Harada, A. (2008) 754–762.
Uezumi, M. Gessler, Y. Ohkawa, S.I. Fukada, Cell-autonomous and redundant roles [62] M. Kurosaka, H. Naito, Y. Ogura, A. Kojima, K. Goto, S. Katamoto, Effects of vo-
of Hey1 and HeyL in muscle stem cells: HeyL requires Hes1 to bind diverse DNA luntary wheel running on satellite cells in the rat plantaris muscle, J. Sports Sci.
sites, Development 146(4) (2019) dev163618. Med. 8 (1) (2009) 51–57.
[38] C.R. Bjornson, T.H. Cheung, L. Liu, P.V. Tripathi, K.M. Steeper, T.A. Rando, Notch [63] P. Li, T. Akimoto, M. Zhang, R.S. Williams, Z. Yan, Resident stem cells are not
signaling is necessary to maintain quiescence in adult muscle stem cells, Stem Cells required for exercise-induced fiber-type switching and angiogenesis but are ne-
30 (2) (2012) 232–242. cessary for activity-dependent muscle growth, Am J Physiol Cell Physiol 290 (6)
[39] P. Mourikis, R. Sambasivan, D. Castel, P. Rocheteau, V. Bizzarro, S. Tajbakhsh, A (2006) C1461–C1468.
critical requirement for Notch signaling in maintenance of the quiescent skeletal [64] A. Irintchev, A. Wernig, Muscle damage and repair in voluntarily running mice:
muscle stem cell state, Stem Cells 30 (2) (2012) 243–252. strain and muscle differences, Cell Tissue Res. 249 (3) (1987) 509–521.
[40] S. Mizuno, M. Yoda, M. Shimoda, T. Tohmonda, Y. Okada, Y. Toyama, S. Takeda, [65] A. Moraska, T. Deak, R.L. Spencer, D. Roth, M. Fleshner, Treadmill running pro-
M. Nakamura, M. Matsumoto, K. Horiuchi, A disintegrin and metalloprotease 10 duces both positive and negative physiological adaptations in Sprague-Dawley
(ADAM10) is indispensable for maintenance of the muscle satellite cell pool, J. rats, Am J Physiol Regul Integr Comp Physiol 279 (4) (2000) R1321–R1329.
Biol. Chem. 290 (47) (2015) 28456–28464. [66] R.B. Armstrong, R.W. Ogilvie, J.A. Schwane, Eccentric exercise-induced injury to

9
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

rat skeletal muscle, J. Appl. Physiol. Respir. Environ. Exerc. Physiol. 54 (1) (1983) exercise in humans stimulates cell fusion of myoblasts along with fusogenic en-
80–93. dogenous retroviral genes in vivo, PLoS One 10 (7) (2015) e0132099.
[67] K.M. McCormick, D.P. Thomas, Exercise-induced satellite cell activation in se- [94] H.K. Smith, L. Maxwell, C.D. Rodgers, N.H. McKee, M.J. Plyley, Exercise-enhanced
nescent soleus muscle, J Appl Physiol (1985) 72(3) (1992) 888–93. satellite cell proliferation and new myonuclear accretion in rat skeletal muscle, J
[68] M.M. Umnova, T.P. Seene, The effect of increased functional load on the activation Appl Physiol (1985) 90(4) (2001) 1407–14.
of satellite cells in the skeletal muscle of adult rats, Int. J. Sports Med. 12 (5) [95] J.D. Lee, C.S. Fry, J. Mula, T.J. Kirby, J.R. Jackson, F. Liu, L. Yang, E.E. Dupont-
(1991) 501–504. Versteegden, J.J. McCarthy, C.A. Peterson, Aged muscle demonstrates Fiber-type
[69] M.V. Franchi, N.D. Reeves, M.V. Narici, Skeletal muscle remodeling in response to adaptations in response to mechanical overload, in the absence of myofiber hy-
eccentric vs. concentric loading: morphological, molecular, and metabolic adap- pertrophy, independent of satellite cell abundance, The Journals of Gerontology.
tations, Frontiers in Physiology 8 (2017) 447. Series A, Biological Sciences and Medical Sciences 71 (4) (2016) 461–467.
