You are on page 1of 10

REVIEW

Molecular and Cellular Determinants of Skeletal


Muscle Atrophy and Hypertrophy
Vittorio Sartorelli* and Marcella Fulco
(Published 3 August 2004)

Introduction the eukaryotic translation initiation factor 2B (eIF-2B) is activated


Two opposing phenomena—muscle growth and muscle atro- through Akt-mediated phosphorylation and inactivation of GSK-3β.
phy—are mechanistically linked, in that either the activity or in- In summary, activation of the IGF-1 pathway leads to a series of
activity of a common set of molecules controlling a few cellular phosphorylation events culminating in the activation of molecules
pathways determines whether the skeletal muscle tissue will re- that regulate protein synthesis and that are likely involved in the
spond to defined stimuli with increased protein synthesis and genesis of muscle hypertrophy.
stimulation of cell growth (muscle hypertrophy) or with in-
creased protein breakdown and reduced cell proliferation Molecular Determinants of Muscle Atrophy
(muscle atrophy). Among these molecules, insulin-like growth Skeletal muscle atrophy is characterized by a decrease in the
factor– 1 (IGF-1) occupies a nodal point. size of preexisting muscle fibers and is observed in several
physiological and pathological settings. Atrophy has been inter-

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


The IGF-1 Pathway preted as the result of a passive adaptation of the muscle to a
Pertinent to the discussion of the mechanisms regulating both lack of electrical or mechanical stimuli. Recently, this view has
muscle atrophy and muscle hypertrophy is the description of been challenged by several findings indicating that the estab-
pathways regulated by IGF-1 [Fig. 1; for a detailed review of lishment of an active transcriptional program is necessary for
the IGF-1 pathway in skeletal muscle, see (1)]. IGF-1 is a se- the induction of muscle atrophy.
creted growth factor that regulates several cellular biochemical The ubiquitin protein ligases MuRF1 and MAFbx. Using dif-
pathways after binding to its membrane receptor, the IGF-1 ferential display and complementary DNA (cDNA) microarray
receptor (IGFR). Upon binding of IGF-1, the IGFR, which is a approaches, the messenger RNAs (mRNAs) obtained from
receptor tyrosine kinase, becomes phosphorylated and recruits muscles of either rats or mice in which atrophy was induced by
the insulin receptor substrate 1 (IRS1), leading to the activation immobilization, denervation, hind-limb suspension, or food de-
of the lipid kinase phosphatidylinositol 3-kinase (PI3K). PI3K privation were compared with controls, and the expression of
catalyzes the transfer of a phosphate group to the membrane- two genes was consistently found to be increased. The two
bound phosphatidylinositol 4,5-bisphosphate (PIP2). Phospho- genes encode muscle RING finger 1 (MuRF1) (9) and muscle
rylation of PIP2 generates phosphatidylinositol 3,4,5-trisphos- atrophy F-box (MAFbx; also known as atrogin-1) (9, 10), two
phate (PIP3), which recruits two additional kinases, Akt1 and ubiquitin protein ligases expressed solely in skeletal and cardiac
phosphoinositide-dependent protein kinase-1 (PDK-1) (2). Atk1 muscles. Ubiquitin ligases are enzymes involved in catalyzing
is bound, phosphorylated, and activated by PDK-1 (3). Once the ubiquitination of proteins fated to be degraded by the pro-
Akt1 is activated, it initiates a cascade of phosphorylation teasome. During atrophy, there is extensive muscle remodeling
events targeting mammalian target of rapamycin (mTOR) (4)— characterized by increased proteolysis, and MuRF1 and MAFbx
which in turn phosphorylates the p70 S6 kinase (p70S6K)—and might mediate such a process. A role for the ubiquitination-
glycogen synthase kinase 3β (GSK-3β) (5). Phosphorylated mediated proteolysis of muscle proteins by MuRF1 is suggested
IRS1 also stimulates the Ras-Raf-MEK-ERK pathway, a mito- by experiments in which MuRF1 interacts with titin, a giant
gen-activated protein kinase (MAPK) pathway. Activation of myofibrillar protein located at the M line of the sarcomere and
this pathway may actually prevent hypertrophy because Ras- mediates titin’s degradation (11).
Raf-MEK-ERK inactivation, rather than its activation, appears An active role for both MuRF1 and MAFbx in mediating
to characterize skeletal muscle hypertrophy (6). Phosphoryla- muscle atrophy can be inferred from experiments conducted
tion of mTOR represses, through an adaptor protein known as with mice in which both alleles for either MAFbx or MuRF1
Raptor, eukaryotic initiation factor 4E (eIF-4E)–binding protein have been deleted. MAFbx−/− and MuRF1−/− animals are partial-
1 [4EBP1, also known as phosphorylated heat- and acid-stable ly refractory to denervation-induced atrophy (9). It should be
protein (PHAS-1)], an inhibitor of eIF-4E (7). Thus, mTOR- possible, in principle, to cross the MAFbx−/− and MuRF1−/− ani-
mediated inhibition of 4EBP1 results in activation of eIF-4E mals to obtain double-knockout MAFbx−/−:MuRF1−/− and to de-
and subsequent increased protein synthesis. An additional level termine whether these animals are viable and whether MAFbx
of regulation on mTOR is provided by glucose and amino acids, and MuRF1 act cooperatively to mediate atrophy. A comple-
which influence the interaction between Raptor and mTOR (8). mentary approach for evaluating the functional role of these two
Therefore, mTOR phosphorylation activates both eIF-4E and ubiquitin ligases could be pursued by generating mice that over-
p70S6K, two positive regulators of protein synthesis. Similarly, express either MAFbx or MuRF1, because they may develop
spontaneous muscle atrophy, have an exacerbated response to
Muscle Gene Expression Group Laboratory of Muscle Biology, atrophic stimuli, or fail to respond to hypertrophic stimuli.
National Institute of Arthritis, Musculoskeletal and Skin Diseases, Finally, because both MAFbx and MuRF1 are expressed in the
National Institutes of Health, Bethesda, MD 20892, USA. heart, it will be important to determine their role in pathological
*Corresponding author. E-mail: sartorev@mail.nih.gov conditions characterized by cardiac remodeling (for example,

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 1
REVIEW

heart failure, acute myocardial infarction, and IGFR IGFR


cardiomyopathies that accompany several
muscular dystrophies). It appears that neither
MuRF1 nor MAFbx plays a role in physio- IGF-1
logical muscle homeostasis, because both
MuRF1- and MAFbx-knockout animals have
normal skeletal muscles (9).
Nonetheless, the PI3K-Akt-mTOR and
ubiquitin-protein ligase pathways seem to be
linked, in that IGF-1-dependent activation of PIP2 PIP3
P
the PI3K-Akt-mTOR pathway blocks accu-
mulation of MAFbx mRNA and prevents P P PI3K P P P
muscle proteolysis in a cellular model of dex- P P
amethasone (DEX)-induced atrophy (12). P
From a mechanistic and therapeutic point of IRS1 Akt PDK-1
view, several models of atrophy—including P P

denervation, immobilization, and atrophies


accompanying chemically induced diabetes,
treatment with dexamethasone or interleukin-
1 (IL-1; a cachexia-inducing cytokine), and

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


P
experimental sepsis—all result in increased
P Raptor
P
GSK-3β mTOR 14-3-3
transcription, mRNA stabilization, or both, of 4EBP1
MuRF1 and MAFbx (9, 13, 14). This sug- P P P
P
gests the possibility that a common set of P P
transcriptional regulators are stimulated by FOXO
diverse atrophic stimuli and that pharmaco- p70S6K
logical modulators targeting these regulators
may be effective in preventing atrophy ob- eIF2B eIF4E
served in disparate pathological conditions.
The FOXO transcription factors and mus-
cle atrophy. Two studies have advanced our
knowledge of the molecular modulators of
muscle atrophy (15, 16). In these studies, the
FOXO family of transcription factors was re-
ported to regulate MAFbx and MuRF1 tran-
scription and to influence muscle atrophy. Akt Fig. 1. The IGF-1 pathway is involved in muscle atrophy and hypertrophy. Upon IGF-
negatively regulates FOXO transcription fac- 1 binding, the IGFR is phosphorylated. The phosphorylated receptor recruits and
tors by phosphorylating them and promoting phosphorylates the IRS1, which activates PI3K. PI3K transfers a phosphate group to
the membrane-bound PIP2 to generate PIP3. PIP3 serves as a nucleation site for
their nuclear export into the cytoplasm, where
Akt1 and PDK-1. PDK-1 phosphorylates and activates Akt1, which, in turn, catalyzes
they are retained through association with the the transfer of phosphate groups to several substrates. Akt-mediated phosphoryla-
14-3-3 proteins (17). Muscle atrophy is mim- tion of the FOXO transcription factors promotes their cytoplasmic retention and func-
icked in cultured myotubes—that is, terminal- tional inactivation through interaction with the 14-3-3 proteins. Because the FOXO
ly differentiated, postmitotic syncytial cells proteins stimulate the transcription of the atrophy-promoting factor MAFbx, FOXO in-
forming the muscle fiber—by exposing them activation prevents muscle atrophy. Akt phosphorylates and inhibits GSK-3β, which
to either DEX or starvation (removal of activates the eukaryotic translation factor eIF-2B and increases protein synthesis.
growth factors, glucose, and amino acids for 6 The mTOR is also a phosphorylation substrate for Akt. With the assistance of the in-
hours). In these situations, transcription of teracting protein Raptor, phosphorylated mTOR promotes phosphorylation and inhibi-
both MAFbx and MuRF1 is increased. Con- tion of 4EBP1. Also, mTOR promotes protein synthesis by relieving 4EBP1-mediated
comitantly, phosphorylation of Akt and of inhibition of eIF-4B. When phosphorylated by mTOR, the ribosomal p70S6K be-
FOXO1 and FOXO3 is reduced. Animals with comes activated and increases protein synthesis. Green arrows indicate phosphory-
denervation atrophy treated with intramuscular lation events that activate the substrate, whereas red arrows point at phosphoryla-
injection of IGF-1, which promotes Akt phos- tions that result in substrate inactivation.
phorylation, do not increase expression of
MAFbx and MuRF1 and, most importantly, are spared from mus- of blocking the increase in MAFbx and MuRF1. These findings
cle loss. FOXO3 dephosphorylation allows transcriptional activa- indicate that the effects of activating the IGF-1-PI3K-Akt path-
tion of the MAFbx promoter and consequently increases MAFbx way that prevents atrophy operate through the FOXO proteins.
mRNA production. Through Akt-mediated phosphorylation of Nonetheless, it is likely that additional targets besides MAFbx are
FOXO1, 3, and 4, IGF-1 counteracts the ability of DEX to acti- affected by the FOXO proteins. In fact, although overexpression
vate MAFbx and MuRF1 transcription. When cultured myotubes of MAFbx alone does not cause muscle atrophy, muscle fibers
exposed to DEX are infected with an adenovirus expressing a expressing a constitutively active form of FOXO3 display a much
constitutively active form of FOXO1, IGF-1 becomes incapable reduced diameter (16). Furthermore, in the same study, FOXO3

