You are on page 1of 12

Critical Review

Myostatin: Expanding Horizons Mridula Sharma1,2*


Craig McFarlane2
Ravi Kambadur2,3
Himani Kukreti1
Sabeera Bonala2
Shruti Srinivasan1

1
Department of Biochemistry, Yong Loo Lin School of Medicine, National
University of Singapore (NUS), Singapore
2
Department of Cell & Molecular Biology, Brenner Centre for Molecular
Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
3
School of Biological Sciences, Nanyang Technological University,
Singapore

Abstract
Myostatin is a secreted growth and differentiation factor that revelations regarding myostatin regulation also offer mecha-
belongs to the TGF-b superfamily. Myostatin is predominantly nisms that could be exploited for developing myostatin antag-
synthesized and expressed in skeletal muscle and thus exerts onists. Increasingly, it is becoming clearer that besides its
a huge impact on muscle growth and function. In keeping with conventional role in muscle, myostatin plays a critical role in
its negative role in myogenesis, myostatin expression is metabolism. Hence, molecular mechanisms by which myosta-
tightly regulated at several levels including epigenetic, tran- tin regulates several key metabolic processes need to be fur-
scriptional, post-transcriptional, and post-translational. New ther explored. V
C 2015 IUBMB Life, 67(8):589–600, 2015

Keywords: myostatin; skeletal muscle; muscle wasting; cancer


cachexia; sarcopenia; insulin resistance

Introduction repair (6–9). Furthermore, high levels of myostatin are linked


with muscle wasting seen during cancer (10), HIV infection
Myostatin, earlier known as growth and differentiation factor
(11), liver disease (12), COPD (13), and during aging (14,15).
8 (GDF-8), is a member of the TGF-b superfamily. Myostatin
Moreover, systemic expression of myostatin in mice has been
has a tremendous impact on the growth and development of shown to induce cachectic-like muscle wasting (16). In addi-
skeletal muscle, such that genetic deletions or mutations in the tion to regulating muscle growth, more recently myostatin has
myostatin gene cause a dramatic increase in skeletal muscle been shown to affect glucose and fat metabolism (1), and as
mass. The muscle hypertrophy phenotype in the absence of such is associated with the development of obesity, insulin
myostatin is present in various mammalian species including resistance, and type 2 diabetes (T2D; (17–20)).
rodents, farm and domestic animals and humans, thus under- In this review article, we discuss the recent developments
scoring the important function of myostatin in muscle (1–5). in our understanding of myostatin regulation and function.
Further evidence for the negative role that myostatin plays
during myogenesis has been observed in several studies that
have assessed myostatin function during muscle growth and
Myostatin Structure and Processing
Similar to other TGF-b superfamily members myostatin is syn-
C 2015 International Union of Biochemistry and Molecular Biology
V
thesised as a precursor protein (375 amino acids) bearing an
Volume 67, Number 8, August 2015, Pages 589–600
N-terminal (NH2) core of hydrophobic amino acids, which acts
Address correspondence to: Mridula Sharma, National University of Singa-
as a signal sequence for secretion ((3); Fig. 1). The C-terminal
pore, Department of Biochemistry, 8 Medical Drive, MD7, Singapore
117597. (COOH) region of myostatin has nine conserved cysteine amino
E-mail: bchmridu@nus.edu.sg acids that are used for homodimerisation and formation of the
Received 20 May 2015; Accepted 29 May 2015 “cysteine knot” structure, a hallmark feature of TGF-b super-
DOI 10.1002/iub.1392 family members ((3,21); Fig. 1). The proteolytic processing of
Published online 25 August 2015 in Wiley Online Library 52 kDa myostatin precursor protein occurs in the Golgi at the
(wileyonlinelibrary.com) RSRR site by the action of the serine protease furin or other

IUBMB Life 589


IUBMB LIFE

Loop-Helix (bHLH) transcription factors, such as MyoD, Myf5


and myogenin. The myostatin promoter is preferentially acti-
vated by MyoD, with peak activity observed during the G1
phase of the cell cycle when MyoD is expressed maximally.
Thus, MyoD activates the myostatin promoter in a temporal
fashion, presumably to allow for timely myostatin-mediated
control of cell cycle arrest (31).
Mef2 transcription factors play an integral role in the tran-
scription of several muscle specific genes during myoblast dif-
Myostatin structure and proteolytic processing. Sche- ferentiation. Putative mef2 binding motifs have been observed
FIG 1 matic representation of the structure of myostatin in the myostatin promoter; however, the number of mef2
protein. Precursor myostatin protein contains an N- motifs is variable across different species (31–34). A report by
terminal (NH2) signal peptide (SP) for secretion and
Wang et al., (2008) indicated that in rat cardiomyocytes,
a C-terminal (COOH) region that contains nine con-
served cysteine residues (c), which are critical for angiotensin II-induced myostatin expression is via mef2 and
eventual homodimerisation and “cysteine knot” for- that this increase in myostatin is believed to inhibit cardiac
mation. Precursor myostatin protein is proteolytically myocyte hypertrophy (35). Mef2 has also been shown to
processed by the serine protease Furin at the Golgi increase myostatin promoter activity in myoblasts, which was
to give rise to LAP and mature myostatin.
proposed to be a potential mechanism through which Mef2
may restrain muscle growth and differentiation (33).
members of the proprotein convertase family ((3,22,23); Fig. Several FoxO binding motifs have also been observed in
1). As a result of the enzymatic reaction, a 36/40 kDa Latency- the myostatin promoter suggesting that the FoxO family of
Associated Peptide (LAP) and a 12.5/26 kDa mature peptide, transcription factors may control myostatin transcription.
corresponding to a C-terminal monomer or dimer respectively, Indeed, type IIB, glycolytic muscle fibers express higher levels
have been observed ((9,22), (24); Fig. 1). Both mature and LAP of myostatin and FoxO1 mRNAs than type I oxidative fibers
forms of myostatin are secreted into the circulation (22,25), (36). Furthermore, an additive effect on the regulation of myo-
allowing myostatin to function in an autocrine, paracrine, or statin transcription was observed when both FoxO and Smad2
endocrine manner. The mature myostatin peptide has been transcription factors were transfected into myoblasts resulting
shown to interact with the Activin Type IIB receptor (ActRIIB) in inhibition of myoblast differentiation (36). The authors fur-
and the TGF-b type I receptor, ALK5, and signal through ther proposed that during atrophy when FoxO activity is
canonical smad2/smad3 proteins (22,26,27). enhanced these factors could in turn increase the expression
The importance of myostatin proteolytic processing is quite of myostatin (36).
clear, as mutation of the myostatin RSRR processing site to the Considering the potent negative function of myostatin dur-
amino acids GLDG leads to prominent skeletal muscle hyper- ing myogenesis, it is not surprising that myostatin is able to
trophy in mice (28). Members of the bone morphogenetic pro- negatively auto regulate its own transcription. In fact, previous
tein-1/tolloid (BMP-1/TLD) family of metalloproteinases have a studies have revealed that myostatin expression at the mRNA
critical function in removal of the myostatin LAP region from level and myostatin promoter activity was reduced in muscle
the latent myostatin complex in the circulation, leading to acti- cells upon exposure to exogenous myostatin (37). This inhibi-
vation of mature myostatin (29). In support, generation of a tion of myostatin transcription was mediated through smad7
mutated LAP peptide, which cannot be cleaved by BMP-1/TLD, (27,37). Consistent with this, an increased expression of myo-
resulted in increased muscle mass in vivo. During myoblast statin mRNA and reduced smad7 expression is observed in
differentiation, the extent of proteolytic processing is downre- Belgian Blue cattle, in which functional myostatin protein is
gulated via a feedback loop wherein the expression of the ser- absent (37).
ine protease furin is negatively regulated by myostatin. This The contextual control of myostatin transcription has been
mechanism has been proposed to regulate myostatin levels highlighted in some contemporary studies. Recent findings
during fetal muscle development to allow for timely myogenic show that inflammatory factors, such as TNF-a, and increased
differentiation (24). Thus, proteolytic processing of myostatin oxidative stress can stimulate myostatin production. The
plays an important role in regulation of myostatin function. increased myostatin transcription due to TNF-a and hydrogen
peroxide treatment was attributed to activation of NF-jB and
its subsequent binding to myostatin promoter sequences (38).
Regulation of Myostatin NF-jB directed regulation of myostatin transcription in myo-
Transcriptional Regulation of Myostatin blasts was also reported under conditions of hyperammonemia
As is typical of many muscle-specific genes, the 50 promoter/ (39). In addition, high glucose loading has been shown to
enhancer region of the myostatin gene contains several evolu- increase myostatin expression in muscle cells and HepG2 liver
tionarily conserved E-Box sequence motifs. (30,31). The E-Box cells, which was shown to be due to carbohydrate-responsive
motif, CANNTG, is the canonical binding site for basic Helix- element-binding protein (ChREBP) (Fig. 2a) binding to