[70] J. Douglas, S. Pearson, A. Ross, M. McGuigan, Chronic adaptations to eccentric [96] T. Moro, C.R. Brightwell, E. Volpi, B.B. Rasmussen, C.S. Fry, Resistance exercise
training: a systematic review, Sports Med. 47 (5) (2017) 917–941. training promotes fiber type-specific myonuclear adaptations in older adults, J
[71] P. Wainman, G. Shipounoff, The effect of castration and testosterone propionate Appl Physiol (1985) 128(4) (2020) 795–804.
on the striated perineal musculature in the rat, Endocrinology 29 (1941) 975. [97] M.T. Webster, U. Manor, J. Lippincott-Schwartz, C.M. Fan, Intravital imaging re-
[72] C. Chambon, D. Duteil, A. Vignaud, A. Ferry, N. Messaddeq, R. Malivindi, S. Kato, veals ghost fibers as architectural units guiding myogenic progenitors during re-
P. Chambon, D. Metzger, Myocytic androgen receptor controls the strength but not generation, Cell Stem Cell 18 (2) (2016) 243–252.
the mass of limb muscles, Proc. Natl. Acad. Sci. U. S. A. 107 (32) (2010) [98] Y. Takemoto, S. Inaba, L. Zhang, K. Tsujikawa, A. Uezumi, S.I. Fukada, Implication
14327–14332. of basal lamina dependency in survival of Nrf2-null muscle stem cells via an an-
[73] I. Sinha-Hikim, S.M. Roth, M.I. Lee, S. Bhasin, Testosterone-induced muscle hy- tioxidative-independent mechanism, J. Cell. Physiol. 234 (2) (2019) 1689–1698.
pertrophy is associated with an increase in satellite cell number in healthy, young [99] D. Hardy, A. Besnard, M. Latil, G. Jouvion, D. Briand, C. Thepenier, Q. Pascal,
men, Am. J. Physiol. Endocrinol. Metab. 285 (1) (2003) E197–E205. A. Guguin, B. Gayraud-Morel, J.M. Cavaillon, S. Tajbakhsh, P. Rocheteau,
[74] J.O. Nnodim, Testosterone mediates satellite cell activation in denervated rat le- F. Chretien, Comparative study of injury models for studying muscle regeneration
vator ani muscle, Anat. Rec. 263 (1) (2001) 19–24. in mice, PLoS One 11 (1) (2016) e0147198.
[75] Y. Chen, J.D. Zajac, H.E. MacLean, Androgen regulation of satellite cell function, J. [100] R. Ogawa, Y. Ma, M. Yamaguchi, T. Ito, Y. Watanabe, T. Ohtani, S. Murakami,
Endocrinol. 186 (1) (2005) 21–31. S. Uchida, P. De Gaspari, A. Uezumi, M. Nakamura, Y. Miyagoe-Suzuki,
[76] M.J. Krahn, J.E. Anderson, Anabolic steroid treatment increases myofiber damage K. Tsujikawa, N. Hashimoto, T. Braun, T. Tanaka, S. Takeda, H. Yamamoto,
in mdx mouse muscular dystrophy, J. Neurol. Sci. 125 (2) (1994) 138–146. S. Fukada, Doublecortin marks a new population of transiently amplifying muscle
[77] C.A. Maltin, S.M. Hay, M.I. Delday, P.J. Reeds, R.M. Palmer, Evidence that the progenitor cells and is required for myofiber maturation during skeletal muscle
hypertrophic action of clenbuterol on denervated rat muscle is not propranolol- regeneration, Development 142 (1) (2015) 51–61.