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 2
REVIEW

overexpression resulted in increased MAFbx transcription, where- Animals overexpressing IGF-1 under the control of muscle-
as in another study (15), MAFbx expression was not affected by specific regulatory elements display muscle hypertrophy and in-
FOXO1. These findings raise the possibility that FOXO3 and creased muscle regeneration during senescence (28, 29). These
FOXO1 may regulate different targets. Alternatively, the differ- findings, along with the observation that IGF-1 overexpression
ences intrinsic to the two experimental settings may be responsi- improves the phenotype of the mdx mouse model for Duchenne
ble for the different results. Indeed, reduced expression of muscular dystrophy, suggest that IGF-1 may positively influ-
FOXO1 achieved with RNA interference reduced activation of ence satellite cell proliferation, activation, or both (30). Consis-
the MAFbx promoter (16). The identification of additional FOXO tent with the existence of a linear pathway proceeding from
targets is expected to shed light on the molecules and mecha- IGF-1 to Akt, deliberate activation of the Akt-mTOR pathway
nisms regulating muscle atrophy. can stimulate fiber hypertrophy both in control and denervated
Another transcription factor, nuclear factor κB (NF-κB), muscles (31). Together, these results suggest a potential thera-
mediates the effects of tumor necrosis factor–α (TNF-α) and peutic value of manipulating the IGF-1–Akt-mTOR pathway in
interferon-γ (IFN-γ) (18) in promoting the muscle wasting that the treatment of muscle atrophy. In contrast, acute physical ex-
accompanies several pathological conditions (19). However, ercise is associated with increased levels of total and phospho-
forced activation of the NF-κB pathway does not increase rylated Akt, increased phosphorylation and reduced activity of
MAFbx expression (16), suggesting that muscle atrophy can be GSK-3β, and reduced levels of phosphorylated β-catenin (32).
elicited through processes that do not involve FOXO proteins, Thus, the Akt to GSK-3β pathway may also be involved in mus-
for example, NF-κB-mediated degradation of the master cle hypertrophy and in the initial phases of muscle activity that
regulatory protein MyoD (18). The possibility exists that contribute to hypertrophy.
different mediators of muscle atrophy may act at distinct stages Although there is little doubt that IGF-1 is involved in medi-

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


and may indeed cross-talk to ensure initiation and completion ating experimental muscle hypertrophy, additional molecules
of the atrophic program. and stimuli may be required to assist IGF-1 in this process.
Transgenic mice in which IGF-1 expression is driven by the
Exercise and Muscle Growth muscle-specific regulatory regions of the gene encoding myosin
Postnatal skeletal muscle growth in response to chronic physical light chain 1/3 (MLC1/3) display pronounced muscle hypertro-
exercise is characterized by cell hypertrophy. Skeletal muscle phy (33). Expression of the endogenous MLC1/3 gene and acti-
hypertrophy is defined as an increase in fiber diameter without vation of the IGF-1 transgene occurred early during develop-
an apparent increase in the number of muscle fibers, accompa- ment. Nonetheless, muscle hypertrophy in MLC1/3–IGF-1 mice
nied by enhanced protein synthesis and augmented contractile first becomes detectable only 10 days after birth (33). It is pos-
force. Microtraumas resulting from chronic exercise also lead to sible that moderate physical exercise may be required for the
satellite cell activation and proliferation, which contribute to the overexpressed IGF-1 to induce hypertrophy or that molecules
increased muscle mass that ensues from extensive physical expressed or activated after birth may be necessary for the hy-
activity. The molecular determinants of muscle growth are be- pertrophic effects of IGF-1. Alternatively, the rapid postnatal
ginning to be characterized. muscle growth in both the transgenic and the wild-type animals
IGF-1 in muscle growth. Increased muscle loading, such as may mask the effects of IGF-1, which become evident in
that resulting from chronic exercise, results in augmented ex- transgenic animals only after muscle growth slows in the con-
pression of the gene encoding IGF-1 in both animal models trol animals. The physiological role of the Akt-mTOR pathway
(20) and humans (21). More specifically, two IGF-1 isoforms, in ensuring muscle trophism also remains to be fully clarified,
IGF-1E and mechanogrowth factor (MGF), seem to be selec- because pharmacological treatment with the mTOR-specific
tively expressed in skeletal muscle and to be regulated by me- inhibitor rapamycin fails to induce muscle atrophy in control
chanical load (22). These observations, along with the findings animals (31).
that the circulating IGF-1 produced by the liver is not necessary Myostatin. Myostatin is a transforming growth factor–β
for postnatal body growth (23), suggest that specific isoforms (TGF-β) family member expressed in the myotome compart-
of IGF-1 act in an autocrine or paracrine manner to regulate ment of developing somites and in the skeletal muscles of adult
skeletal muscle biology. Availability of IGF-1 for muscle IGFR animals (34). Naturally occurring mutations in the myostatin
is controlled by the combination of IGF-1, IGFR, and IGF-1 [Mstn, also known as growth and differentiation factor 8
binding proteins (IGFBPs). Binding of IGF-1 to certain IGFBPs (GDF-8)] gene are responsible for the “double-muscling” phe-
results in opposing biological effects. For instance, binding of notype, which is characterized by a dramatic increase in mus-
IGF-1 to IGFBP-4 appears to inhibit cell proliferation and dif- cle mass of certain breeds of cattle (35). Mstn-null mice show
ferentiation, whereas binding to IGFBP-5 has a dual effect (that an increase in muscle mass due to muscle hyperplasia (in-
is, either inhibition or stimulation of IGF-1 effects), depending creased number of muscle fibers) and hypertrophy (increased
on the culture conditions (24, 25). The analysis of the physio- muscle fiber diameter) (34). Recently, a child with muscle hy-
logical contribution of the individual IGFBPs in the animal has pertrophy was found to have a loss-of-function mutation in the
been hampered by the redundancy in the IGFBP system (26). Mstn gene (36), although whether the child’s muscles have an
Nonetheless, muscle overloading is associated with increased increased number of fibers or a normal number of enlarged
abundance of IGFBP-4 and decreased abundance of IGFBP-5, myofibers remains to be established. These findings indicate
suggesting that these two isoforms may be involved in regulat- that inactivation of myostatin has similar effects in humans,
ing muscle adaptation to mechanical forces (27). Thus, whether mice, and cattle.
IGF-1 acts as an extracellular cue in muscle biology depends on After an enzymatic cleavage, the active myostatin peptide in-
a finely tuned and regulated series of events leading to either teracts with the membrane-bound type IIB activin receptor,
the stimulation or the inactivity of the IGF-1–IGFR axis. initiating a signaling cascade (likely mediated by the Smad tran-