590 Myostatin
through binding to a sterol regulatory element present in the
myostatin promoter to promote insulin resistance (17). In
addition, myostatin transcription is upregulated in skeletal
muscle of streptozotocin-induced type 1 diabetic mice through
the action of the Foxa2 transcription factor, which is activated
due to reduced insulin signaling in type 1 diabetic muscle (40).
Post-Transcriptional Regulation of Myostatin
One of the modes of post-transcriptional regulation of genes is
through the action of microRNAs. Clop et al., (2006) first
reported the involvement of microRNAs in regulation of myo-
statin in Texel sheep, which express low levels of myostatin
concomitant with a muscle hypertrophy phenotype (41). The
authors reported a G to C single nucleotide polymorphism in
the 3’UTR of the myostatin gene in Texel sheep making it a
target of the muscle-specific microRNA miR206, which
resulted in inhibition of myostatin and increased musculature
(41). After this initial finding there have been additional
reports highlighting myostatin regulation by microRNAs,
including miR-27a/b, miR-208a/b and miR-499. Several inde-
pendent groups have revealed myostatin as a target of miR-
Summary of myostatin function during muscle wast- 27a/b (42–45) using both in vitro and in vivo models. Allen
FIG 2 ing and insulin resistance. (a) Increased myostatin et al., (2007) observed higher expression of mature myostatin
levels can promote the development of insulin resist- mRNA in fast twitch muscles when compared with slow-twitch
ance. High glucose and palmitate result in transcrip-
tional activation of myostatin through the action of
muscles, although it was noted that myostatin pre-mRNA
carbohydrate-responsive element-binding protein expression levels were similar between fast and slow muscle
(ChREBP) and sterol regulatory element binding types (42). This observation led them to investigate post-
protein-1c (SREBP1c), respectively. Elevated myosta- transcriptional control of myostatin, and results revealed that
tin levels lead to increased expression of Casitas B- miR-27a/b repressed myostatin expression and was highly
lineage lymphoma b (Cblb) and phosphotyrosine
interaction domain containing 1 (PID1) through expressed in slow-twitch muscles, where reduced myostatin
Smad3- and NFjB-dependent signaling mechanisms expression was noted (42). Taken together these data reveal
respectively, which in turn target and repress insulin that miR-27a/b may play an important role in regulating mus-
signaling. Impaired insulin signaling results in cle fibre type-specific expression of myostatin. Huang et al.,
reduced glucose uptake and the eventual develop-
(2012) further demonstrated an inverse relationship between
ment of insulin resistance. (b) Myostatin promotes
the development of skeletal muscle wasting. Epige- miR-27a and myostatin expression in proliferating C2C12
netic and genetic factors lead to increased levels of myoblasts and postulated that miR-27a promotes myoblast
myostatin during skeletal muscle wasting. Increased proliferation by targeting myostatin (46). In addition, further
levels of myostatin result in activation of canonical work has shown that miR-27a is responsible for decreased
Smad3 signaling and increased expression of Fork-
myostatin expression in C2C12 myoblasts treated with leucine,
head Box O (FoxO) transcription factors. Inhibition of
pAkt by myostatin promotes the activation and sub- thereby promoting leucine-induced proliferation of myoblasts
sequent nuclear translocation of FoxO transcription (43). MiR-27a has also been shown to target bovine myostatin
factors to increase the expression of downstream tar- 30 UTR in the Piedmontese cattle breed (45). Piedmontese cattle
get genes. Increased FoxO activation results in have low levels of mature myostatin because of C313Y mis-
increased expression of the ubiquitin E3 ligases
Atrogin-1 and MuRF1, enhanced ubiquitin-
sense mutation in the myostatin gene and hence exhibit skele-
proteasome pathway (UPP)-mediated protein degra- tal muscle hypertrophy and hyperplasia (2,47,48). Although
dation and muscle wasting. Myostatin has further the majority of Piedmontese cattle have this mutation, the
been shown to increase the expression of miR-1, degree of resulting skeletal muscle hypertrophy is variable.
which inhibits HSP70 and subsequently reduces pAkt
Miretti et al., (2013) have suggested that this conformational
levels leading to increased FoxO, Atrogin-1 and
MuRF1 expression and the development of skeletal difference could be due to the differential expression of miR-
muscle wasting. 27a (45).
Recent work from our laboratory has revealed that miR-
carbohydrate response element (ChoRE) present in the myo- 27a/b plays a role in feedback negative autoregulation of myo-
statin promoter region (17). Similarly, palmitate-mediated statin (44). Our results show that myostatin up-regulates the
activation of sterol regulatory element binding protein-1c expression of miR-27a/b, via a smad3-dependent mechanism,
(SREBP1c) can also activate myostatin transcription (Fig. 2a) to inhibit its own expression. Further analysis revealed that

SHARMA ET AL. 591


IUBMB LIFE

miR-27a/b-mediated inhibition of myostatin has a role in pro- Post-Translational Regulation of Myostatin


moting satellite cell (SC) activation, increasing myoblast prolif- Several reports have revealed that myostatin function is regu-
eration, and preventing muscle wasting (44). Similarly, Yang lated via protein-protein interactions. The sarcomeric protein
et al., (2014) have also demonstrated that miR-27a positively titin-cap has been shown to bind to mature myostatin protein
regulates porcine myoblast proliferation through inhibiting (60). Overexpression of titin-cap in myoblasts did not alter
myostatin (49). myostatin synthesis and processing, however myoblast prolif-
In an earlier study in which the effect of essential amino eration increased. Based on these results it was concluded
acids (EAA) on muscle protein synthesis and muscular hyper- that titin-cap modulates the secretion of myostatin (60).
trophy in humans was analyzed, expression analysis for miR- Another protein that regulates myostatin secretion is human
NAs revealed that miR-499 and miR-208b were upregulated small glutamine-rich tetratricopeptide repeat-containing pro-
within 3 h of ingestion of 10g of EAA in vastus lateralis human tein (hSGT), which was found to interact with myostatin within
skeletal muscle (50). Both miR-499 and miR-208b are encoded myoblasts (61). The C-terminal region of hSGT and the N-
by the introns of myosin genes, miR-208b from the Myhc7 terminal signal peptide region of myostatin were essential for
gene and miR-499 from the Myhc7b gene, respectively (51). this association (61). Previously, follistatin had been shown to
While Drummond et al., (2009) reported that reduced expres- interact with mature myostatin thereby inhibiting its binding
sion of myostatin correlated with increase expression of these to ActRIIB (22). Consistent with this, follistatin inhibits myosta-
microRNAs, myostatin was not validated as a direct target of tin activity and rescues the inhibition of muscle differentiation
miR-499 and miR-208b in the study. However, a later study imposed by myostatin (62). Conversely, follistatin-null mice
established myostatin to be a valid target of miR-499 (52). have diminished muscle mass at birth, concomitant with
Another related microRNA, miR-208a that is heart specific and increased myostatin activity (63). Interestingly, transgenic
encoded by an intron of the Myhc6 gene (51) was also shown mice overexpressing follistatin show a hypermuscular pheno-
to target myostatin (53). Callis et al., 2009 revealed that over- type that is greater than the observed myostatin-null pheno-
expression of miR-208a in mice heart induced hypertrophic type (22), indicating that follistatin may in fact control the
growth and led to repression of myostatin, suggesting myosta- activity of additional negative regulators of muscle growth.
tin to be a potential target of miR-208a (53). Similar to follistatin, follistatin-related gene also interacts with
mature myostatin peptide indicated by a dose-dependent
decrease in myostatin activity in reporter gene analysis experi-
Epigenetic Regulation of Myostatin ments (25). In the circulation, Growth and differentiation
Apart from the regulation of myostatin by microRNAs, epige- factor-associated serum protein-1 (GASP-1) was found to bind
netic mechanisms also regulate myostatin. Fan et al., (2012) to mature and LAP portions of myostatin. GASP-1 also inhibits
identified that sulphoraphane (SFN), a bioactive compound myostatin signaling as indicated by reporter gene analysis
that is abundant in cruciferous vegetables, like broccoli and (64). In agreement with these results, overexpression of GASP-
cauliflower, is a negative regulator of myostatin in porcine SCs 1 leads to muscle hypertrophy in mice (65). Furthermore, our
(54). The authors found that in addition to being a histone group has reconciled the role of GASP-1 as a positive regulator
deacetylase inhibitor, SFN is also a DNA methyltransferase of myogenesis by establishing it as a transcriptional target of
inhibitor, and when compared with trichostatin A and 5-aza- peroxisome proliferator-activated receptorb/d (PPARb/d). Spe-
2’-deoxycytidine, SFN significantly repressed myostatin cifically, our data revealed that PPARb/d positively regulated
expression. This epigenetic repression of myostatin was attrib- myogenesis by enhancing the transcription of GASP-1, which
uted in part to hypoacetylation of the MyoD binding sites in the in turn abrogates myostatin activity during myogenesis (66).
myostatin promoter (54). Additionally, decorin, a leucine-rich repeat extracellular
Histone methyl transferases belonging to the SMYD family proteoglycan, has been shown to interact with mature myosta-
are highly expressed in myogenic tissue (55,56), thus showing tin, in a Zn21-dependent manner (67). This interaction was
the potential to be important regulators of skeletal muscle demonstrated to relieve the inhibitory effect of myostatin on
mass. Recently one of the SMYD family members, SMYD3, was myoblast proliferation in vitro. Recently, myostatin was found
demonstrated to be involved in epigenetic regulation of myo- to interact with another extracellular matrix protein, laminin,
statin. Proserpio et al., (2013) have shown that SMYD3 helps and this interaction was able to inhibit myostatin signaling in
chromatin recruitment of BRD4 (Bromodomain protein) and p- myoblasts (68). Laminins are large proteins present in basal
TEFb (positive transcription elongation factor complex) to the lamina and mutations in laminin are known to cause myopa-
myostatin promoter. In doing so, these proteins facilitate early thy. In a previous report, whereby laminin alpha 2-null mice
transcriptional elongation of myostatin mRNA thus positively (dy) were crossed with mstn-null mice, an improvement in
regulating myostatin expression. Moreover, SMYD3 expression muscle regeneration was observed; however, the pathology
was increased during Dex-induced atrophy in C2C12 myotubes due to loss of laminin was exacerbated (69). These results sug-
(57), a condition known to increase myostatin expression gest that while laminin can regulate myostatin activity, myo-
(58,59). statin might not regulate or improve laminin function.