sensitive, Br. J. Pharmacol. 96 (4) (1989) 817–822. [101] C.L. Mendias, A.J. Schwartz, J.A. Grekin, J.P. Gumucio, K.B. Sugg, Changes in
[78] J.J. McCarthy, J. Mula, M. Miyazaki, R. Erfani, K. Garrison, A.B. Farooqui, muscle fiber contractility and extracellular matrix production during skeletal
R. Srikuea, B.A. Lawson, B. Grimes, C. Keller, G. Van Zant, K.S. Campbell, muscle hypertrophy, J Appl Physiol (1985) 122(3) (2017) 571–579.
K.A. Esser, E.E. Dupont-Versteegden, C.A. Peterson, Effective fiber hypertrophy in [102] W.A. He, E. Berardi, V.M. Cardillo, S. Acharyya, P. Aulino, J. Thomas-Ahner,
satellite cell-depleted skeletal muscle, Development 138 (17) (2011) 3657–3666. J. Wang, M. Bloomston, P. Muscarella, P. Nau, N. Shah, M.E. Butchbach,
[79] I.M. Egner, J.C. Bruusgaard, K. Gundersen, Satellite cell depletion prevents fiber K. Ladner, S. Adamo, M.A. Rudnicki, C. Keller, D. Coletti, F. Montanaro,
hypertrophy in skeletal muscle, Development 143 (16) (2016) 2898–2906. D.C. Guttridge, NF-kappaB-mediated Pax7 dysregulation in the muscle micro-
[80] Q. Goh, D.P. Millay, Requirement of myomaker-mediated stem cell fusion for environment promotes cancer cachexia, J. Clin. Invest. 123 (11) (2013)
skeletal muscle hypertrophy, Elife 6 (2017) e20007. 4821–4835.
[81] A. Stromberg, M. Jansson, H. Fischer, E. Rullman, H. Hagglund, T. Gustafsson, [103] R.J. Boutrup, J. Farup, K. Vissing, M. Kjaer, U.R. Mikkelsen, Skeletal muscle stem
Bone marrow derived cells in adult skeletal muscle tissue in humans, Skelet. cell characteristics and myonuclei content in patients with rheumatoid arthritis: a
Muscle 3 (1) (2013) 12. cross-sectional study, Rheumatol. Int. 38 (6) (2018) 1031–1041.
[82] N. Liu, G.A. Garry, S. Li, S. Bezprozvannaya, E. Sanchez-Ortiz, B. Chen, [104] S. Fujimaki, M. Machida, T. Wakabayashi, M. Asashima, T. Takemasa,
J.M. Shelton, P. Jaichander, R. Bassel-Duby, E.N. Olson, A Twist2-dependent T. Kuwabara, Functional overload enhances satellite cell properties in skeletal
progenitor cell contributes to adult skeletal muscle, Nat. Cell Biol. 19 (3) (2017) muscle, Stem Cells Int. 2016 (2016) 7619418.
202–213. [105] R.D. Hyldahl, T. Olson, T. Welling, L. Groscost, A.C. Parcell, Satellite cell activity is
[83] V. Randrianarison-Huetz, A. Papaefthymiou, G. Herledan, C. Noviello, differentially affected by contraction mode in human muscle following a work-
U. Faradova, L. Collard, A. Pincini, E. Schol, J.F. Decaux, P. Maire, matched bout of exercise, Front. Physiol. 5 (2014) 485.
S. Vassilopoulos, A. Sotiropoulos, Srf controls satellite cell fusion through the [106] B. Evano, S. Khalilian, G. Le Carrou, G. Almouzni, S. Tajbakhsh, Dynamics of
maintenance of actin architecture, J. Cell Biol. 217 (2) (2018) 685–700. asymmetric and symmetric divisions of muscle stem cells in vivo and on artificial
[84] K.A. Murach, S.H. White, Y. Wen, A. Ho, E.E. Dupont-Versteegden, J.J. McCarthy, niches, Cell Reports 30(10) (2020) 3195–3206 e7.