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 3
REVIEW

scription factors) that ultimately blocks muscle differentiation, animals (54, 55). At the doses employed in those studies, val-
possibly by inhibiting the master regulatory genes MyoD (37) proic acid (a known teratogen) did not cause apparent toxicity
and Pax-3 (38), which encode proteins controlling skeletal myo- or malformations. In cultured cells, the effects of HDAC in-
genesis (39, 40). These findings raise the possibility that phar- hibitors on follistatin are restricted to skeletal muscle cells, be-
macological inhibition of myostatin may be of therapeutic value cause treatment with these inhibitors of mouse primary ker-
to promote muscle growth and differentiation in human diseases atinocytes and of other cell lines fated to differentiate toward
(41). Accordingly, antibody-mediated myostatin blockade effec- the adipose, bone, or pituitary phenotypes fails to activate Fst
tively ameliorates anatomical and physiological muscle parame- gene expression (51). These findings are relevant because they
ters in the mdx mouse (42). Consistent with these findings, suggest that systemic delivery of HDAC inhibitors may activate
knockout of the Mstn gene in the mdx mouse improves its dys- follistatin expression selectively in skeletal muscles. Iezzi et al.
trophic muscle phenotype (43). Whether down-regulation of (51) found that the effects exerted by HDAC inhibitors on
Mstn gene expression is involved in exercise-induced muscle skeletal muscle are due to an increased ability of undifferentiat-
hypertrophy remains to be firmly determined (44). Interestingly, ed myoblasts to be recruited and to fuse with already formed
Mstn-null mice have a substantial reduction in fat accumulation myotubes, and appear not to be mediated by either IGF-1 or
despite normal food intake. Furthermore, when Mstn-null mice IL-4 (56), two molecules involved in myoblast fusion and
were bred with two different models of obesity, agouti lethal growth. Follistatin overexpression in skeletal muscle cells can
yellow [A(y)] and obese [Lep(ob/ob)], the double-mutant mice fully recapitulate the morphological and biochemical modifica-
exhibited partly corrected glucose metabolism (45). Therefore, tions induced by the HDAC inhibitors, indicating the pivotal
pharmacological inactivation of myostatin may have the double role of follistatin in promoting myoblast accretion. Consistent
benefit of increasing muscle mass and retarding or preventing with these observations, blockade of follistatin, by either

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


the development of obesity and type II diabetes. functional inactivation with recombinant myostatin or RNA
Follistatin and deacetylase inhibitors. Follistatin is a secret- interference-meditated “knock-down” of follistatin, render
ed protein that interacts with and inhibits the activity of several HDAC inhibitors incapable of promoting myoblast accretion. A
TGF-β family members, including GDF-11 [also known as role for follistatin during skeletal muscle regeneration is indi-
bone morphogenetic protein 11 (BMP-11)] (46), a protein with cated by its expression in activated satellite cells during both
high sequence similarity to myostatin (47). Furthermore, myo- initial (51, 57) and later (51) stages of muscle regeneration.
statin circulates as a part of a latent complex containing myo- Systemic delivery of HDAC inhibitors in a mouse model of
statin propeptide and either follistatin-related gene (FLRG) or muscle degeneration and regeneration results in the activation
GDF-associated serum protein-1 (GASP-1), two molecules of markers, suggesting the existence of an active and sustained
closely related to follistatin (48, 49). Finally, follistatin interacts process of muscle regeneration. Increased muscle regeneration
with myostatin and reverses the inhibition exerted by the latter promoted by HDAC inhibitors requires the presence of activat-
on muscle differentiation of primary chick cells (38). Biochemi- ed satellite cells, indicating that the activity of HDAC inhibitors
cal experiments indicate that follistatin can block the binding of may be not only cell-type specific, but also limited to damaged
myostatin to the activin IIB receptor by interacting with the C- muscles (51).
terminal region of a myostatin dimer
(50). Mice overexpressing follistatin
under the control of muscle-specific
regulatory elements display a muscle
phenotype that recapitulates that ob-
served in the absence of myostatin
(50). Thus, follistatin counteracts the
biological activity of myostatin and
possibly of other TGF-β family mem-
bers.
Follistatin (Fst) gene expression is
activated by exposure of skeletal mus-
cle cells to deacetylase inhibitors (51),
a class of small, membrane-permeable
molecules that block the activity of Fig. 2. The deacetylase inhibitor trichostatin A induces an increase in both the diameter of
class I-II histone deacetylases differentiated myotubes and the number of myonuclei. Undifferentiated C2C12 skeletal my-
oblasts were either mock-treated (control) or exposed to 50 nM trichostatin A and allowed
(HDACs). Some of these molecules, in-
to differentiate in low growth factor–containing medium for 2 days before immunostaining
cluding valproic acid, are currently em- with an antibody to MHC.
ployed to treat various clinical condi-
tions such as epilepsy (52) and mood
disorders (53). Exposure of cultured skeletal muscle cells to In conclusion, an alternative method to blocking myostatin
HDAC inhibitors results in the formation of myotubes with in- activity to promote muscle growth may be by inducing Fst gene
creased diameter, increased number of myonuclei (Fig. 2), and expression with HDAC inhibitors. Although it is not known
enhanced abundance of muscle structural proteins such as whether exercise-induced muscle hypertrophy is associated with
myosins (54). Similarly, mouse embryos exposed to HDAC in- an increase of Fst expression, it is tempting to speculate that
hibitors display a premature formation of caudal somites and this might be the case, because Fst expression is observed in ac-
augmented muscle gene expression when compared to control tivated satellite cells (57).

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 4
REVIEW

Cell Determinants of Muscle Growth sibility that some satellite cells could enter terminal differentia-
Satellite cells. Satellite cells are lineage-committed adult stem tion and fuse with damaged muscle fibers after one cell division
cells (58), located between the basal lamina and the sarcolemma at most, thus evading the antiproliferative effect of radiation
of myofibers, that contribute to postnatal muscle growth (59) damage (82). Alternatively, some satellite cells may have sur-
and muscle regeneration after injury (60). Upon myotrauma, vived, given the existence of cell subpopulations resistant to ra-
quiescent satellite cells become activated, proliferate, and ulti- diation (83). It should also be possible to address the satellite
mately fuse to existing damaged muscle fibers or among them- cell requirement for the genesis of muscle hypertrophy in Pax7-
selves to form new myofibers (61). Satellite cells are activated deficent mice subjected to compensatory hypertrophy. In fact,
in response to hypertrophic stimuli, such as those occurring because Pax7-deficient mice lack satellite cells, any increase in
during muscle mechanical overload (59, 62, 63). In several muscle mass would have to be ascribed either to the expansion
animal models of compensatory hypertrophy (64–66) or after of other nonsatellite adult stem cells (see Nonsatellite Adult
resistance training in humans (67–69), the total number of Stem Cells, below) or to increased protein synthesis in existing
activated satellite cells is substantially increased. In most of myofibers.
these early works, activation, proliferation, and incorporation of Satellite cell activation. The mechanisms leading to satellite
satellite cells into myofibers was documented by electron mi- cell activation during muscle hypertrophy are not entirely un-
croscopy. Quiescent satellite cells are recognizable for their dis- derstood. It is postulated that extensive physical activity, such as
tinct location and morphological features, such as a very high resistance training or muscle overloading (chronic stretch, ago-
nuclear-to-cytoplasm ratio with few organelles and a smaller nist muscle ablation, tenotomy), inflicts muscle injury (84, 85)
nuclear size with increased amounts of heterochromatin com- that, similar to more severe muscle damage, may initiate a pro-
pared to fiber myonuclei (70). When activated, satellite cells are cess of regeneration. An indirect proof of muscle damage after

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


easily identified, because they appear as swellings on the my- mechanical stress is given by an increase of serum markers such
ofiber, have an increased cytoplasmic-to-nuclear ratio, and have as muscle creatine kinase, an enzyme that is usually restricted
increased numbers of intracellular organelles (61). Satellite to the myofiber cytosol (86). Muscle injury initiates an inflam-
cells have been the object of extensive studies aimed at the matory response with the attraction of nonmuscle mononucleat-
identification of specific molecular markers. Among the specif- ed cells, such as neutrophils and macrophages, into the dam-
ic satellite cell markers so far identified are Pax7, a paired-box- aged zone (87, 88), with consequent release of several growth
containing transcription factor essential for the specification of factors (by either the infiltrating cells or the damaged myofibers
satellite cells (71); myocyte nuclear factor (MNF, also called themselves). These growth factors may ultimately regulate
Foxk1), a winged-helix transcription factor; c-Met, the receptor satellite cell proliferation and differentiation (89). Indeed, sever-
for hepatocyte growth factor (HGF); M-cadherin, a calcium-de- al cytokines have been described that modulate proliferation
pendent cell adhesion molecule that is expressed only in a small and differentiation of satellite cells in vitro or during regenera-
subset of quiescent satellite cells; neural cell adhesion molecule tion after muscle injury (72, 90). HGF, which was initially de-
(NCAM) and vascular adhesion molecule-1 (VCAM-1); and tected in hepatic tissue, is considered to be a key regulator of
syndecan-3 and syndecan-4 [reviewed in (72)]. All these mark- satellite cell activity during muscle regeneration (91, 92). HGF
ers are present in either quiescent or proliferating satellite cells. is secreted by damaged tissue during the early phase of muscle
Upon exposure to signals emanating from the damaged area, regeneration in amounts proportional to the extent of muscle
quiescent satellite cells become activated and start proliferating injury (93, 94). It seems that HGF directly regulates satellite
(73). Once satellite cells are activated, they express the genes cell activation, as suggested by the presence of its receptor c-
encoding the two myogenic regulatory factors Myf 5 and Met in quiescent and activated satellite cells. Upon HGF bind-
MyoD, which are not detectable in quiescent cells (74). The ap- ing, the c-Met receptor tyrosine kinase transmits intracellular
pearance of MyoD temporally precedes any signs of cellular di- signals through a unique multidocking site at its C terminus that
vision, such as the expression of proliferative cell nuclear anti- is generated by autophosphorylation of two tyrosine residues.
gen (PCNA) (75). After proliferation, satellite cells initiate ex- The multisubstrate docking site mediates the binding of several
pression of the genes encoding myogenin, muscle regulatory adaptor proteins, such as Grb2, SHC, Crk (also known as
factor 4 (MRF4), cyclin-dependent kinase inhibitor p21, and, CRKL), PI3K, and the large adaptor protein Gab1. These adap-
after a permanent exit from the cell cycle, they fuse with dam- tor proteins then participate in transducing extracellular signals
aged myofibers or give rise to new myofibers (76, 77). Newly elicited by HGF to downstream targets (95). The PI3K-Akt
formed myofibers can be identified by the central location of pathway, for instance, mediates HGF-induced survival and pro-
myonuclei and expression of embryonic or developmental tection from apoptosis (96).
forms of myosin heavy chain (MHC) (78). In a process called Other secreted factors may also participate in satellite cell
self-renewal, a small proportion of satellite cells return to the activation. For example, fibroblast growth factor 6 (FGF-6) is
quiescent state to replenish the satellite cell pool. another potential regulator of satellite cell activity (97, 98).
Because satellite cells in their mitotic phase are sensitive to However, FGF-6-deficient mice generated by two different lab-
DNA damage, γ-irradiation has been used to impair their activa- oratories display different phenotypes during muscle regenera-
tion and proliferation and to test for their functional relevance tion. In one case, FGF-6-null animals have impaired satellite
in the development of skeletal muscle hypertrophy. Compen- cell proliferation and defects in muscle regeneration in response
satory hypertrophy can be largely prevented by muscle irradia- to a crush injury (99), whereas another study found no evidence
tion, providing compelling evidence that the presence of activat- of regenerative defects in response to several kinds of muscle
ed and proliferating satellite cells is indeed necessary for this damage (100). IL-4 is secreted by both myotubes and
process (79–81). However, compensatory hypertrophy is not macrophages present in the regenerating muscles. By promot-
completely prevented by muscle irradiation, suggesting the pos- ing fusion of myoblasts with preformed myotubes (56), IL-4