592 Myostatin
Altogether, it is clear that myostatin levels are tightly regu- myostatin-Cblb signaling resulted in hypophosphorylation of
lated through a combination of transcriptional, posttranscrip- Akt, and subsequent development of insulin resistance (17).
tional, epigenetic, and post-translational mechanisms. Recent work from our lab has established myostatin as an
inducer of Phosphotyrosine Interaction Domain containing 1
(PID1) protein in human muscle cells (81). PID1 is well known
Myostatin in Obesity and T2D for its role in the development of insulin resistance in both
Recently, our understanding of function of myostatin outside white adipocytes and murine C2C12 muscle cells (82,83).
its conventional role in muscle growth is gaining momentum. Bonala et al. identified that myostatin signals via NF-jB to
Early reports have shown that absence of myostatin affects activate PID1 gene transcription (Fig. 2a) and that myostatin
body metabolism leading to enhanced resistance to obesity activation of PID1 affected the phosphorylation status of IRS1,
and improved sensitivity to insulin in both high fat fed (HFD) but not total IRS1 protein levels in human muscle cells (81).
mouse models and in genetically obese mice (18,70–72). More The subsequent hypophosphorylation of IRS1 prevented fur-
recently, work from our laboratory has revealed that elevated ther PI3K/Akt/Glut-4 signaling resulting in reduced glucose
AMPK and PPAR signaling in myostatin-null muscle, as well as uptake by muscle cells (81). These results suggest that myosta-
enhanced COX-2-mediated prostaglandin synthesis, lead to the tin can act as potent inducer of insulin resistance through both
improved insulin sensitivity, increased peripheral tissue fatty Cblb- and PID1-mediated inhibition of insulin signaling.
acid oxidation and enhanced beige phenotype of white adipo- Elevated serum levels and expression of myostatin have
cytes observed in myostatin-null mice upon HFD feeding also been observed in muscle from a streptozotocin-induced
(19,20). However, elevated levels of myostatin have been type 1 diabetic mouse model. Interestingly, the levels of myo-
reported in both mice and humans suffering from obesity statin could be abrogated by administering insulin to the type
(73,74). Allen et al., reported increased mRNA and protein lev- 1 diabetic mice (84). However, results from this study indicate
els of myostatin in muscle and adipose tissues of leptin defi- a primary role for myostatin in promoting muscle atrophy in
type 1 diabetic mice, as opposed to regulating insulin
cient obese mice as well as in HFD fed wild type mice (73).
sensitivity.
Furthermore, Hittel et al., reported higher levels of serum
myostatin and increased secretion of myostatin by myotubes
derived from muscle biopsies of obese women (74). In addition, Myostatin and Skeletal Muscle Wasting
activation of myostatin signaling resulted in increased fat
Myostatin has been shown to play a causative role during skel-
accumulation in mice with subsequent muscle loss (75). Collec-
etal muscle wasting. In fact, intramuscular injection of myosta-
tively, these studies suggest that myostatin behaves as a cru-
tin protein producing and secreting CHO cells leads to the
cial factor in regulating whole body metabolism.
induction of cachectic-like skeletal muscle wasting in mice
Palsgaard et al. for the first time reported higher mRNA
(16). Mechanistically myostatin-induced muscle wasting results
levels of myostatin in type 2 diabetic muscles as well as in
from both impaired protein synthesis and enhanced ubiquitin-
non-obese insulin resistant subjects (76). Later, Hittel et al.
proteasome pathway-mediated protein degradation of critical
reported that exogenous injection of myostatin elevates insulin
muscle-specific proteins (85–87). Specifically, work from our
resistance in both muscle and liver of mice and that the levels
lab has revealed that excess myostatin results in impaired
of myostatin decreased with aerobic exercise, thus improving IGF-1/Akt/PI3K signaling, which resulted in enhanced activa-
insulin sensitivity in humans (77). Moreover, inhibition of myo- tion of the transcription factor Forkhead box O1 (FoxO1; Fig.
statin activity in liver protects from developing insulin resist- 2b). Myostatin-mediated activation of FoxO1 led to increased
ance upon HFD feeding in mice (70). Taken together, these expression of atrophy-related genes, including the Ubiquitin
conclusions suggest that myostatin levels correlate with the E3 ligases Atrogin-1 and MuRF1 and enhanced protein ubiqui-
extent of insulin resistance with or without associated obesity. tination (Fig. 2(86)). A further study by Durieux et al., (2007)
However, these studies do not delineate the major signaling revealed that over expression of myostatin in muscle resulted
mechanism behind myostatin-mediated insulin resistance. in significant loss of skeletal muscle mass, which was associ-
Subsequent studies from our group have identified that ated with reduced expression of muscle structural genes,
myostatin signals via smad3 to increase the expression of Casi- including myosin heavy chain and desmin (88). More recently,
tas B-lineage lymphoma b (Cblb) (Fig. 2a) in both muscle and Lokireddy et al., (2011) have established that myostatin signals
liver tissues (17). Cblb is an ubiquitin E3 ligase that has been in a smad3-dependent manner to activate FoxO1 and Atrogin-
shown to specifically degrade insulin receptor substrate 1 1, to promote the ubiquitination and subsequent proteasome-
(IRS1) protein in muscle. The myostatin-Cblb-IRS1 signaling mediated degradation of sarcomeric proteins during muscle
mechanism was further validated in both in vitro and in vivo wasting (85).
insulin resistance models (17). IRS1 is a critical mediator in Results from our laboratory have also demonstrated that
the insulin-signaling pathway (78–80); as such degradation of myostatin facilitates translocation of the glucocorticoid recep-
IRS1 is associated with impairment in further activation of tor to the nucleus during dexamethasone-induced muscle
PI3K/Akt/Glut-4 signaling. Consistent with this, activation of wasting to induce the expression of miR-1 (58). It was further

SHARMA ET AL. 593


IUBMB LIFE

shown that subsequently miR-1 targets and represses HSP70, (96). Although loss of muscle mass has a detrimental effect in
leading to reduced p-Akt levels, enhanced FoxO3 activity and liver cirrhosis patients, the mechanisms behind muscle loss
upregulation of the Ubiquitin E3 ligases MuRF1 and Atrogin-1 linked with liver diseases had until recently remained unex-
(Fig. 2(58)). These results reveal a novel mechanism for plored. Recent research indicates that myostatin is a key factor
dexamethasone-mediated muscle wasting as well as a poten- involved in muscle wasting seen in patients with liver diseases.
tial mechanism through which myostatin downregulates the Indeed, higher levels of circulatory myostatin were noted in
levels of p-Akt. patients with end-stage liver disease (12). Increased myostatin
expression was also observed in skeletal muscle of patients
with cirrhosis (39,97). In cirrhosis urea metabolism gets dis-
Myostatin and Chronic Obstructive turbed and as a result hyperammonemia is seen in muscle.
Pulmonary Disease (COPD) Qiu et al., (2013) recently demonstrated that hyperammonemia
COPD is characterized by obstruction of airflow, which not induces NF-jB signaling leading to increased myostatin tran-
only affects lung function but also has significant systemic con- scription (39). Myostatin has also been implicated in muscle
sequences. One of the secondary complications of COPD is pul- wasting associated with post-transplantation of liver during
monary cachexia, manifested as peripheral muscle dysfunc- which, steroid therapy, mTOR inhibitors or calcineurin inhibi-
tion, respiratory muscle dysfunction and a loss of total body tors are used. Specifically, glucocorticoids and calcineurin
mass. Approximately 30–40% of people with COPD undergo inhibitors are known to induce myostatin and hence are likely
muscle wasting. Consistent with this, Doucet et al., (2010) to exacerbate muscle wasting in liver transplant patients
noted increased expression of the ubiquitin proteasome E3 (98–100).
ligases Atrogin-1 and MuRF-1 in muscle of COPD patients
together with increased FoxO1 protein levels, when compared
with healthy controls (89). Another phenomenon that occurs in
Myostatin and Chronic Kidney Disease
patients suffering from COPD is the decline in oxidative muscle (CKD)
phenotype, resulting in reduced energy output and increased One of the comorbidities frequently noted in chronic kidney
oxidative stress (90). In fact, cachectic COPD patients display disease is a rapid onset of muscle wasting. The loss of muscle
higher levels of oxidative stress as compared with noncachec- mass in these patients is marked by increased protein break-
tic COPD patients and healthy controls, at rest and following down, insulin resistance, excessive glucocorticoid production,
physical exercise (91,92), as well as increased mitochondrial and impaired p-Akt signaling. Work by Zhang et al., 2011 has
reactive oxygen species production (93). The manifestations revealed a 2- to 3-fold increase in the expression of myostatin
associated with COPD progression, including increased protea- in the muscles of mice with CKD (101). Subsequent pharmaco-
somal activity and elevated oxidative stress, point to a possible logical inhibition of myostatin, by subcutaneous injections of
role for myostatin in COPD. Most certainly, myostatin has been an anti-myostatin peptibody, reversed the loss of body weight
implicated in development of oxidative stress in skeletal mus- and muscle mass and reduced the levels of circulating inflam-
cle through increasing TNF-a signaling, via a mechanism matory cytokines in mice with CKD. Furthermore, antagonism
involving NF-jB and NADPH oxidase (38). Interestingly, myo- of myostatin led to reduced muscle protein degradation, con-
statin activation of TNF-a acts as a novel feed forward mecha- comitant with increased muscle protein synthesis (101). More
nism to promote further production of myostatin (38). Several recently, Stat3-mediated induction of myostatin has been
additional studies support a role for myostatin during COPD- implicated in promoting the muscle loss associated with CKD
induced muscle wasting. Myostatin expression has been shown (102). Specifically, Zhang et al., 2013 demonstrated that acti-
to negatively correlate with muscle strength during COPD (94). vation of Stat3 resulted in increased levels of myostatin, via a
Plant et al., (2010) have further shown increased expression of mechanism involving the transcription factor CCAAT/
myostatin as well as the Ubiquitin E3 ligases Atrogin-1 and enhancer-binding protein d (C/EBPd) to stimulate skeletal mus-
Nedd4 in the vastus lateralis muscle of COPD patients (95). cle loss during CKD (102). In another study, TNF-a induced
Furthermore, circulating myostatin levels are increased in the myostatin was alleged to activate autophagy related genes and
serum of patients with COPD and correlate with decreased the ubiquitin proteosomal system in muscle of rats with CKD,
skeletal muscle mass (13). resulting in muscle wasting (103).