C.A. Peterson, Differential requirement for satellite cells during overload-induced [107] S. Kuang, K. Kuroda, F. Le Grand, M.A. Rudnicki, Asymmetric self-renewal and
muscle hypertrophy in growing versus mature mice, Skelet. Muscle 7 (1) commitment of satellite stem cells in muscle, Cell 129 (5) (2007) 999–1010.
(2017) 14. [108] I.M. Conboy, T.A. Rando, The regulation of Notch signaling controls satellite cell
[85] C.S. Fry, T.J. Kirby, K. Kosmac, J.J. McCarthy, C.A. Peterson, Myogenic progenitor activation and cell fate determination in postnatal myogenesis, Dev. Cell 3 (3)
cells control extracellular matrix production by fibroblasts during skeletal muscle (2002) 397–409.
hypertrophy, Cell Stem Cell 20 (1) (2017) 56–69. [109] R.E. Allen, L.K. Boxhorn, Regulation of skeletal muscle satellite cell proliferation
[86] I. Sinha-Hikim, J. Artaza, L. Woodhouse, N. Gonzalez-Cadavid, A.B. Singh, and differentiation by transforming growth factor-beta, insulin-like growth factor
M.I. Lee, T.W. Storer, R. Casaburi, R. Shen, S. Bhasin, Testosterone-induced in- I, and fibroblast growth factor, J. Cell. Physiol. 138 (2) (1989) 311–315.
crease in muscle size in healthy young men is associated with muscle fiber hy- [110] R. Tatsumi, J.E. Anderson, C.J. Nevoret, O. Halevy, R.E. Allen, HGF/SF is present
pertrophy, Am. J. Physiol. Endocrinol. Metab. 283 (1) (2002) E154–E164. in normal adult skeletal muscle and is capable of activating satellite cells, Dev.
[87] D.A. Englund, B.D. Peck, K.A. Murach, A.C. Neal, H.A. Caldwell, J.J. McCarthy, Biol. 194 (1) (1998) 114–128.
C.A. Peterson, E.E. Dupont-Versteegden, Resident muscle stem cells are not re- [111] T.J. Hawke, D.J. Garry, Myogenic satellite cells: physiology to molecular biology,
quired for testosterone-induced skeletal muscle hypertrophy, Am J Physiol Cell J. Appl. Physiol. 91 (2) (2001) 534–551.
Physiol 317 (4) (2019) C719–C724. [112] M.T. Webster, C.M. Fan, c-MET regulates myoblast motility and myocyte fusion
[88] B. Blaauw, M. Canato, L. Agatea, L. Toniolo, C. Mammucari, E. Masiero, during adult skeletal muscle regeneration, PLoS One 8(11) (2013) e81757.
R. Abraham, M. Sandri, S. Schiaffino, C. Reggiani, Inducible activation of Akt [113] H. Zhu, F. Xiao, G. Wang, X. Wei, L. Jiang, Y. Chen, L. Zhu, H. Wang, Y. Diao,
increases skeletal muscle mass and force without satellite cell activation, FASEB J. H. Wang, N.Y. Ip, T.H. Cheung, Z. Wu, STAT3 regulates self-renewal of adult
23 (11) (2009) 3896–3905. muscle satellite cells during injury-induced muscle regeneration, Cell Rep. 16 (8)
[89] H. Amthor, A. Otto, A. Vulin, A. Rochat, J. Dumonceaux, L. Garcia, E. Mouisel, (2016) 2102–2115.
C. Hourde, R. Macharia, M. Friedrichs, F. Relaix, P.S. Zammit, A. Matsakas, [114] A.L. Serrano, B. Baeza-Raja, E. Perdiguero, M. Jardi, P. Munoz-Canoves,
K. Patel, T. Partridge, Muscle hypertrophy driven by myostatin blockade does not Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle
require stem/precursor-cell activity, Proc. Natl. Acad. Sci. U. S. A. 106 (18) (2009) hypertrophy, Cell Metab. 7 (1) (2008) 33–44.