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 5
REVIEW

may actively participate in the regenerative process. Leukemia cle regeneration are currently being intensively pursued. In one
inhibitory factor (LIF), IL-6, IL-15, TNF-α, and several other study, multipotent progenitors of mesodermal tissues [termed
factors have been described as potentially involved in the mus- “mesoangioblasts” (120)] that express Flk1 (vascular
cle remodeling response after injury (101). However, a defini- endothelial growth factor–receptor 2), the key marker of
tive role for all these factors in regulating regeneration, hyper- angiopoietic progenitors, were intraarterially delivered into
trophy, or both remains to be further elucidated. α-sarcoglycan–null dystrophic mice. This resulted in restora-
As already described, a large body of evidence supports the tion of the α-sarcoglycan–dystrophin complex and, most impor-
importance of IGF-1 in the genesis of skeletal muscle hypertro- tantly, in complete functional muscle recovery of the dystrophic
phy. The expression of genes encoding two IGF-1 muscle- animals (121). Although of extreme biological and clinical rele-
specific isoforms, IGF-1E and MGF, is stimulated by mechani- vance, the discussion of these nonmuscle resident stem cells is
cal overload (102) with apparently different kinetics. MGF beyond the scope of this review. Novel populations of adult
seems to be expressed early after mechanical damage and to stem cells have been isolated from uninjured skeletal muscle
temporally precede the activation of satellite cells, whereas that express the CD45 and Sca1 (122, 123), two surface mark-
IGF-1E expression peaks later and is likely to be involved in ers expressed on hematopoietic stem cells. When cultured in
maintaining protein synthesis required to complete the muscle vitro, these stem cells form hematopoietic colonies but do not
repair (103). IGF-1 can promote both proliferation (104, 105) differentiate into muscle, with the exception of a small percent-
and differentiation of cultured satellite cells (106, 107), and age that can adopt the myogenic phenotype when cocultured
these findings have been confirmed in animal models. For in- with satellite cell-derived myoblasts (124). This population of
stance, both IGF-1–mediated muscle hypertrophy and compen- cells is clearly distinct from satellite cells, because they contain
satory hypertrophy are halved by γ-irradiation (108). Further- the CD45 and Sca1 markers, which are not present on satellite

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


more, IGF-1–induced muscle hypertrophy is accompanied by cells (124). Upon muscle injury, the number of cells in the
increased DNA content of myofibers (109, 110), appearance of CD45+:Sca1+ population increases approximately 10-fold and
centrally located nuclei, and appearance of developmental gives rise to myogenic cells (125). This suggests that a factor, or
forms of myosin (29, 111). All these findings support the in- factors, released by the injured or regenerating muscle may
volvement of satellite cells in IGF-1–mediated muscle hypertro- switch the fate of the CD45+:Sca1+ population and redirect it
phy. Further evidence for a role of satellite cells is suggested by from the hematopoietic to the myogenic phenotype. Indeed,
experiments in which muscle-localized expression of IGF-1 Polesskaya et al. (125) demonstrated that the expression of
prevented, through an increase of the regenerative potential of genes encoding selected members of the secreted Wnt protein
satellite cells, the aging-related loss of muscle mass (29). Final- family are induced in regenerating muscles. Consistent with
ly, satellite cells derived from mice overexpressing IGF-1 dis- this, ectopic expression of Wnts causes muscle commitment of
play an increased proliferative potential (112), which seems to CD45+:Sca1+ cells. The results of this study indicate that resi-
be mediated by activation of the PI3K-Akt pathway and down- dent, nonsatellite-derived progenitors can, in response to Wnt
regulation of the activity of cyclin-dependent inhibitor p27kip1 signaling, participate in muscle repair. Because mesoan-
(113), which results from an inhibitory effect on FOXO1 (114). gioblasts contain both Sca1 and Wnts (120), it should be infor-
Therefore, the molecular pathways activated by IGF-1 in the mative to compare the expression profiles of mesoangioblasts
muscle fibers to promote increased protein translation appear to and of muscle resident CD45+:Sca1+ cells to establish whether
also be activated in satellite cells. these cells have a common origin. Although these findings are
Satellite cells and aging. Aging is accompanied by a decline important in suggesting a potential clinical use of Wnt modula-
in muscle mass and strength, a phenomenon referred as to sar- tors to influence muscle regeneration, whether muscle-resident
copenia (115). It is clear that fitness is greater at any age in in- adult stem cells may participate in muscle hypertrophy remains
dividuals who exercise regularly versus those who do not and to be experimentally tested. Finally, it is interesting to note that
that sarcopenia is reduced—albeit not abolished—in physically IGF-1 has been reported to enhance the proliferation potential
active elderly people. A decline in the number of satellite cells, of a subpopulation of bone marrow stem cells, and their recruit-
their proliferative capacity, or both may contribute to sarcope- ment to sites of muscle damage, in lethally irradiated mice that
nia. One of the mechanisms responsible for the reduced regen- received bone marrow transplants (126). Thus, it is possible that
erative potential of old muscle seems to be the decline in Notch also adult stem cells may respond to IGF-1 signaling.
signaling (116), a pathway involved in the formation of several
tissues during embryogenesis. Notch signaling also contributes Conclusions and Future Directions
to satellite cell activation, proliferation, and cell lineage deter- It has been known for some time that muscle hypertrophy is the
mination (117). The regenerative potential of old muscle can be result of the activity both of molecules that stimulate growth of
experimentally restored either by forced activation of Notch sig- existing myofibers and of fiber accretion obtained through re-
naling or, as mentioned, by IGF-1. Whether physical exercise cruitment of activated satellite cells. Nonetheless, only recently
activates the Notch signaling pathway is not known. However, has a detailed description of the molecular and cellular mecha-
manipulations involving the Notch and IGF-1 pathways may re- nisms underlying postnatal muscle growth become available. It
sult in the activation and proliferation of satellite cells and may is now clear that the IGF-1 pathway is central to both increased
be useful in retarding age-related loss of muscle mass and protein synthesis and satellite cell activation. Similarly, the IGF-
strength. 1 pathway controls the activity of the FOXO transcription
Nonsatellite adult stem cells. In recent years, there have been factors. Under physiological conditions, IGF-1 prevents FOXOs
several reports indicating that cells deriving from bone marrow from activating the gene expression program leading to muscle
can contribute to the regeneration of injured muscle (118, 119). atrophy. It does not seem likely that the IGF-1 pathway is dedi-
The ontology and effectiveness of these cells in promoting mus- cated to controlling cell growth exclusively in skeletal muscle,