Myostatin and Liver Diseases Myostatin and Cancer Cachexia


Chronic necrosis and inflammation of liver eventually results Cachexia is a multifactorial syndrome associated with chronic
in cirrhosis of the liver that in turn is compounded by loss of disease that is accompanied by several symptoms including
skeletal muscle mass (96). Loss of muscle mass contributes to loss of body weight, skeletal muscle loss, and wasting with
weakness and frailty in cirrhotic patients, furthermore skeletal associated asthenia (loss of muscle strength), loss of body fat
muscle mass is not restored even after liver transplantation and changes in lipid, carbohydrate and protein metabolism

594 Myostatin
(104,105). Cachexia is associated with many chronic diseases recent study by Aversa et al. has revealed that myostatin pro-
including cancer and importantly cancer cachexia is exhibited tein levels were increased in skeletal muscle from patients
by 80% of all patients who possess advanced tumors of pan- with gastric cancer; although in the same study no change in
creatic, lung and gastric origin, and it is responsible for over skeletal muscle myostatin protein levels were noted in patients
30% of cancer-related fatalities (106,107). with lung cancer (10). A study by Sim et al. detected a signifi-
Myostatin is a secreted pro-cachectic growth factor that cant increase in myostatin expression in Central neurocytoma
promotes the development of cachectic-like skeletal muscle tumor tissue when compared with healthy normal tissue (119).
wasting (16). Elevated levels of myostatin expression as well Several subsequent articles have failed to show increased myo-
as enhanced myostatin signaling have been shown to correlate statin during cancer cachexia and in fact a study by D’orlando
with the progression of cancer and cancer-associated cachexia et al. reports no change in myostatin mRNA expression in gas-
in several experimental models of cancer cachexia. Myostatin tric cancer patients, when compared with healthy individuals
mRNA and protein levels were increased in rats 7 days follow- (120), although no assessment of myostatin muscle protein lev-
ing Yoshida AH-130 hepatoma tumor transplantation (108) els or serum levels were made. In a separate study, myostatin
and in mice following inoculation with the S-180 mouse ascitic mRNA was further assessed in gastric cancer patients, who
tumor (109). In addition, transplantation of C26 adenocarci- have minimal or no loss of muscle mass, and results revealed
noma cells and LP07 lung tumor cells into mice also correlated a significant decrease in myostatin mRNA in muscle tissue
with elevated myostatin protein levels in skeletal muscle from gastric cancer patients (121). However, in this study, the
(110–112). Increased serum and muscle-specific expression of patients were at an early stage of cancer progression and it
myostatin was further noted in a chemically induced model of was suggested that changes in myostatin may become appa-
urothelial carcinogenesis (113). Further evidence for the role rent as the cancer progresses. In a further study, Breitbart
of myostatin during cancer cachexia is provided through inhib- et al. have described the use of an immunoradiometric sand-
itor studies. Liu et al. demonstrated that injection of antisense wich assay to detect myostatin prodomain in patients with gas-
RNA oligonucleotides targeted to myostatin into S-180 mouse trointestinal or hepatic cancer undergoing acute weight loss.
ascitic tumor inoculated mice resulted in decreased myostatin Results revealed no increase, but a decrease, in myostatin pro-
expression and an associated increase in skeletal muscle mass domain levels in serum samples from cancer patients, when
(109). Furthermore, antibody-mediated myostatin inhibition compared with healthy controls (122). It is important to high-
prevented loss of muscle mass and muscle weakness associ- light that this assay only detects the N-terminal myostatin pro-
ated with injection of Lewis lung carcinoma cells into mice domain, also referred to as the latency associated peptide
(114). Antagonism of myostatin signaling through addition of a (LAP) region, and not the biologically active C-terminal mature
soluble receptor antagonist (sActRIIB) has also shown utility in ligand of myostatin. As the prodomain acts as a potent antago-
overcoming cancer associated cachexia. Addition of sActRIIB nist of myostatin function (123) it remains to be determined
in both mice injected with Lewis lung carcinoma and C26 cells how lowered myostatin prodomain serum levels, as observed
resulted in reversal of muscle wasting (112,115,116) and in this cancer cachexia model, would in turn affect myostatin
improved muscle strength (112,116). In addition, prolonged biological activity.
survival was noted in C26 tumor bearing mice administered Although evidence to date has clearly linked myostatin
sActRIIB (112). In a recent report, genetic inactivation of myo- with the progression of cancer-associated cachexia in rodent
statin in Apcmin/1 mice (a model of colorectal cancer) resulted models, additional studies should be undertaken using larger
in a significant reduction in the number and size of intestinal samples sizes and well-stratified patients to fully characterize
polyps and prevented loss of muscle mass (117). In the same myostatin function during cancer-cachexia in humans. Fur-
report, wildtype and myostatin-null (Mstn2/2) mice were thermore, several molecular and biochemical techniques need
injected with Lewis lung carcinoma tumor cells (LLC); LLC to be employed in these studies to carefully assess not only
tumor growth was reduced and the expression of both MuRF1 myostatin gene expression changes but also myostatin protein
and Atrogin-1 was inhibited in Mstn2/2 muscle, when com- levels in tumor tissue, skeletal muscle and in circulation.
pared with wildtype controls. Another noteworthy observation Nevertheless, recent clinical trials with myostatin inhibitors
was the reduced protein levels of lipidated LC3b-II (an autoph- (including LY2495655 and BYM-338) appear to be promising
agy related marker) in the muscle of LLC tumor bearing in preserving muscle mass and thus offer hope of treatment
Mstn2/2 mice when compared with LLC tumor bearing wild- for cachetic patients (124).
type mice, suggesting that loss of myostatin also prevents
induction of autophagy-related genes. With regards to autoph-
agy, the in vivo results seen here concur with the report by Myostatin and Sarcopenia
Lee et al., (2011), wherein myostatin was shown to induce Sarcopenia is the decline in skeletal muscle mass and function
autophagy in C2C12 myoblasts (118). associated with the normal aging process (125,126). Differen-
While several lines of evidence support a role for myosta- ces in the expression of myostatin have been described during
tin in cancer-associated cachexia in rodents, the role of myo- aging-related skeletal muscle wasting. myostatin mRNA has
statin in cancer cachexia in humans remains less clear. A been shown to decrease in aged rats (127,128), although