7479–7484. [115] A. Guerci, C. Lahoute, S. Hebrard, L. Collard, D. Graindorge, M. Favier,
[90] A. Raffaello, G. Milan, E. Masiero, S. Carnio, D. Lee, G. Lanfranchi, A.L. Goldberg, N. Cagnard, S. Batonnet-Pichon, G. Precigout, L. Garcia, D. Tuil, D. Daegelen,
M. Sandri, JunB transcription factor maintains skeletal muscle mass and promotes A. Sotiropoulos, Srf-dependent paracrine signals produced by myofibers control
hypertrophy, J. Cell Biol. 191 (1) (2010) 101–113. satellite cell-mediated skeletal muscle hypertrophy, Cell Metab. 15 (1) (2012)
[91] A.I. McKenzie, A.C. D’Lugos, M.J. Saunders, K.D. Gworek, N.D. Luden, Fiber type- 25–37.
specific satellite cell content in cyclists following heavy training with carbohy- [116] L. Collard, G. Herledan, A. Pincini, A. Guerci, V. Randrianarison-Huetz,
drate and carbohydrate-protein supplementation, Front. Physiol. 7 (2016) 550. A. Sotiropoulos, Nuclear actin and myocardin-related transcription factors control
[92] A.R. Konopka, W.M. Castor, C.A. Wolff, R.V. Musci, J.J. Reid, J.L. Laurin, Z.J. disuse muscle atrophy through regulation of Srf activity, J. Cell Sci. 127 (Pt 24)
Valenti, K.L. Hamilton, B.F. Miller, Skeletal muscle mitochondrial protein synth- (2014) 5157–5163.
esis and respiration in response to the energetic stress of an ultra-endurance race, J [117] K. Kuwahara, H. Kinoshita, Y. Kuwabara, Y. Nakagawa, S. Usami, T. Minami,
Appl Physiol (1985) 123(6) (2017) 1516–1524. Y. Yamada, M. Fujiwara, K. Nakao, Myocardin-related transcription factor A is a
[93] S. Frese, M. Ruebner, F. Suhr, T.M. Konou, K.A. Tappe, M. Toigo, H.H. Jung, common mediator of mechanical stress- and neurohumoral stimulation-induced
C. Henke, R. Steigleder, P.L. Strissel, H. Huebner, M.W. Beckmann, P. van der cardiac hypertrophic signaling leading to activation of brain natriuretic peptide
Keylen, B. Schoser, T. Schiffer, L. Frese, W. Bloch, R. Strick, Long-term endurance gene expression, Mol. Cell. Biol. 30 (17) (2010) 4134–4148.

10
S.-i. Fukada, et al. BBA - Molecular Cell Research 1867 (2020) 118742

[118] L. Montel, A. Sotiropoulos, S. Henon, The nature and intensity of mechanical growth, Sci. Rep. 9 (1) (2019) 969.
stimulation drive different dynamics of MRTF-A nuclear redistribution after actin [138] M.L. Novak, W. Billich, S.M. Smith, K.B. Sukhija, T.J. McLoughlin,
remodeling in myoblasts, PLoS One 14 (3) (2019) e0214385. T.A. Hornberger, T.J. Koh, COX-2 inhibitor reduces skeletal muscle hypertrophy in
[119] T. Panciera, L. Azzolin, M. Cordenonsi, S. Piccolo, Mechanobiology of YAP and mice, Am J Physiol Regul Integr Comp Physiol 296 (4) (2009) R1132–R1139.