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 6
REVIEW

because overexpression of the Drosophila IRS, Akt, or p70S6K upregulates the gene expression of multiple ubiquitin ligases in skeletal
can cause hypertrophy in disparate cell lineages (127), includ- muscle. Int. J. Biochem. Cell Biol. 35, 698–705 (2003).
14. S. H. Lecker, R. T. Jagoe, A. Gilbert, M. Gomes, V. Baracos, J. Bailey,
ing those giving rise to the eye and wing (128). Rather, the au- S. R. Price, W. E. Mitch, A. L. Goldberg, Multiple types of skeletal mus-
tocrine or paracrine activity of locally produced IGF-1 may en- cle atrophy involve a common program of changes in gene expression.
sure skeletal muscle specificity. FASEB J. 18, 39–51 (2004).
15. T. N. Stitt, D. Drujan, B. A. Clarke, F. Panaro, Y. Timofeyva, W. O. Kline,
In several pathological settings, it would be desirable to ei- M. Gonzalez, G. D. Yancopoulos, D. J. Glass, The IGF-1/PI3K/Akt
ther prevent muscle atrophy or activate pathways that lead to in- pathway prevents expression of muscle atrophy-induced ubiquitin ligas-
creased muscle growth and that promote muscle hypertrophy. In es by inhibiting FOXO transcription factors. Mol. Cell 14, 395–403
(2004).
this respect, it is of interest to discuss the experimental use of 16. M. Sandri, C. Sandri, A. Gilbert, C. Skurk, E. Calabria, A. Picard, K.
small molecules—whose therapeutic efficacy is currently being Walsh, S. Schiaffino, S. H. Lecker, A. L. Goldberg, Foxo transcription
evaluated in clinical trials (129, 130)—that modulate the activi- factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause
ty of nuclear deacetylases. The transcriptional activity of FOXO skeletal muscle atrophy. Cell 117, 399–412 (2004).
17. H. Tran, A. Brunet, E. C. Griffith, M. E. Greenberg, The many forks in
proteins is regulated by Sir2, an NAD+-dependent deacetylase, FOXO’s road. Sci. STKE 2003, REre5 (2003).
with the expression of several FOXO targets being reduced by 18. D. C. Guttridge, M. W. Mayo, L. V. Madrid, C. Y. Wang, A. S. Baldwin,
Sir2 overexpression (131, 132). Because inhibition of Jr., NF-κB-induced loss of MyoD messenger RNA: Possible role in
muscle decay and cachexia. Science 289, 2363–2366 (2000).
FOXO-transcriptional activity is associated with reduced ex- 19. Y. P. Li, S. H. Lecker, Y. Chen, I. D. Waddell, A. L. Goldberg, M. B. Reid,
pression of MAFbx, it will be important to determine whether TNF-α increases ubiquitin-conjugating activity in skeletal muscle by up-
the Sir2 agonist resveratrol (133) is of therapeutic value in re- regulating UbcH2/E220k. FASEB J. 17, 1048–1057 (2003).
20. D. L. DeVol, P. Rotwein, J. L. Sadow, J. Novakofski, P. J. Bechtel, Acti-
ducing FOXO-dependent activation of the muscle atrophy pro- vation of insulin-like growth factor gene expression during work-induced
gram. Similarly, the use of class I-II deacetylase inhibitors has

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


skeletal muscle growth. Am. J. Physiol. 259, E89–E95 (1990).
given encouraging results in promoting muscle regeneration in 21. M. M. Bamman, J. R. Shipp, J. Jiang, B. A. Gower, G. R. Hunter, A.
Goodman, C. L. McLafferty Jr., R. J. Urban, Mechanical load increases
an animal model of muscle injury through activation of muscle IGF-I and androgen receptor mRNA concentrations in humans.
follistatin gene expression (51). Future experiments will deter- Am. J. Physiol. Endocrinol. Metab. 280, E383–E390 (2001).
mine whether these agents are effective in modulating the pro- 22. G. Goldspink, Cellular and molecular aspects of muscle growth, adap-
tation and ageing. Gerodontology 15, 35–43 (1998).
gression of muscle wasting in animal models of cancer, sepsis, 23. K. Sjogren, K. Wallenius, J. L. Liu, Y. M. Bohlooly, G. Pacini, L. Svens-
and degenerative diseases. son, J. Tornell, O. G. Isaksson, B. Ahren, J. O. Jansson, C. Ohlsson,
Liver-derived IGF-I is of importance for normal carbohydrate and lipid
metabolism. Diabetes 50, 1539–1545 (2001).
References 24. J. R. Florini, D. Z. Ewton, S. A. Coolican, Growth hormone and the
1. D. J. Glass, Molecular mechanisms modulating muscle mass. Trends insulin-like growth factor system in myogenesis. Endocr. Rev. 17,
Mol. Med. 9, 344–350 (2003). 481–517 (1996).
2. L. C. Cantley, The phosphoinositide 3-kinase pathway. Science 296, 25. D. Z. Ewton, S. A. Coolican, S. Mohan, S. D. Chernausek, J. R. Florini,
1655–1657 (2002). Modulation of insulin-like growth factor actions in L6A1 myoblasts by in-
3. K. M. Nicholson, N. G. Anderson, The protein kinase B/Akt signalling sulin-like growth factor binding protein (IGFBP)-4 and IGFBP-5: A dual
pathway in human malignancy. Cell. Signal. 14, 381–395 (2002). role for IGFBP-5. J. Cell. Physiol. 177, 47–57 (1998).
4. B. T. Nave, M. Ouwens, D. J. Withers, D. R. Alessi, P. R. Shepherd, 26. J. E. Pintar, A. Schuller, J. A. Cerro, M. Czick, A. Grewal, B. Green,
Mammalian target of rapamycin is a direct target for protein kinase B: Genetic ablation of IGFBP-2 suggests functional redundancy in the
Identification of a convergence point for opposing effects of insulin and IGFBP family. Prog. Growth Factor Res. 6, 437–445 (1995).
amino-acid deficiency on protein translation. Biochem. J. 344, 427–431 27. B. Awede, J. Thissen, P. Gailly, J. Lebacq, Regulation of IGF-I, IGFBP-4
(1999). and IGFBP-5 gene expression by loading in mouse skeletal muscle.
5. D. A. Cross, D. R. Alessi, P. Cohen, M. Andjelkovich, B. A. Hemmings, FEBS Lett. 461, 263–267 (1999).
Inhibition of glycogen synthase kinase-3 by insulin mediated by protein 28. M. E. Coleman, F. DeMayo, K. C. Yin, H. M. Lee, R. Geske, C. Mont-
kinase B. Nature 378, 785–789 (1995). gomery, R. J. Schwartz, Myogenic vector expression of insulin-like
6. C. Rommel, B. A. Clarke, S. Zimmermann, L. Nunez, R. Rossman, K. growth factor I stimulates muscle cell differentiation and myofiber hy-
Reid, K. Moelling, G. D. Yancopoulos, D. J. Glass, Differentiation stage- pertrophy in transgenic mice. J. Biol. Chem. 270, 12109–12116 (1995).
specific inhibition of the Raf-MEK-ERK pathway by Akt. Science 286, 29. A. Musaro, K. McCullagh, A. Paul, L. Houghton, G. Dobrowolny, M.
1738–1741 (1999). Molinaro, E. R. Barton, H. L. Sweeney, N. Rosenthal, Localized Igf-1
7. K. Hara, K. Yonezawa, M. T. Kozlowski, T. Sugimoto, K. Andrabi, Q. P. transgene expression sustains hypertrophy and regeneration in senes-
Weng, M. Kasuga, I. Nishimoto, J. Avruch, Regulation of eIF-4E BP1 cent skeletal muscle. Nat. Genet. 27, 195–200 (2001).
phosphorylation by mTOR. J. Biol. Chem. 272, 26457–26463 (1997). 30. E. R. Barton, L. Morris, A. Musaro, N. Rosenthal, H. L. Sweeney, Mus-
8. K. Hara, K. Yonezawa, Q. P. Weng, M. T. Kozlowski, C. Belham, J. cle-specific expression of insulin-like growth factor I counters muscle
Avruch, Amino acid sufficiency and mTOR regulate p70 S6 kinase and decline in mdx mice. J. Cell Biol. 157, 137–148 (2002).
eIF-4E BP1 through a common effector mechanism. J. Biol. Chem. 31. S. C. Bodine, T. N. Stitt, M. Gonzalez, W. O. Kline, G. L. Stover, R.
273, 14484–14494 (1998). Bauerlein, E. Zlotchenko, A. Scrimgeour, J. C. Lawrence, D. J. Glass,
9. S. C. Bodine, E. Latres, S. Baumhueter, V. K. Lai, L. Nunez, B. A. G. D. Yancopoulos, Akt/mTOR pathway is a crucial regulator of skeletal
Clarke, W. T. Poueymirou, F. J. Panaro, E. Na, K. Dharmarajan, Z. Q. muscle hypertrophy and can prevent muscle atrophy in vivo. Nat. Cell
Pan, D. M. Valenzuela, T. M. DeChiara, T. N. Stitt, G. D. Yancopoulos, Biol. 3, 1014–1019 (2001).
D. J. Glass, Identification of ubiquitin ligases required for skeletal mus- 32. K. Sakamoto, D. E. Arnolds, I. Ekberg, A. Thorell, L. J. Goodyear, Exer-
cle atrophy. Science 294, 1704–1708 (2001). cise regulates Akt and glycogen synthase kinase-3 activities in human
10. M. D. Gomes, S. H. Lecker, R. T. Jagoe, A. Navon, A. L. Goldberg, skeletal muscle. Biochem. Biophys. Res. Commun. 319, 419–425
Atrogin-1, a muscle-specific F-box protein highly expressed during (2004).
muscle atrophy. Proc. Natl. Acad. Sci. U.S.A. 98, 14440–14445 (2001). 33. A. Musaro, K. J. McCullagh, F. J. Naya, E. N. Olson, N. Rosenthal, IGF-
11. A. S. McElhinny, K. Kakinuma, H. Sorimachi, S. Labeit, C. C. Gregorio, 1 induces skeletal myocyte hypertrophy through calcineurin in associa-
Muscle-specific RING finger-1 interacts with titin to regulate sarcomeric tion with GATA-2 and NF-ATc1. Nature 400, 581–585 (1999).
M-line and thick filament structure and may have nuclear functions via 34. A. C. McPherron, A. M. Lawler, S. J. Lee, Regulation of skeletal muscle
its interaction with glucocorticoid modulatory element binding protein-1. mass in mice by a new TGF-β superfamily member. Nature 387, 83–90
J. Cell Biol. 157, 125–136 (2002). (1997).
12. J. M. Sacheck, A. Ohtsuka, S. C. McLary, A. L. Goldberg, IGF-1 stimu- 35. A. C. McPherron, S. J. Lee, Double muscling in cattle due to mutations
lates muscle growth by suppressing protein breakdown and expression in the myostatin gene. Proc. Natl. Acad. Sci. U.S.A. 94, 12457–12461
of atrophy-related ubiquitin-ligases, atrogin-1 and MuRF1. Am. J. Phys- (1997).
iol. Endocrinol. Metab. (2004). 36. M. Schuelke, K. R. Wagner, L. E. Stolz, C. Hubner, T. Riebel, W.
13. C. J. Wray, J. M. Mammen, D. D. Hershko, P. O. Hasselgren, Sepsis Komen, T. Braun, J. F. Tobin, S. J. Lee, Myostatin mutation associated