SHARMA ET AL. 595


IUBMB LIFE

Baumann et al. noted increased myostatin protein levels dur- Work from our laboratory has shown that antagonism of myo-
ing aging (127). In a separate study, age-induced reduction of statin through addition of a dominant negative mimetic protein
skeletal muscle mass in rats was associated with increased (Mstn-ant1) leads to enhanced muscle regeneration following
muscle-specific myostatin protein and mRNA expression (129). notexin injection, with accelerated nascent fiber formation,
However, in contrast, a study by Kawada et al. failed to show reduced fibrosis, and increased myofibre size, when compared
age-related differences in skeletal muscle myostatin protein with saline injected controls (142). Furthermore, treatment
levels in mice (130). Similar to the variations in myostatin with Mstn-ant1 resulted in increased SC activation and
expression observed in rodent studies, decreased myostatin enhanced grip strength in aged mice (142). Studies by LeBras-
mRNA expression (131), unaltered myostatin mRNA (132,133) seur et al. and Murphy et al. have assessed the utility of
and serum levels (134) as well as increased myostatin mRNA, antibody-mediated blockade of myostatin during aging in mice
protein and serum levels (14,15,135–137), have all been previ- (143,144). Myostatin inhibition resulted in increased muscle
ously reported in humans during aging. More recently, work mass and function in aged mice (143,144), with a noticeable
by Patel et al. has revealed that myostatin mRNA expression is increase in muscle performance in response to exercise (144).
reduced in aged individuals that have a higher grip strength, In addition, treatment of aged mice with a soluble form of the
suggesting that myostatin may play a role in regulating muscle myostatin receptor (ActRIIB-Fc) and recombinant myostatin
strength during aging (138). propeptide (Propeptide-Fc), both inhibitors of myostatin,
The disparity between the findings from these different
resulted in increased muscle mass (145,146) and function
rodent and human studies has complicated our understanding
(146) when compared with vehicle treated controls. Taken
of the role of myostatin during sarcopenia. The differences
together these data suggest that postnatal inactivation of myo-
observed in myostatin expression suggest that the effect of
statin may be an effective approach to overcome the loss of
myostatin during aging may be limited to specific stages of
muscle mass and function observed during sarcopenia.
sarcopenia, or more probably, that changes in myostatin activ-
ity cannot be directly implied from expression analysis alone.
Most certainly previous studies, as reviewed above, have Conclusions
revealed that myostatin expression and activity is tightly regu-
Since the discovery of myostatin in 1997, considerable pro-
lated at several levels, including transcriptionally, post-
transcriptionally and in circulation through the action of sev- gress has been made in understanding myostatin function not
eral interacting proteins. Furthermore, it is established that only in skeletal muscle but also in heart and adipose tissue.
myostatin can in fact inhibit its own expression and activity Myostatin appears to be a versatile protein with a wide range
through negative feedback mechanisms involving smad7 (37) of functions (Figs. 2a and 2b); hence, exciting new develop-
and more recently microRNA-27a/b (44). Taken together these ments are expected in myostatin research in the near future.
data suggest that changes in myostatin expression observed
during aging may not be reflective of myostatin activity, or for Acknowledgements
that matter myostatin function, during sarcopenia. This sug- The authors thank all the researchers who have contributed to
gests that a more robust approach should be undertaken,
myostatin research. They acknowledge the support of National
involving a more complete assessment of both inactive and
Research Foundation (NRF), and NMRC (BnB), Singapore.
active forms of myostatin, which can then be correlated to the
progression of sarcopenia.
That being said, further evidence underscoring the role of References
myostatin during sarcopenia can be seen in studies on myosta- [1] Elliott, B., Renshaw, D., Getting, S., and Mackenzie, R. (2012) The central role
tin-null (mstn-null) mice. Aged mstn-null mice maintain of myostatin in skeletal muscle and whole body homeostasis. Acta Physiol.
(Oxf) 205, 324–340.
increased muscle mass and myofibre area when compared
[2] Kambadur, R., Sharma, M., Smith, T. P., and Bass, J. J. (1997) Mutations in
with aged wild type mice (139–141). Further work from our
myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle.
lab revealed that the pool of muscle stem cells (SCs) was con- Genome Res. 7, 910–916.
sistently higher in mstn-null mice and although SC activation [3] McPherron, A. C., Lawler, A. M., and Lee, S. J. (1997) Regulation of skeletal
declined during aging in both wildtype and mstn-null mice, a muscle mass in mice by a new TGF-beta superfamily member. Nature 387,
higher proportion of active SCs was maintained in mstn-null 83–90.
mice irrespective of age (140). Muscle from aged mstn-null [4] Schuelke, M., Wagner, K. R., Stolz, L. E., Hubner, C., Riebel, T., et al. (2004)
mice also regenerated more robustly when compared with Myostatin mutation associated with gross muscle hypertrophy in a child. N.
Engl. J. Med. 350, 2682–2688.
wildtype counterparts following both cardiotoxin and notexin-
[5] Shelton, G. D., and Engvall, E. (2007) Gross muscle hypertrophy in whippet
induced injury (140,141). These data suggest that prolonged
dogs is caused by a mutation in the myostatin gene. Neuromuscul. Disord.
absence of myostatin has beneficial effects during aging and 17, 721–722.
reduces the extent of sarcopenia. [6] Langley, B., Thomas, M., Bishop, A., Sharma, M., Gilmour, S., et al. (2002)
Several additional studies have also assessed the effect of Myostatin inhibits myoblast differentiation by down-regulating MyoD expres-
postnatal myostatin inhibition on aging-associated muscle loss. sion. J. Biol. Chem. 277, 49831–49840.

596 Myostatin
[7] McCroskery, S., Thomas, M., Maxwell, L., Sharma, M., and Kambadur, R. [27] Zhu, X., Topouzis, S., Liang, L. F., and Stotish, R. L. (2004) Myostatin
(2003) Myostatin negatively regulates satellite cell activation and self- signaling through Smad2, Smad3 and Smad4 is regulated by the inhibitory
renewal. J. Cell. Biol. 162, 1135–1147. Smad7 by a negative feedback mechanism. Cytokine 26, 262–272.
[8] McCroskery, S., Thomas, M., Platt, L., Hennebry, A., Nishimura, T., et al. [28] Zhu, X., Hadhazy, M., Wehling, M., Tidball, J. G., and McNally, E. M. (2000)
(2005) Improved muscle healing through enhanced regeneration and Dominant negative myostatin produces hypertrophy without hyperplasia in
reduced fibrosis in myostatin-null mice. J. Cell. Sci. 118, 3531–3541. muscle. FEBS Lett. 474, 71–75.
[9] Thomas, M., Langley, B., Berry, C., Sharma, M., Kirk, S., et al. (2000) [29] Wolfman, N. M., McPherron, A. C., Pappano, W. N., Davies, M. V., Song, K., et al.
Myostatin, a negative regulator of muscle growth, functions by inhibiting (2003) Activation of latent myostatin by the BMP-1/tolloid family of
myoblast proliferation. J. Biol. Chem. 275, 40235–40243. metalloproteinases. Proc. Natl. Acad. Sci. USA 100, 15842–15846.
[10] Aversa, Z., Bonetto, A., Penna, F., Costelli, P., Di Rienzo, G. et al. (2012) [30] Salerno, M. S., Thomas, M., Forbes, D., Watson, T., Kambadur, R., et al.
Changes in myostatin signaling in non-weight-losing cancer patients. Ann. (2004) Molecular analysis of fiber type-specific expression of murine myo-
Surg. Oncol. 19, 1350–1356. statin promoter. Am J. Physiol. Cell Physiol. 287, C1031–C1040.
[11] Gonzalez-Cadavid, N. F., Taylor, W. E., Yarasheski, K., Sinha-Hikim, I., Ma, [31] Spiller, M. P., Kambadur, R., Jeanplong, F., Thomas, M., Martyn, J. K., et al.
K., et al. (1998) Organization of the human myostatin gene and expression (2002) The myostatin gene is a downstream target gene of basic helix-loop-
in healthy men and HIV-infected men with muscle wasting. Proc. Natl. Acad helix transcription factor MyoD. Mol. Cell. Biol. 22, 7066–7082.
Sci. USA 95, 14938–14943. [32] Du, R., Chen, Y. F., An, X. R., Yang, X. Y., Ma, Y., et al. (2005) Cloning and
[12] Garcia, P. S., Cabbabe, A., Kambadur, R., Nicholas, G., and Csete, M. (2010) sequence analysis of myostatin promoter in sheep. DNA Seq. 16, 412–417.
Brief-reports: elevated myostatin levels in patients with liver disease: a [33] Li, J., Deng, J., Yu, S., Zhang, J., Cheng, D., et al. (2012) The virtual element
potential contributor to skeletal muscle wasting. Anesth. Analg. 111, 707– in proximal promoter of porcine myostatin is regulated by myocyte
709. enhancer factor 2C. Biochem. Biophys. Res. Commun. 419, 175–181.
[13] Ju, C. R., and Chen, R. C. Serum myostatin levels and skeletal muscle [34] Ma, K., Mallidis, C., Artaza, J., Taylor, W., Gonzalez-Cadavid, N., et al. (2001)
wasting in chronic obstructive pulmonary disease. Respir. Med. 106, 102– Characterization of 5’-regulatory region of human myostatin gene:
108. regulation by dexamethasone in vitro. Am. J. Physiol. Endocrinol. Metab.
[14] Leger, B., Derave, W., De Bock, K., Hespel, P., and Russell, A. P. (2008) 281, E1128–E1136.
Human sarcopenia reveals an increase in SOCS-3 and myostatin and a [35] Wang, B. W., Chang, H., Kuan, P., and Shyu, K. G. (2008) Angiotensin II
reduced efficiency of Akt phosphorylation. Rejuvenation Res. 11, 163–175B. activates myostatin expression in cultured rat neonatal cardiomyocytes via
[15] McKay, B. R., Ogborn, D. I., Bellamy, L. M., Tarnopolsky, M. A., and Parise, p38 MAP kinase and myocyte enhance factor 2 pathway. J. Endocrinol. 197,
G. (2012) Myostatin is associated with age-related human muscle stem cell 85–93.
dysfunction. FASEB J. 26, 2509–2521. [36] Allen, D. L., and Unterman, T. G. (2007) Regulation of myostatin expression
[16] Zimmers, T. A., Davies, M. V., Koniaris, L. G., Haynes, P., Esquela, A. F., and myoblast differentiation by FoxO and SMAD transcription factors. Am.
et al. (2002) Induction of cachexia in mice by systemically administered J. Physiol. Cell. Physiol. 292, C188–C199.
myostatin. Science 296, 1486–1488. [37] Forbes, D., Jackman, M., Bishop, A., Thomas, M., Kambadur, R., et al.
[17] Bonala, S., Lokireddy, S., McFarlane, C., Patnam, S., Sharma, M., et al. (2006) Myostatin auto-regulates its expression by feedback loop through
(2014) Myostatin induces insulin resistance via Casitas B-lineage lymphoma Smad7 dependent mechanism. J. Cell. Physiol. 206, 264–272.
b (Cblb)-mediated degradation of insulin receptor substrate 1 (IRS1) protein [38] Sriram, S., Subramanian, S., Sathiakumar, D., Venkatesh, R., Salerno, M. S.,
in response to high calorie diet intake. J. Biol. Chem. 289, 7654–7670. et al. (2011) Modulation of reactive oxygen species in skeletal muscle by
[18] McPherron, A. C., and Lee, S. J. (2002) Suppression of body fat myostatin is mediated through NF-kappaB. Aging Cell 10, 931–948.
accumulation in myostatin-deficient mice. J. Clin. Invest. 109, 595–601. [39] Qiu, J., Thapaliya, S., Runkana, A., Yang, Y., Tsien, C., et al. (2013)
[19] Zhang, C., McFarlane, C., Lokireddy, S., Bonala, S., Ge, X., et al. (2011) Hyperammonemia in cirrhosis induces transcriptional regulation of
Myostatin-deficient mice exhibit reduced insulin resistance through myostatin by an NF-kappaB-mediated mechanism. Proc. Natl. Acad. Sci.
activating the AMP-activated protein kinase signalling pathway. Diabetolo- USA 110, 18162–18167.
gia 54, 1491–501. [40] Sriram, S., Subramanian, S., Juvvuna, P. K., McFarlane, C., Salerno, M. S.,
[20] Zhang, C., McFarlane, C., Lokireddy, S., Masuda, S., Ge X., et al. (2012) et al. (2014) Myostatin induces DNA damage in skeletal muscle of
Inhibition of myostatin protects against diet-induced obesity by enhancing streptozotocin-induced type 1 diabetic mice. J. Biol. Chem. 289, 5784–5798.
fatty acid oxidation and promoting a brown adipose phenotype in mice. [41] Clop, A., Marcq, F., Takeda, H., Pirottin, D., Tordoir, X., et al. (2006) A
Diabetologia 55, 183–193. mutation creating a potential illegitimate microRNA target site in the
[21] McPherron, A. C., and Lee, S. L. (1996) The transforming growth factor-b myostatin gene affects muscularity in sheep. Nat. Genet. 38, 813–818.
superfamily. Growth Factors Cytokines Health Dis 1, 357–393. [42] Allen, D. L., and Loh, A. S. (2011) Posttranscriptional mechanisms involving
[22] Lee, S. J., and McPherron, A. C. (2001) Regulation of myostatin activity and microRNA-27a and b contribute to fast-specific and glucocorticoid-mediated
muscle growth. Proc. Natl. Acad. Sci. USA 98, 9306–9311. myostatin expression in skeletal muscle. Am. J. Physiol. Cell. Physiol. 300,
[23] Sharma, M., Kambadur, R., Matthews, K. G., Somers, W. G., Devlin, G. P., c124–c137.
et al. (1999) Myostatin, a transforming growth factor-beta superfamily mem- [43] Chen, X., Huang, Z., Chen, D., Yang, T., and Liu, G. (2013) MicroRNA-27a is
ber, is expressed in heart muscle and is upregulated in cardiomyocytes induced by leucine and contributes to leucine-induced proliferation promo-
after infarct. J. Cell. Physiol. 180, 1–9. tion in C2C12 cells. Int. J. Mol. Sci. 14, 14076–14084.
[24] McFarlane, C., Langley, B., Thomas, M., Hennebry, A., Plummer, E., et al. [44] McFarlane, C., Vajjala, A., Arigela, H., Lokireddy, S., Ge, X., et al. (2014)
(2005) Proteolytic processing of myostatin is auto-regulated during myogen- Negative auto-regulation of myostatin expression is mediated by Smad3
esis. Dev. Biol. 283, 58–69. and microRNA-27. PLoS One 9, e87687
[25] Hill, J. J., Davies, M. V., Pearson, A. A., Wang, J. H., Hewick, R. M., et al. [45] Miretti, S., Martignani, E., Accornero, P., and Baratta, M. (2013) Functional
(2002) The myostatin propeptide and the follistatin-related gene are inhibi- effect of mir-27b on myostatin expression: a relationship in Piedmontese
tory binding proteins of myostatin in normal serum. J. Biol. Chem. 277, cattle with double-muscled phenotype. BMC Genomics 14, 194
40735–40741. [46] Huang, Z., Chen, X., Yu, B., He, J., and Chen, D. (2012) MicroRNA-27a
[26] Rebbapragada, A., Benchabane, H., Wrana, J. L., Celeste, A. J., and promotes myoblast proliferation by targeting myostatin. Biochem. Biophys.
Attisano, L. (2003) Myostatin signals through a transforming growth factor Res. Commun. 423, 265–269.
beta-like signaling pathway to block adipogenesis. Mol. Cell. Biol. 23, 7230– [47] Berry, C., Thomas, M., Langley, B., Sharma, M., and Kambadur, R. (2002)
7242. Single cysteine to tyrosine transition inactivates the growth inhibitory