TAZ in physiology and disease, Nat. Rev. Mol. Cell Biol. 18 (12) (2017) 758–770. [139] A. Heydemann, J.M. Huber, A. Demonbreun, M. Hadhazy, E.M. McNally, Genetic
[120] T. Kim, S.J. Yang, D. Hwang, J. Song, M. Kim, S. Kyum Kim, K. Kang, J. Ahn, background influences muscular dystrophy, Neuromuscul. Disord. 15 (9–10)
D. Lee, M.Y. Kim, S. Kim, J. Seung Koo, S. Seok Koh, S.Y. Kim, D.S. Lim, A basal- (2005) 601–609.
like breast cancer-specific role for SRF-IL6 in YAP-induced cancer stemness, Nat. [140] S. Fukada, D. Morikawa, Y. Yamamoto, T. Yoshida, N. Sumie, M. Yamaguchi,
Commun. 6 (2015) 10186. T. Ito, Y. Miyagoe-Suzuki, S. Takeda, K. Tsujikawa, H. Yamamoto, Genetic back-
[121] C. Canto, Z. Gerhart-Hines, J.N. Feige, M. Lagouge, L. Noriega, J.C. Milne, ground affects properties of satellite cells and mdx phenotypes, Am. J. Pathol. 176
P.J. Elliott, P. Puigserver, J. Auwerx, AMPK regulates energy expenditure by (5) (2010) 2414–2424.
modulating NAD+ metabolism and SIRT1 activity, Nature 458 (7241) (2009) [141] W.D. Coley, L. Bogdanik, M.C. Vila, Q. Yu, J.H. Van Der Meulen, S. Rayavarapu,
1056–1060. J.S. Novak, M. Nearing, J.L. Quinn, A. Saunders, C. Dolan, W. Andrews,
[122] E. Koltai, Z. Bori, C. Chabert, H. Dubouchaud, H. Naito, S. Machida, K.J. Davies, C. Lammert, A. Austin, T.A. Partridge, G.A. Cox, C. Lutz, K. Nagaraju, Effect of
Z. Murlasits, A.C. Fry, I. Boldogh, Z. Radak, SIRT1 may play a crucial role in genetic background on the dystrophic phenotype in mdx mice, Hum. Mol. Genet.
overload-induced hypertrophy of skeletal muscle, J. Physiol. 595 (11) (2017) 25 (1) (2016) 130–145.
3361–3376. [142] H. Gordish-Dressman, R. Willmann, L. Dalle Pazze, A. Kreibich, M. van Putten, A.
[123] J.A. Ross, Y. Levy, K. Svensson, A. Philp, S. Schenk, J. Ochala, SIRT1 regulates Heydemann, L. Bogdanik, C. Lutz, K. Davies, A.R. Demonbreun, D. Duan, D. Elsey,
nuclear number and domain size in skeletal muscle fibers, J. Cell. Physiol. 233 (9) S.I. Fukada, M. Girgenrath, J. Patrick Gonzalez, M.D. Grounds, A. Nichols, T.
(2018) 7157–7163. Partridge, M. Passini, F. Sanarica, F.J. Schnell, D.J. Wells, T. Yokota, C.S. Young, Z.
[124] D.P. Millay, J.R. O’Rourke, L.B. Sutherland, S. Bezprozvannaya, J.M. Shelton, Zhong, C. Spurney, M. Spencer, A. De Luca, K. Nagaraju, A. Aartsma-Rus, “of mice
R. Bassel-Duby, E.N. Olson, Myomaker is a membrane activator of myoblast fusion and measures”: a project to improve how we advance Duchenne muscular dys-
and muscle formation, Nature 499 (7458) (2013) 301–305. trophy therapies to the clinic, J Neuromuscul Dis 5(4) (2018) 407–417.