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 7
REVIEW

with gross muscle hypertrophy in a child. N. Engl. J. Med. 350, cells during compensatory muscle hypertrophy. Virchows Arch. B Cell
2682–2688 (2004). Pathol. 21, 113–118 (1976).
37. B. Langley, M. Thomas, A. Bishop, M. Sharma, S. Gilmour, R. Kam- 64. V. Hanzlikova, E. V. Mackova, P. Hnik, Satellite cells of the rat soleus
badur, Myostatin inhibits myoblast differentiation by down-regulating muscle in the process of compensatory hypertrophy combined with
MyoD expression. J. Biol. Chem. 277, 49831–49840 (2002). denervation. Cell Tissue Res. 160, 411–421 (1975).
38. H. Amthor, G. Nicholas, I. McKinnell, C. F. Kemp, M. Sharma, R. Kam- 65. M. H. Snow, Satellite cell response in rat soleus muscle undergoing hy-
badur, K. Patel, Follistatin complexes myostatin and antagonises myo- pertrophy due to surgical ablation of synergists. Anat. Rec. 227,
statin-mediated inhibition of myogenesis. Dev. Biol. 270, 19–30 (2004). 437–446 (1990).
39. M. Maroto, R. Reshef, A. E. Munsterberg, S. Koester, M. Goulding, A. 66. M. Ishido, K. Kami, M. Masuhara, Localization of MyoD, myogenin and
B. Lassar, Ectopic Pax-3 activates MyoD and Myf-5 expression in em- cell cycle regulatory factors in hypertrophying rat skeletal muscles. Acta
bryonic mesoderm and neural tissue. Cell 89, 139–148 (1997). Physiol. Scand. 180, 281–289 (2004).
40. S. Tajbakhsh, D. Rocancourt, G. Cossu, M. Buckingham, Redefining 67. F. Kadi, A. Eriksson, S. Holmner, G. S. Butler-Browne, L. E. Thornell,
the genetic hierarchies controlling skeletal myogenesis: Pax-3 and Cellular adaptation of the trapezius muscle in strength-trained athletes.
Myf-5 act upstream of MyoD. Cell 89, 127–138 (1997). Histochem. Cell Biol. 111, 189–195 (1999).
41. T. S. Khurana, K. E. Davies, Pharmacological strategies for muscular 68. F. Kadi, Adaptation of human skeletal muscle to training and anabolic
dystrophy. Nat. Rev. Drug Discov. 2, 379–390 (2003). steroids. Acta Physiol. Scand. Suppl. 646, 1–52 (2000).
42. S. Bogdanovich, T. O. Krag, E. R. Barton, L. D. Morris, L. A. Whitte- 69. S. M. Roth, G. F. Martel, F. M. Ivey, J. T. Lemmer, B. L. Tracy, E. J. Met-
more, R. S. Ahima, T. S. Khurana, Functional improvement of dystroph- ter, B. F. Hurley, M. A. Rogers, Skeletal muscle satellite cell characteris-
ic muscle by myostatin blockade. Nature 420, 418–421 (2002). tics in young and older men and women after heavy resistance strength
43. K. R. Wagner, A. C. McPherron, N. Winik, S. J. Lee, Loss of myostatin training. J. Gerontol. A Biol. Sci. Med. Sci. 56, B240–B247 (2001).
attenuates severity of muscular dystrophy in mdx mice. Ann. Neurol. 70. E. Schultz, M. C. Gibson, T. Champion, Satellite cells are mitotically
52, 832–836 (2002). quiescent in mature mouse muscle: An EM and radioautographic study.
44. K. S. Walker, R. Kambadur, M. Sharma, H. K. Smith, Resistance train- J. Exp. Zool. 206, 451–456 (1978).
ing alters plasma myostatin but not IGF-1 in healthy men. Med. Sci. 71. P. Seale, L. A. Sabourin, A. Girgis-Gabardo, A. Mansouri, P. Gruss, M.
Sports Exerc. 36, 787–793 (2004). A. Rudnicki, Pax7 is required for the specification of myogenic satellite