SHARMA ET AL. 597


IUBMB LIFE

function of Piedmontese myostatin. Am. J. Physiol. Cell. Physiol. 283, C135– [67] Miura, T., Kishioka, Y., Wakamatsu, J., Hattori, A., Hennebry, A., et al. (2006)
C141. Decorin binds myostatin and modulates its activity to muscle cells.
[48] McPherron, A. C., and Lee S. J. (1997) Double muscling in cattle due to Biochem. Biophys. Res. Commun. 340, 675–680.
mutations in the myostatin gene. Proc. Natl. Acad Sci. USA 94, 12457– [68] Yasaka, N., Suzuki, K., Kishioka, Y., Wakamatsu, J., and Nishimura, T. (2013)
12461. Laminin binds to myostatin and attenuates its signaling. Anim. Sci. J. 84,
[49] Yang, T., Chen, X. L., Huang, Z. Q., Wen, W. X., Xu, M., et al. (2014) 663–668.
MicroRNA-27a promotes porcine myoblast proliferation by downregulating [69] Li, Z. F., Shelton, G. D., and Engvall, E. (2005) Elimination of myostatin does
myostatin expression. Animal 8, 1867–1872. not combat muscular dystrophy in dy mice but increases postnatal lethality.
[50] Drummond, M. J., Glynn, E. L., Fry, C. S., Dhanani, S., Volpi, E., et al. (2009) Am. J. Pathol. 166, 491–497.
Essential amino acids increase microRNA-499, 2208b, and 223a and down- [70] Wilkes, J. J., Lloyd, D. J., and Gekakis, N. (2009) Loss-of-function mutation
regulate myostatin and myocyte enhancer factor 2C mRNA expression in in myostatin reduces tumor necrosis factor alpha production and protects
human skeletal muscle. J. Nutr. 139, 2279–2284. liver against obesity-induced insulin resistance. Diabetes 58, 1133–1143.
[71] Yang, J., and Zhao, B. (2006) Postnatal expression of myostatin propeptide
[51] van Rooij, E., Quiat, D., Johnson, B. A., Sutherland, L. B., Qi, X., et al. (2009)
cDNA maintained high muscle growth and normal adipose tissue mass in
A family of microRNAs encoded by myosin genes governs myosin
transgenic mice fed a high-fat diet. Mol. Reprod. Dev. 73, 462–469.
expression and muscle performance. Dev. Cell 17, 662–673.
[72] Zhao, B., Wall, R. J., and Yang, J. (2005) Transgenic expression of
[52] Bell, M. L., Buvoli, M., and Leinwand, L. A. (2010) Uncoupling of expression
myostatin propeptide prevents diet-induced obesity and insulin resistance.
of an intronic microRNA and its myosin host gene by exon skipping. Mol.
Biochem. Biophys. Res. Commun. 337, 248–255.
Cell. Biol. 30, 1937–1945.
[73] Allen, D. L., Cleary, A. S., Speaker, K. J., Lindsay, S. F., Uyenishi, J., et al.
[53] Callis, T. E., Pandya, K., Seok, H. Y., Tang, R. H., Tatsuguchi, M., et al. (2009)
(2008) Myostatin, activin receptor IIb, and follistatin-like-3 gene expression
MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in
are altered in adipose tissue and skeletal muscle of obese mice. Am. J.
mice. J. Clin. Invest. 119, 2772–2786.
Physiol. Endocrinol. Metab. 294, E918–E927.
[54] Fan, H., Zhang, R., Tesfaye, D., Tholen, E., Looft, C., et al. (2012) [74] Hittel, D. S., Berggren, J. R., Shearer, J., Boyle, K., and Houmard, J. A.
Sulforaphane causes a major epigenetic repression of myostatin in porcine (2009) Increased secretion and expression of myostatin in skeletal muscle
satellite cells. Epigenetics 7, 1379–1390. from extremely obese women. Diabetes 58, 30–38.
[55] Kawamura, S., Yoshigai, E., Kuhara, S., and Tashiro, K. (2008) smyd1 and [75] Reisz-Porszasz, S., Bhasin, S., Artaza, J. N., Shen, R., Sinha-Hikim, I., et al.
smyd2 are expressed in muscle tissue in Xenopus laevis. Cytotechnology (2003) Lower skeletal muscle mass in male transgenic mice with muscle-
57, 161–168. specific overexpression of myostatin. Am. J. Physiol. Endocrinol. Metab.
[56] Thompson, E. C., and Travers, A. A. (2008) A Drosophila Smyd4 homologue 285, E876–E888.
is a muscle-specific transcriptional modulator involved in development. [76] Palsgaard, J., Brons, C., Friedrichsen, M., Dominguez, H., Jensen, M., et al.
PLoS One 3, e3008 (2009) Gene expression in skeletal muscle biopsies from people with type 2
[57] Proserpio, V., Fittipaldi, R., Ryall, J. G., Sartorelli, V., and Caretti, G. (2013) diabetes and relatives: differential regulation of insulin signaling pathways.
The methyltransferase SMYD3 mediates the recruitment of transcriptional PLoS One, 4, e6575
cofactors at the myostatin and c-Met genes and regulates skeletal muscle [77] Hittel, D. S., Axelson, M., Sarna, N., Shearer, J., Huffman, K. M., et al.
atrophy. Genes Dev. 27, 1299–1312. (2010) Myostatin decreases with aerobic exercise and associates with
[58] Kukreti, H., Amuthavalli, K., Harikumar, A., Sathiyamoorthy, S., Feng, P. Z., insulin resistance. Med. Sci. Sports Exerc. 42, 2023–2029.
et al. (2013) Muscle-specific MicroRNA1 (miR1) Targets Heat Shock Protein [78] Saltiel, A. R., and Kahn, C. R. (2001) Insulin signalling and the regulation of
70 (HSP70) during Dexamethasone-mediated Atrophy. J. Biol. Chem. 288, glucose and lipid metabolism. Nature 414, 799–806.
6663–6678. [79] Shepherd, P. R., and Kahn, B. B. (1999) Glucose transporters and insulin
[59] Ma, K., Mallidis, C., Bhasin, S., Mahabadi, V., Artaza, J., et al. (2003) action–implications for insulin resistance and diabetes mellitus. N. Engl. J.
Glucocorticoid-induced skeletal muscle atrophy is associated with Med. 341, 248–257.
upregulation of myostatin gene expression. Am. J. Physiol. Endocrinol. [80] Thirone, A. C., Huang, C., and Klip, A. (2006) Tissue-specific roles of IRS
Metab. 285, E363–E371. proteins in insulin signaling and glucose transport. Trends Endocrinol.
Metab. 17, 72–78.
[60] Nicholas, G., Thomas, M., Langley, B., Somers, W., Patel, K., et al. (2002)
[81] Bonala, S., McFarlane, C., Ang, J., Lim, R., Lee, M., et al. (2013) Pid1
Titin-cap associates with, and regulates secretion of, Myostatin. J. Cell.
induces insulin resistance in both human and mouse skeletal muscle during
Physiol. 193, 120–131.
obesity. Mol. Endocrinol. 27, 1518–1535.
[61] Wang, H., Zhang, Q., and Zhu, D. (2003) hSGT interacts with the N-terminal
[82] Wu, W. L., Gan, W. H., Tong, M. L., Li, X. L., Dai, J. Z., et al. (2011) Over-
region of myostatin. Biochem. Biophys. Res. Commun. 311, 877–883.
expression of NYGGF4 (PID1) inhibits glucose transport in skeletal
[62] Amthor, H., Nicholas, G., McKinnell, I., Kemp, C. F., Sharma, M., et al.
myotubes by blocking the IRS1/PI3K/AKT insulin pathway. Mol. Genet.
(2004) Follistatin complexes Myostatin and antagonises Myostatin-mediated
Metab. 102, 374–377.
inhibition of myogenesis. Dev. Biol. 270, 19–30.
[83] Zhao, Y., Zhang, C., Chen, X., Gao, C., Ji, C., et al. (2010) Overexpression of
[63] Matzuk, M. M., Lu, N., Vogel, H., Sellheyer, K., Roop, D. R., et al. (1995) NYGGF4 (PID1) induces mitochondrial impairment in 3T3-L1 adipocytes.
Multiple defects and perinatal death in mice deficient in follistatin. Nature Mol. Cell. Biochem. 340, 41–48.
374, 360–363. [84] Chen, Y., Cao, L., Ye, J., and Zhu, D. (2009) Upregulation of myostatin gene
[64] Hill, J. J., Qiu, Y., Hewick, R. M., and Wolfman, N. M. (2003) Regulation of expression in streptozotocin-induced type 1 diabetes mice is attenuated by
myostatin in vivo by growth and differentiation factor-associated serum pro- insulin. Biochem. Biophys. Res. Commun. 388, 112–116.
tein-1: a novel protein with protease inhibitor and follistatin domains. Mol. [85] Lokireddy, S., Mouly, V., Butler-Browne, G., Gluckman, P. D., Sharma, M.,
Endocrinol. 17, 1144–1154. et al. (2011) Myostatin promotes the wasting of human myoblast cultures
[65] Monestier, O., Brun, C., Heu, K., Passet, B., Malhouroux, M., et al. (2012) through promoting ubiquitin-proteasome pathway-mediated loss of sarco-
Ubiquitous Gasp1 overexpression in mice leads mainly to a hypermuscular meric proteins. Am. J. Physiol. Cell. Physiol. 301, C1316–C1324.
phenotype. BMC Genomics 13, 541 [86] McFarlane, C., Plummer, E., Thomas, M., Hennebry, A., Ashby, M., et al.
[66] Bonala, S., Lokireddy, S., Arigela, H., Teng, S., Wahli, W., et al. (2012) (2006) Myostatin induces cachexia by activating the ubiquitin proteolytic
Peroxisome proliferator-activated receptor beta/delta induces myogenesis system through an NF-kappaB-independent, FoxO1-dependent mechanism.
by modulating myostatin activity. J. Biol. Chem. 287, 12935–12951. J. Cell. Physiol. 209, 501–514.