[125] P. Bi, A. Ramirez-Martinez, H. Li, J. Cannavino, J.R. McAnally, J.M. Shelton, [143] M. Rodrigues, Y. Echigoya, R. Maruyama, K.R. Lim, S.I. Fukada, T. Yokota,
E. Sanchez-Ortiz, R. Bassel-Duby, E.N. Olson, Control of muscle formation by the Impaired regenerative capacity and lower revertant fibre expansion in dystrophin-
fusogenic micropeptide myomixer, Science 356 (6335) (2017) 323–327. deficient mdx muscles on DBA/2 background, Sci. Rep. 6 (2016) 38371.
[126] M.E. Quinn, Q. Goh, M. Kurosaka, D.G. Gamage, M.J. Petrany, V. Prasad, [144] A.S. Brack, H. Bildsoe, S.M. Hughes, Evidence that satellite cell decrement con-
D.P. Millay, Myomerger induces fusion of non-fusogenic cells and is required for tributes to preferential decline in nuclear number from large fibres during murine
skeletal muscle development, Nat. Commun. 8 (2017) 15665. age-related muscle atrophy, J. Cell Sci. 118 (Pt 20) (2005) 4813–4821.
[127] Q. Zhang, A.A. Vashisht, J. O’Rourke, S.Y. Corbel, R. Moran, A. Romero, [145] A.C. Keefe, J.A. Lawson, S.D. Flygare, Z.D. Fox, M.P. Colasanto, S.J. Mathew,
L. Miraglia, J. Zhang, E. Durrant, C. Schmedt, S.C. Sampath, S.C. Sampath, The M. Yandell, G. Kardon, Muscle stem cells contribute to myofibres in sedentary
microprotein Minion controls cell fusion and muscle formation, Nat. Commun. 8 adult mice, Nat. Commun. 6 (2015) 7087.
(2017) 15664. [146] C.S. Fry, J.D. Lee, J. Mula, T.J. Kirby, J.R. Jackson, F. Liu, L. Yang, C.L. Mendias,
[128] M.J. Petrany, D.P. Millay, Cell fusion: merging membranes and making muscle, E.E. Dupont-Versteegden, J.J. McCarthy, C.A. Peterson, Inducible depletion of
Trends Cell Biol. 29 (12) (2019) 964–973. satellite cells in adult, sedentary mice impairs muscle regenerative capacity
[129] E. Leikina, D.G. Gamage, V. Prasad, J. Goykhberg, M. Crowe, J. Diao, M.M. without affecting sarcopenia, Nat. Med. 21 (1) (2015) 76–80.
Kozlov, L.V. Chernomordik, D.P. Millay, Myomaker and myomerger work in- [147] T. Snijders, T. Aussieker, A. Holwerda, G. Parise, L.J.C. van Loon, L.B. Verdijk, The
dependently to control distinct steps of membrane remodeling during myoblast concept of skeletal muscle memory: evidence from animal and human studies,
fusion, Dev Cell 46(6) (2018) 767–780 e7. Acta Physiol (Oxf) (2020) e13465.
[130] V. Horsley, K.M. Jansen, S.T. Mills, G.K. Pavlath, IL-4 acts as a myoblast recruit- [148] H. Taylor-Weiner, C.L. Grigsby, D.M.S. Ferreira, J.M. Dias, M.M. Stevens,
ment factor during mammalian muscle growth, Cell 113 (4) (2003) 483–494. J.L. Ruas, A.I. Teixeira, Modeling the transport of nuclear proteins along single
[131] S. Orimo, E. Hiyamuta, K. Arahata, H. Sugita, Analysis of inflammatory cells and skeletal muscle cells, Proc. Natl. Acad. Sci. U. S. A. 117 (6) (2020) 2978–2986.
complement C3 in bupivacaine-induced myonecrosis, Muscle Nerve 14 (6) (1991) [149] D.A. Englund, K.A. Murach, C.M. Dungan, V.C. Figueiredo, I.J. Vechetti Jr.,
515–520. E.E. Dupont-Versteegden, J.J. McCarthy, C.A. Peterson, Depletion of resident
[132] M. Cantini, U. Carraro, Macrophage-released factor stimulates selectively myo- muscle stem cells negatively impacts running volume, physical function and
genic cells in primary muscle culture, J. Neuropathol. Exp. Neurol. 54 (1) (1995) muscle hypertrophy in response to lifelong physical activity, Am J Physiol Cell
121–128. Physiol (2020), https://doi.org/10.1152/ajpcell.00090.2020.