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


45. A. C. McPherron, S. J. Lee, Suppression of body fat accumulation in cells. Cell 102, 777–786 (2000).
myostatin-deficient mice. J. Clin. Invest. 109, 595–601 (2002). 72. S. B. Charge, M. A. Rudnicki, Cellular and molecular regulation of mus-
46. L. W. Gamer, N. M. Wolfman, A. J. Celeste, G. Hattersley, R. Hewick, V. cle regeneration. Physiol. Rev. 84, 209–238 (2004).
Rosen, A novel BMP expressed in developing mouse limb, spinal cord, 73. E. Schultz, D. L. Jaryszak, C. R. Valliere, Response of satellite cells to
and tail bud is a potent mesoderm inducer in Xenopus embryos. Dev. focal skeletal muscle injury. Muscle Nerve 8, 217–222 (1985).
Biol. 208, 222–232 (1999). 74. R. N. Cooper, S. Tajbakhsh, V. Mouly, G. Cossu, M. Buckingham, G. S.
47. A. C. McPherron, A. M. Lawler, S. J. Lee, Regulation of anterior/posteri- Butler-Browne, In vivo satellite cell activation via Myf5 and MyoD in re-
or patterning of the axial skeleton by growth/differentiation factor 11. generating mouse skeletal muscle. J. Cell Sci. 112, 2895–2901 (1999).
Nat. Genet. 22, 260–264 (1999). 75. C. K. Smith 2nd, M. J. Janney, R. E. Allen, Temporal expression of
48. J. J. Hill, M. V. Davies, A. A. Pearson, J. H. Wang, R. M. Hewick, N. M. myogenic regulatory genes during activation, proliferation, and differen-
Wolfman, Y. Qiu, The myostatin propeptide and the follistatin-related tiation of rat skeletal muscle satellite cells. J. Cell. Physiol. 159,
gene are inhibitory binding proteins of myostatin in normal serum. J. Bi- 379–385 (1994).
ol. Chem. 277, 40735–40741 (2002). 76. D. D. Cornelison, B. J. Wold, Single-cell analysis of regulatory gene ex-
49. J. J. Hill, Y. Qiu, R. M. Hewick, N. M. Wolfman, Regulation of myostatin pression in quiescent and activated mouse skeletal muscle satellite
in vivo by growth and differentiation factor-associated serum protein-1: cells. Dev. Biol. 191, 270–283 (1997).
A novel protein with protease inhibitor and follistatin domains. Mol. En- 77. T. A. Robertson, J. M. Papadimitriou, M. D. Grounds, Fusion of myo-
docrinol. 17, 1144–1154 (2003). genic cells to the newly sealed region of damaged myofibres in skeletal
50. S. J. Lee, A. C. McPherron, Regulation of myostatin activity and muscle muscle regeneration. Neuropathol. Appl. Neurobiol. 19, 350–358
growth. Proc. Natl. Acad. Sci. U.S.A. 98, 9306–9311 (2001). (1993).
51. S. Iezzi, M. Di Padova, C. Serra, G. Caretti, C. Simone, E. Maklan, G. 78. R. G. Whalen, J. B. Harris, G. S. Butler-Browne, S. Sesodia, Expres-
Minetti, P. Zhao, E. P. Hoffman, P. L. Puri, V. Sartorelli, Deacetylase in- sion of myosin isoforms during notexin-induced regeneration of rat
hibitors increase muscle cell size by promoting myoblast recruitment soleus muscles. Dev. Biol. 141, 24–40 (1990).
and fusion through induction of follistatin. Dev. Cell 6, 673–684 (2004). 79. J. D. Rosenblatt, D. Yong, D. J. Parry, Satellite cell activity is required
52. C. L. Deckers, Y. A. Hekster, A. Keyser, H. J. van Lier, H. Meinardi, W. for hypertrophy of overloaded adult rat muscle. Muscle Nerve 17,
O. Renier, Monotherapy versus polytherapy for epilepsy: A multicenter 608–613 (1994).
double-blind randomized study. Epilepsia 42, 1387–1394 (2001). 80. J. N. Phelan, W. J. Gonyea, Effect of radiation on satellite cell activity
53. T. Puzantian, Criteria for use of valproate in adult psychiatric inpatients and protein expression in overloaded mammalian skeletal muscle.
and outpatients. Am. J. Health Syst. Pharm. 53, 1187–1188 (1996). Anat. Rec. 247, 179–188 (1997).
54. S. Iezzi, G. Cossu, C. Nervi, V. Sartorelli, P. L. Puri, Stage-specific 81. G. R. Adams, V. J. Caiozzo, F. Haddad, K. M. Baldwin, Cellular and
modulation of skeletal myogenesis by inhibitors of nuclear deacety- molecular responses to increased skeletal muscle loading after
lases. Proc. Natl. Acad. Sci. U.S.A. 99, 7757–7762 (2002). irradiation. Am. J. Physiol. Cell Physiol. 283, C1182–C1195 (2002).
55. C. Nervi, U. Borello, F. Fazi, V. Buffa, P. G. Pelicci, G. Cossu, Inhibition 82. J. K. McGeachie, M. D. Grounds, Initiation and duration of muscle pre-
of histone deacetylase activity by trichostatin A modulates gene expres- cursor replication after mild and severe injury to skeletal muscle of
sion during mouse embryogenesis without apparent toxicity. Cancer mice. An autoradiographic study. Cell Tissue Res. 248, 125–130
Res. 61, 1247–1249 (2001). (1987).
56. V. Horsley, K. M. Jansen, S. T. Mills, G. K. Pavlath, IL-4 Acts as a My- 83. L. Heslop, J. E. Morgan, T. A. Partridge, Evidence for a myogenic stem
oblast recruitment factor during mammalian muscle growth. Cell 113, cell that is exhausted in dystrophic muscle. J. Cell Sci. 113, 2299–2308
483–494 (2003). (2000).
57. A. S. Armand, B. Della Gaspera, T. Launay, F. Charbonnier, C. L. Gal- 84. J. A. Faulkner, S. V. Brooks, J. A. Opiteck, Injury to skeletal muscle
lien, C. Chanoine, Expression and neural control of follistatin versus fibers during contractions: Conditions of occurrence and prevention.
myostatin genes during regeneration of mouse soleus. Dev. Dyn. 227, Phys. Ther. 73, 911–921 (1993).
256–265 (2003). 85. R. B. Armstrong, G. L. Warren, J. A. Warren, Mechanisms of exercise-
58. A. Mauro, W. R. Adams, The structure of the sarcolemma of the frog induced muscle fibre injury. Sports Med. 12, 184–207 (1991).
skeletal muscle fiber. J. Biophys. Biochem. Cytol. 10 (suppl.), 177–185 86. S. Sorichter, J. Mair, A. Koller, E. Muller, C. Kremser, W. Judmaier, C.
(1961). Haid, C. Calzolari, B. Puschendorf, Creatine kinase, myosin heavy
59. F. P. Moss, C. P. Leblond, Satellite cells as the source of nuclei in mus- chains and magnetic resonance imaging after eccentric exercise. J.
cles of growing rats. Anat. Rec. 170, 421–435 (1971). Sports Sci. 19, 687–691 (2001).
60. L. A. Sabourin, M. A. Rudnicki, The molecular regulation of myogene- 87. R. A. Fielding, T. J. Manfredi, W. Ding, M. A. Fiatarone, W. J. Evans, J.
sis. Clin. Genet. 57, 16–25 (2000). G. Cannon, Acute phase response in exercise. III. Neutrophil and IL-1β
61. E. Schultz, K. M. McCormick, Skeletal muscle satellite cells. Rev. Phys- accumulation in skeletal muscle. Am. J. Physiol. 265, R166–R172
iol. Biochem. Pharmacol. 123, 213–257 (1994). (1993).
62. K. C. Darr, E. Schultz, Exercise-induced satellite cell activation in grow- 88. D. L. MacIntyre, S. Sorichter, J. Mair, A. Berg, D. C. McKenzie, Markers
ing and mature skeletal muscle. J. Appl. Physiol. 63, 1816–1821 of inflammation and myofibrillar proteins following eccentric exercise in
(1987). humans. Eur. J. Appl. Physiol. 84, 180–186 (2001).
63. S. Schiaffino, S. P. Bormioli, M. Aloisi, The fate of newly formed satellite 89. T. J. Hawke, D. J. Garry, Myogenic satellite cells: Physiology to molecu-

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 8
REVIEW

lar biology. J. Appl. Physiol. 91, 534–551 (2001). skeletal muscle satellite cells by enhancing G1/S cell cycle progression
90. J. Vierck, B. O’Reilly, K. Hossner, J. Antonio, K. Byrne, L. Bucci, M. via the activation of phosphatidylinositol 3′-kinase/Akt signaling
Dodson, Satellite cell regulation following myotrauma caused by resis- pathway. J. Biol. Chem. 275, 35942–35952 (2000).
tance exercise. Cell Biol. Int. 24, 263–272 (2000). 114. S. Machida, E. E. Spangenburg, F. W. Booth, Forkhead transcription
91. R. E. Allen, S. M. Sheehan, R. G. Taylor, T. L. Kendall, G. M. Rice, Hep- factor FoxO1 transduces insulin-like growth factor’s signal to p27Kip1 in
atocyte growth factor activates quiescent skeletal muscle satellite cells primary skeletal muscle satellite cells. J. Cell. Physiol. 196, 523–531
in vitro. J. Cell. Physiol. 165, 307–312 (1995). (2003).
92. R. Bischoff, Chemotaxis of skeletal muscle satellite cells. Dev. Dyn. 115. T. J. Marcell, Sarcopenia: Causes, consequences, and preventions. J.
208, 505–515 (1997). Gerontol. A Biol. Sci. Med. Sci. 58, M911–M916 (2003).
93. S. M. Sheehan, R. E. Allen, Skeletal muscle satellite cell proliferation in 116. I. M. Conboy, M. J. Conboy, G. M. Smythe, T. A. Rando, Notch-mediat-
response to members of the fibroblast growth factor family and hepato- ed restoration of regenerative potential to aged muscle. Science 302,
cyte growth factor. J. Cell. Physiol. 181, 499–506 (1999). 1575–1577 (2003).
94. R. Tatsumi, J. E. Anderson, C. J. Nevoret, O. Halevy, R. E. Allen, 117. I. M. Conboy, T. A. Rando, The regulation of Notch signaling controls
HGF/SF is present in normal adult skeletal muscle and is capable of satellite cell activation and cell fate determination in postnatal
activating satellite cells. Dev. Biol. 194, 114–128 (1998). myogenesis. Dev. Cell 3, 397–409 (2002).
95. K. A. Furge, Y. W. Zhang, G. F. Vande Woude, Met receptor tyrosine 118. G. Ferrari, G. Cusella-De Angelis, M. Coletta, E. Paolucci, A. Stornaiuo-
kinase: Enhanced signaling through adapter proteins. Oncogene 19, lo, G. Cossu, F. Mavilio, Muscle regeneration by bone marrow-derived
5582–5589 (2000). myogenic progenitors. Science 279, 1528–1530 (1998).
96. G. H. Xiao, M. Jeffers, A. Bellacosa, Y. Mitsuuchi, G. F. Vande Woude, 119. M. A. LaBarge, H. M. Blau, Biological progression from adult bone mar-
J. R. Testa, Anti-apoptotic signaling by hepatocyte growth factor/Met via row to mononucleate muscle stem cell to multinucleate muscle fiber in
the phosphatidylinositol 3-kinase/Akt and mitogen-activated protein ki- response to injury. Cell 111, 589–601 (2002).
nase pathways. Proc. Natl. Acad. Sci. U.S.A. 98, 247–252 (2001). 120. G. Cossu, P. Bianco, Mesoangioblasts–vascular progenitors for ex-
97. S. Dusterhoft, D. Pette, Evidence that acidic fibroblast growth factor travascular mesodermal tissues. Curr. Opin. Genet. Dev. 13, 537–542
promotes maturation of rat satellite-cell-derived myotubes in vitro. (2003).
Differentiation 65, 161–169 (1999). 121. M. Sampaolesi, Y. Torrente, A. Innocenzi, R. Tonlorenzi, G. D’Antona,