598 Myostatin
[87] Trendelenburg, A. U., Meyer, A., Rohner, D., Boyle, J., Hatakeyama, S., [108] Costelli, P., Muscaritoli, M., Bonetto, A., Penna, F., Reffo, P., et al. (2008)
et al. (2009) Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting Muscle myostatin signalling is enhanced in experimental cancer cachexia.
myoblast differentiation and myotube size. Am. J. Physiol. Cell. Physiol. Eur. J. Clin. Invest. 38, 531–538.
296, C1258–C1270. [109] Liu, C. M., Yang, Z., Liu, C. W., Wang, R., Tien, P., et al. (2008) Myostatin
[88] Durieux, A. C., Amirouche, A., Banzet, S., Koulmann, N., Bonnefoy, R., et al. antisense RNA-mediated muscle growth in normal and cancer cachexia
(2007) Ectopic expression of myostatin induces atrophy of adult skeletal mice. Gene Ther. 15, 155–160.
muscle by decreasing muscle gene expression. Endocrinology 148, 3140– [110] Bonetto, A., Penna, F., Minero, V. G., Reffo, P., Bonelli, G., et al. (2009)
3147. Deacetylase inhibitors modulate the myostatin/follistatin axis without
[89] Doucet, M., Dube, A., Joanisse, D. R., Debigare, R., Michaud, A., et al. (2010) improving cachexia in tumor-bearing mice. Curr. Cancer Drug Targets 9,
Atrophy and hypertrophy signalling of the quadriceps and diaphragm in 608–616.
COPD. Thorax 65, 963–970. [111] Chacon-Cabrera, A., Fermoselle, C., Urtreger, A. J., Mateu-Jimenez, M.,
[90] Schols, A. M., and Gosker, H. R. (2009) The pathophysiology of cachexia in Diament, M. J., et al. (2014) Pharmacological strategies in lung cancer-
chronic obstructive pulmonary disease. Curr. Opin. Support. Palliat. Care 3, induced cachexia: effects on muscle proteolysis, autophagy, structure, and
282–287. weakness. J. Cell. Physiol. 229, 1660–1672.
[91] Barreiro, E., Schols, A. M., Polkey, M. I., Galdiz, J. B., Gosker, H. R., et al. [112] Zhou, X., Wang, J. L., Lu, J., Song, Y., Kwak, K. S., et al. (2010) Reversal of
(2008) Cytokine profile in quadriceps muscles of patients with severe COPD. cancer cachexia and muscle wasting by ActRIIB antagonism leads to
Thorax 63, 100–107. prolonged survival. Cell 142, 531–543.
[92] van Helvoort, H. A., Heijdra, Y. F., de Boer, R. C., Swinkels, A., Thijs, H. M., [113] Padrao, A. I., Oliveira, P., Vitorino, R., Colaco, B., Pires, M. J., et al. (2013)
et al. (2007) Six-minute walking-induced systemic inflammation and oxida- Bladder cancer-induced skeletal muscle wasting: disclosing the role of
tive stress in muscle-wasted COPD patients. Chest 131, 439–445. mitochondria plasticity. Int. J. Biochem. Cell. Biol. 45, 1399–1409.
[93] Fermoselle, C., Rabinovich, R., Ausin, P., Puig-Vilanova, E., Coronell, C., [114] Murphy, K. T., Chee, A., Gleeson, B. G., Naim, T., Swiderski, K., et al.
et al. (2012) Does oxidative stress modulate limb muscle atrophy in severe (2011) Antibody-directed myostatin inhibition enhances muscle mass and
COPD patients? Eur. Respir. J. 40, 851–862. function in tumor-bearing mice. Am. J. Physiol. Regul. Integr. Comp. Phys-
iol. 301, R716–R726.
[94] Man, W. D., Natanek, S. A., Riddoch-Contreras, J., Lewis, A., Marsh, G. S.,
[115] Benny Klimek, M. E., Aydogdu, T., Link, M. J., Pons, M., Koniaris, L. G.,
et al. (2010) Quadriceps myostatin expression in COPD. Eur. Respir. J. 36,
et al. (2010) Acute inhibition of myostatin-family proteins preserves skele-
686–688.
tal muscle in mouse models of cancer cachexia. Biochem. Biophys. Res.
[95] Plant, P. J., Brooks, D., Faughnan, M., Bayley, T., Bain, J., et al. (2010)
Commun. 391, 1548–1554.
Cellular markers of muscle atrophy in chronic obstructive pulmonary
[116] Busquets, S., Toledo, M., Orpi, M., Massa, D., Porta, M., et al. (2012) Myostatin
disease. Am. J. Respir. Cell. Mol. Biol. 42, 461–471.
blockage using actRIIB antagonism in mice bearing the Lewis lung carcinoma
[96] Dasarathy, S. (2012) Consilience in sarcopenia of cirrhosis. J. Cachexia
results in the improvement of muscle wasting and physical performance. J.
Sarcopenia Muscle 3, 225–237.
Cachexia Sarcopenia Muscle 3, 37–43.
[97] Tsien, C., Davuluri, G., Singh, D., Allawy, A., Ten Have, G. A., et al.
[117] Gallot, Y. S., Durieux, A. C., Castells, J., Desgeorges, M. M., Vernus, B.,
Metabolic and molecular responses to leucine-enriched branched chain
et al. (2014) Myostatin gene inactivation prevents skeletal muscle wasting
amino acid supplementation in the skeletal muscle of alcoholic cirrhosis.
in cancer. Cancer Res. 74, 7344–7356.
Hepatology 61, 2018–2029.
[118] Lee, J. Y., Hopkinson, N. S., and Kemp, P. R. (2011) Myostatin induces
[98] Dasarathy, S. (2013) Posttransplant sarcopenia: an underrecognized early
autophagy in skeletal muscle in vitro. Biochem. Biophys. Res. Commun.
consequence of liver transplantation. Dig. Dis. Sci. 58, 3103–3111.
415, 632–636.
[99] Sakuma, K., and Yamaguchi, A. (2010) The functional role of calcineurin in
[119] Sim, F. J., Keyoung, H. M., Goldman, J. E., Kim, D. K., Jung, H. W., et al.
hypertrophy, regeneration, and disorders of skeletal muscle. J. Biomed.
(2006) Neurocytoma is a tumor of adult neuronal progenitor cells. J.
Biotechnol. 2010, 8.
Neurosci. 26, 12544–12555.
[100] Tsien, C., Garber, A., Narayanan, A., Shah, S. N., Barnes, D., et al. (2014)
[120] D’Orlando, C., Marzetti, E., Francois, S., Lorenzi, M., Conti, V., et al. (2014)
Post-liver transplantation sarcopenia in cirrhosis: a prospective evaluation.
Gastric cancer does not affect the expression of atrophy-related genes in
J. Gastroenterol. Hepatol. 29, 1250–1257.
human skeletal muscle. Muscle Nerve 49, 528–533.
[101] Zhang, L., Rajan, V., Lin, E., Hu, Z., Han, H. Q., et al. (2011) [121] Bonetto, A., Penna, F., Aversa, Z., Mercantini, P., Baccino, F. M., et al.
Pharmacological inhibition of myostatin suppresses systemic inflammation (2013) Early changes of muscle insulin-like growth factor-1 and myostatin
and muscle atrophy in mice with chronic kidney disease. Faseb J. 25, gene expression in gastric cancer patients. Muscle Nerve 48, 387–392.
1653–1663. [122] Breitbart, A., Scharf, G. M., Duncker, D., Widera, C., Gottlieb, J., et al. (2013)
[102] Zhang, L., Pan, J., Dong, Y., Tweardy, D. J., Dong, Y., et al. (2013) Stat3 Highly specific detection of myostatin prodomain by an immunoradiometric
activation links a C/EBPdelta to myostatin pathway to stimulate loss of sandwich assay in serum of healthy individuals and patients. PLoS One 8,
muscle mass. Cell. Metab. 18, 368–379. e80454
[103] Wang, D.-T., Yang, Y.-J., Huang, R.-H., Zhang, Z.-H., and Lin, X. (2014) [123] Jiang, M. S., Liang, L. F., Wang, S., Ratovitski, T., Holmstrom, J., et al. (2004)
Myostatin activates the ubiquitin-proteasome and autophagy-lysosome Characterization and identification of the inhibitory domain of GDF-8 propep-
systems contributing to muscle wasting in chronic kidney disease. Oxid. tide. Biochem. Biophys. Res. Commun. 315, 525–531.
Med. Cell. Longev, In Press. [124] Smith, R. C., and Lin, B. K. (2013) Myostatin inhibitors as therapies for
[104] Argiles, J. M., Fontes-Oliveira, C. C., Toledo, M., Lopez-Soriano, F. J., and muscle wasting associated with cancer and other disorders. Curr. Opin.
Busquets, S. (2014) Cachexia: a problem of energetic inefficiency. Support. Palliat. Care 7, 352–360.
J Cachexia Sarcopenia Muscle 5, 279–286. [125] Kim, T. N., and Choi, K. M. (2013) Sarcopenia: definition, epidemiology,
[105] Tisdale, M. J. (2010) Cancer cachexia. Curr. Opin. Gastroenterol. 26, 146– and pathophysiology. J. Bone Metab. 20, 1–10.
151. [126] Pereira, A. F., Silva, A. J., Matos Costa, A., Monteiro, A. M., Bastos, E. M.,
[106] Fearon, K. C. (2008) Cancer cachexia: developing multimodal therapy for a et al. (2013) Muscle tissue changes with aging. Acta Med. Port 26, 51–55.
multidimensional problem. Eur. J. Cancer 44, 1124–1132. [127] Baumann, A. P., Ibebunjo, C., G. W. A., and Paralkar, V. M. (2003) Myostatin
[107] Tisdale, M. J. (2009) Mechanisms of cancer cachexia. Physiol. Rev. 89, expression in age and denervation-induced skeletal muscle atrophy. J. Mus-
381–410. culoskelet. Neuron Interact. 3, 8–16.