[133] L. Arnold, A. Henry, F. Poron, Y. Baba-Amer, N. van Rooijen, A. Plonquet, [150] W. Liu, A. Klose, S. Forman, N.D. Paris, L. Wei-LaPierre, M. Cortes-Lopez, A. Tan,
R.K. Gherardi, B. Chazaud, Inflammatory monocytes recruited after skeletal M. Flaherty, P. Miura, R.T. Dirksen, J.V. Chakkalakal, Loss of adult skeletal muscle
muscle injury switch into antiinflammatory macrophages to support myogenesis, stem cells drives age-related neuromuscular junction degeneration, Elife 6 (2017)
J. Exp. Med. 204 (5) (2007) 1057–1069. e26464.
[134] M. Segawa, S. Fukada, Y. Yamamoto, H. Yahagi, M. Kanematsu, M. Sato, T. Ito, [151] H. Lee, K. Kim, B. Kim, J. Shin, S. Rajan, J. Wu, X. Chen, M.D. Brown, S. Lee,
A. Uezumi, S. Hayashi, Y. Miyagoe-Suzuki, S. Takeda, K. Tsujikawa, H. Yamamoto, J.Y. Park, A cellular mechanism of muscle memory facilitates mitochondrial re-
Suppression of macrophage functions impairs skeletal muscle regeneration with modelling following resistance training, J. Physiol. 596 (18) (2018) 4413–4426.
severe fibrosis, Exp. Cell Res. 314 (17) (2008) 3232–3244. [152] I.M. Egner, J.C. Bruusgaard, E. Eftestol, K. Gundersen, A cellular memory me-
[135] R. Mounier, M. Theret, L. Arnold, S. Cuvellier, L. Bultot, O. Goransson, N. Sanz, chanism aids overload hypertrophy in muscle long after an episodic exposure to
A. Ferry, K. Sakamoto, M. Foretz, B. Viollet, B. Chazaud, AMPKalpha1 regulates anabolic steroids, J. Physiol. 591 (24) (2013) 6221–6230.
macrophage skewing at the time of resolution of inflammation during skeletal [153] K. Gundersen, Muscle memory and a new cellular model for muscle atrophy and
muscle regeneration, Cell Metab. 18 (2) (2013) 251–264. hypertrophy, J. Exp. Biol. 219 (Pt 2) (2016) 235–242.
[136] N. Dumont, J. Frenette, Macrophages protect against muscle atrophy and promote [154] J.C. Bruusgaard, I.B. Johansen, I.M. Egner, Z.A. Rana, K. Gundersen, Myonuclei
muscle recovery in vivo and in vitro: a mechanism partly dependent on the in- acquired by overload exercise precede hypertrophy and are not lost on detraining,
sulin-like growth factor-1 signaling molecule, Am. J. Pathol. 176 (5) (2010) Proc. Natl. Acad. Sci. U. S. A. 107 (34) (2010) 15111–15116.
2228–2235. [155] K.A. Murach, C.M. Dungan, E.E. Dupont-Versteegden, J.J. McCarthy, C.A.
[137] R.G. Walton, K. Kosmac, J. Mula, C.S. Fry, B.D. Peck, J.S. Groshong, B.S. Finlin, Peterson, “Muscle memory” not mediated by myonuclear number? Secondary
B. Zhu, P.A. Kern, C.A. Peterson, Human skeletal muscle macrophages increase analysis of human detraining data, J Appl Physiol (1985) 127(6) (2019) 1814-
following cycle training and are associated with adaptations that may facilitate 1816.

11

You might also like