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


98. S. Kastner, M. C. Elias, A. J. Rivera, Z. Yablonka-Reuveni, Gene ex- M. A. Pellegrino, R. Barresi, N. Bresolin, M. G. De Angelis, K. P. Camp-
pression patterns of the fibroblast growth factors and their receptors bell, R. Bottinelli, G. Cossu, Cell therapy of α-sarcoglycan null dys-
during myogenesis of rat satellite cells. J. Histochem. Cytochem. 48, trophic mice through intra-arterial delivery of mesoangioblasts. Science
1079–1096 (2000). 301, 487–492 (2003).
99. T. Floss, H. H. Arnold, T. Braun, A role for FGF-6 in skeletal muscle 122. E. Gussoni, Y. Soneoka, C. D. Strickland, E. A. Buzney, M. K. Khan, A.
regeneration. Genes Dev. 11, 2040–2051 (1997). F. Flint, L. M. Kunkel, R. C. Mulligan, Dystrophin expression in the mdx
100. F. Fiore, A. Sebille, D. Birnbaum, Skeletal muscle regeneration is not mouse restored by stem cell transplantation. Nature 401, 390–394
impaired in Fgf6−/− mutant mice. Biochem. Biophys. Res. Commun. (1999).
272, 138–143 (2000). 123. K. A. Jackson, T. Mi, M. A. Goodell, Hematopoietic potential of stem
101. I. Husmann, L. Soulet, J. Gautron, I. Martelly, D. Barritault, Growth fac- cells isolated from murine skeletal muscle. Proc. Natl. Acad. Sci. U.S.A.
tors in skeletal muscle regeneration. Cytokine Growth Factor Rev. 7, 96, 14482–14486 (1999).
249–258 (1996). 124. A. Asakura, P. Seale, A. Girgis-Gabardo, M. A. Rudnicki, Myogenic
102. G. McKoy, W. Ashley, J. Mander, S. Y. Yang, N. Williams, B. Russell, G. specification of side population cells in skeletal muscle. J. Cell Biol.
Goldspink, Expression of insulin growth factor-1 splice variants and 159, 123–134 (2002).
structural genes in rabbit skeletal muscle induced by stretch and 125. A. Polesskaya, P. Seale, M. A. Rudnicki, Wnt signaling induces the
stimulation. J. Physiol. 516, 583–592 (1999). myogenic specification of resident CD45+ adult stem cells during mus-
103. M. Hill, A. Wernig, G. Goldspink, Muscle satellite (stem) cell activation cle regeneration. Cell 113, 841–852 (2003).
during local tissue injury and repair. J. Anat. 203, 89–99 (2003). 126. A. Musaro, C. Giacinti, G. Borsellino, G. Dobrowolny, L. Pelosi, L.
104. M. E. Doumit, D. R. Cook, R. A. Merkel, Fibroblast growth factor, epi- Cairns, S. Ottolenghi, G. Cossu, G. Bernardi, L. Battistini, M. Molinaro,
dermal growth factor, insulin-like growth factors, and platelet-derived N. Rosenthal, Stem cell-mediated muscle regeneration is enhanced by
growth factor-BB stimulate proliferation of clonally derived porcine myo- local isoform of insulin-like growth factor 1. Proc. Natl. Acad. Sci. U.S.A.
genic satellite cells. J. Cell. Physiol. 157, 326–332 (1993). 101, 1206–1210 (2004).
105. R. E. Allen, L. K. Boxhorn, Regulation of skeletal muscle satellite cell 127. R. Bohni, J. Riesgo-Escovar, S. Oldham, W. Brogiolo, H. Stocker, B. F.
proliferation and differentiation by transforming growth factor-β, insulin- Andruss, K. Beckingham, E. Hafen, Autonomous control of cell and or-
like growth factor I, and fibroblast growth factor. J. Cell. Physiol. 138, gan size by CHICO, a Drosophila homolog of vertebrate IRS1-4. Cell
311–315 (1989). 97, 865–875 (1999).
106. S. A. Coolican, D. S. Samuel, D. Z. Ewton, F. J. McWade, J. R. Florini, 128. S. J. Leevers, D. Weinkove, L. K. MacDougall, E. Hafen, M. D. Water-
The mitogenic and myogenic actions of insulin-like growth factors utilize field, The Drosophila phosphoinositide 3-kinase Dp110 promotes cell
distinct signaling pathways. J. Biol. Chem. 272, 6653–6662 (1997). growth. EMBO J. 15, 6584–6594 (1996).
107. S. Adi, B. Bin-Abbas, N. Y. Wu, S. M. Rosenthal, Early stimulation and late 129. M. H. Aziz, R. Kumar, N. Ahmad, Cancer chemoprevention by resvera-
inhibition of extracellular signal-regulated kinase 1/2 phosphorylation by trol: In vitro and in vivo studies and the underlying mechanisms. Int. J.
IGF-I: A potential mechanism mediating the switch in IGF-I action on skeletal Oncol. 23, 17–28 (2003).
muscle cell differentiation. Endocrinology 143, 511–516 (2002). 130. J. P. Secrist, X. Zhou, V. M. Richon, HDAC inhibitors for the treatment
108. E. R. Barton-Davis, D. I. Shoturma, H. L. Sweeney, Contribution of of cancer. Curr. Opin. Investig. Drugs 4, 1422–1427 (2003).
satellite cells to IGF-I induced hypertrophy of skeletal muscle. Acta 131. M. C. Motta, N. Divecha, M. Lemieux, C. Kamel, D. Chen, W. Gu, Y.
Physiol. Scand. 167, 301–305 (1999). Bultsma, M. McBurney, L. Guarente, Mammalian SIRT1 represses fork-
109. G. R. Adams, S. A. McCue, Localized infusion of IGF-I results in skele- head transcription factors. Cell 116, 551–563 (2004).
tal muscle hypertrophy in rats. J. Appl. Physiol. 84, 1716–1722 (1998). 132. A. Brunet, L. B. Sweeney, J. F. Sturgill, K. F. Chua, P. L. Greer, Y. Lin,
110. M. L. Fiorotto, R. J. Schwartz, M. C. Delaughter, Persistent IGF-I over- H. Tran, S. E. Ross, R. Mostoslavsky, H. Y. Cohen, L. S. Hu, H. L.
expression in skeletal muscle transiently enhances DNA accretion and Cheng, M. P. Jedrychowski, S. P. Gygi, D. A. Sinclair, F. W. Alt, M. E.
growth. FASEB J. 17, 59–60 (2003). Greenberg, Stress-dependent regulation of FOXO transcription factors
111. E. R. Barton-Davis, D. I. Shoturma, A. Musaro, N. Rosenthal, H. L. by the SIRT1 deacetylase. Science 303, 2011–2015 (2004).
Sweeney, Viral mediated expression of insulin-like growth factor I 133. K. T. Howitz, K. J. Bitterman, H. Y. Cohen, D. W. Lamming, S. Lavu, J.
blocks the aging-related loss of skeletal muscle function. Proc. Natl. G. Wood, R. E. Zipkin, P. Chung, A. Kisielewski, L. L. Zhang, B. Scher-
Acad. Sci. U.S.A. 95, 15603–15607 (1998). er, D. A. Sinclair, Small molecule activators of sirtuins extend Saccha-
112. M. V. Chakravarthy, B. S. Davis, F. W. Booth, IGF-I restores satellite romyces cerevisiae lifespan. Nature 425, 191–196 (2003).
cell proliferative potential in immobilized old skeletal muscle. J. Appl.
Physiol. 89, 1365–1379 (2000).
113. M. V. Chakravarthy, T. W. Abraha, R. J. Schwartz, M. L. Fiorotto, F. W. Citation: V. Sartorelli, M. Fulco, Molecular and cellular determinants of
Booth, Insulin-like growth factor-I extends in vitro replicative life span of skeletal muscle atrophy and hypertrophy. Sci. STKE 2004, re11 (2004).

www.stke.org/cgi/content/full/sigtrans;2004/244/re11 Page 9
Molecular and Cellular Determinants of Skeletal Muscle Atrophy
and Hypertrophy
Vittorio Sartorelli and Marcella Fulco (July 27, 2004)
Science Signaling 2004 (244), re11. [doi: 10.1126/stke.2442004re11]

The following resources related to this article are available online at http://stke.sciencemag.org.
This information is current as of June 21, 2015.

Article Tools Visit the online version of this article to access the personalization and article
tools:
http://stke.sciencemag.org/content/2004/244/re11

Related Content The editors suggest related resources on Science's sites:


http://sageke.sciencemag.org/cgi/content/full/2003/46/pe31

Downloaded from http://stke.sciencemag.org/ on June 21, 2015


http://stke.sciencemag.org/content
http://www.sciencemag.org/content/sci/305/5684/631.full.html
http://stke.sciencemag.org/content/sigtrans/3/152/pe49.full.html
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://sageke.sciencemag.org/cgi/content/full/2003/48/pe34
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content
http://stke.sciencemag.org/content/sigtrans/2003/212/re15.full.html
http://stke.sciencemag.org/content

References This article cites 132 articles, 48 of which you can access for free at:
http://stke.sciencemag.org/content/2004/244/re11#BIBL

Glossary Look up definitions for abbreviations and terms found in this article:
http://stke.sciencemag.org/cgi/glossarylookup

Permissions Obtain information about reproducing this article:


http://www.sciencemag.org/about/permissions.dtl

Science Signaling (ISSN 1937-9145) is published weekly, except the last December, by the American
Association for the Advancement of Science, 1200 New York Avenue, NW, Washington, DC 20005.
Copyright 2015 by the American Association for the Advancement of Science; all rights reserved.

You might also like