SHARMA ET AL. 599


IUBMB LIFE

[128] Haddad, F., and Adams, G. R. (2006) Aging-sensitive cellular and increased in 60-92 year old women and men with muscle wasting. J. Nutr.
molecular mechanisms associated with skeletal muscle hypertrophy. J. Health Aging 6, 343–348.
Appl. Physiol. 100, 1188–1203. [138] Patel, H. P., Al-Shanti, N., Davies, L. C., Barton, S. J., Grounds, M. D., et al.
[129] Ko, I. G., Jeong, J. W., Kim, Y. H., Jee, Y. S., Kim, S. E., et al. (2014) (2014) Lean mass, muscle strength and gene expression in community
Aerobic exercise affects myostatin expression in aged rat skeletal muscles: dwelling older men: findings from the Hertfordshire Sarcopenia Study
a possibility of antiaging effects of aerobic exercise related with pelvic (HSS). Calcif. Tissue Int 95, 308–316.
floor muscle and urethral rhabdosphincter. Int. Neurourol. J. 18, 77–85. [139] Morissette, M. R., Stricker, J. C., Rosenberg, M. A., Buranasombati, C.,
[130] Kawada, S., Tachi, C., and Ishii, N. (2001) Content and localization of Levitan, E. B., et al. (2009) Effects of myostatin deletion in aging mice.
myostatin in mouse skeletal muscles during aging, mechanical unloading Aging Cell 8, 573–583.
and reloading. J. Muscle Res. Cell. Motil. 22, 627–633. [140] Siriett, V., Platt, L., Salerno, M. S., Ling, N., Kambadur, R., et al. (2006)
[131] Dennis, R. A., Przybyla, B., Gurley, C., Kortebein, P. M., Simpson, P., et al. Prolonged absence of myostatin reduces sarcopenia. J. Cell. Physiol. 209,
(2008) Aging alters gene expression of growth and remodeling factors in 866–873.
[141] Wagner, K. R., Liu, X., Chang, X., and Allen, R. E. (2005) Muscle
human skeletal muscle both at rest and in response to acute resistance
regeneration in the prolonged absence of myostatin. Proc. Natl. Acad Sci.
exercise. Physiol. Genomics 32, 393–400.
USA 102, 2519–2524.
[132] Marcell, T. J., Harman, S. M., Urban, R. J., Metz, D. D., Rodgers, B. D.,
[142] Siriett, V., Salerno, M. S., Berry, C., Nicholas, G., Bower, R., et al. (2007)
et al. (2001) Comparison of GH, IGF-I, and testosterone with mRNA of
Antagonism of Myostatin Enhances Muscle Regeneration During
receptors and myostatin in skeletal muscle in older men. Am. J. Physiol.
Sarcopenia. Mol. Ther 15, 1463–1470.
Endocrinol. Metab. 281, E1159–E1164.
[143] LeBrasseur, N. K., Schelhorn, T. M., Bernardo, B. L., Cosgrove, P. G., Loria,
[133] Welle, S., Bhatt, K., Shah, B., and Thornton, C. (2002) Insulin-like growth
P. M., et al. (2009) Myostatin inhibition enhances the effects of exercise on
factor-1 and myostatin mRNA expression in muscle: comparison between
performance and metabolic outcomes in aged mice. J. Gerontol. A Biol.
62-77 and 21-31 yr old men. Exp. Gerontol. 37, 833–839.
Sci. Med. Sci. 64, 940–948.
[134] Ratkevicius, A., Joyson, A., Selmer, I., Dhanani, T., Grierson, C., et al. [144] Murphy, K. T., Koopman, R., Naim, T., Leger, B., Trieu, J., et al. (2010)
(2011) Serum concentrations of myostatin and myostatin-interacting pro- Antibody-directed myostatin inhibition in 21-mo-old mice reveals novel
teins do not differ between young and sarcopenic elderly men. J. Gerontol. roles for myostatin signaling in skeletal muscle structure and function.
A Biol. Sci. Med. Sci. 66, 620–626. FASEB J. 24, 4433–4442.
[135] Raue, U., Slivka, D., Jemiolo, B., Hollon, C., and Trappe, S. (2006) [145] Arounleut, P., Bialek, P., Liang, L. F., Upadhyay, S., Fulzele, S., et al. (2013)
Myogenic gene expression at rest and after a bout of resistance exercise A myostatin inhibitor (propeptide-Fc) increases muscle mass and muscle
in young (18-30 yr) and old (80-89 yr) women. J. Appl. Physiol. 101, 53–59. fiber size in aged mice but does not increase bone density or bone
[136] Schulte, J. N., and Yarasheski, K. E. (2001) Effects of resistance training on strength. Exp. Gerontol. 48, 898–904.
the rate of muscle protein synthesis in frail elderly people. Int. J. Sport [146] Chiu, C. S., Peekhaus, N., Weber, H., Adamski, S., Murray, E. M., et al.
Nutr. Exerc. Metab. 11 Suppl, S111–S118. (2013) Increased muscle force production and bone mineral density in
[137] Yarasheski, K. E., Bhasin, S., Sinha-Hikim, I., Pak-Loduca, J., and Gonzalez- ActRIIB-Fc-treated mature rodents. J. Gerontol. A Biol. Sci. Med. Sci. 68,
Cadavid, N. F. (2002) Serum myostatin-immunoreactive protein is 1181–1192.

600 Myostatin

You might also like