Anabolic and Catabolic Signaling Pathways that

0 downloads 0 Views 303KB Size Report
review of catabolic signaling pathways that promote muscle atrophy by ...... mechanisms for beta-adrenergic activation of NKCC activity in skeletal muscle.
Anabolic and Catabolic Signaling Pathways that Regulate Skeletal Muscle Mass

John J. McCarthy1,2*, Kevin A. Murach1,3

Contact information:

800 South Rose St. 1

Center for Muscle Biology

2

Department of Physiology

3

Department of Rehabilitation Sciences

College of Medicine University of Kentucky Lexington, KY 40536 *Corresponding Author Phone:

(859) 323-4730

Fax:

(859) 323-1070

e-mail:

[email protected]

Abstract Skeletal muscle mass is primarily dictated by the balance between the rates of protein synthesis and degradation. Over the last decade, significant progress has been made in defining the anabolic and catabolic signaling pathways that control skeletal muscle mass through the regulation of protein synthesis and degradation. The purpose of this review is to briefly describe known and emerging signaling pathways involved in the regulation of skeletal muscle mass. Two important themes have come to light: 1) the degree of crosstalk between and among anabolic and catabolic signaling pathways involved in the control of skeletal muscle hypertrophy and atrophy, and 2) the balance between anabolism and catabolism that is required to facilitate a proper hypertrophic response. A more in-depth understanding of the anabolic and catabolic signaling pathways that regulate skeletal muscle mass will provide a critical foundation for the development of more effective training programs and nutritional aids to enhance athletic performance.

Key words AMPK, β-adrenergic, β-catenin, anabolic, atrophy, catabolic, FoxO, hypertrophy, MAFbx, mechanotransduction, MuRF-1, myostatin, NF-κB, nitric oxide, PGC-1α4, ribosome biogenesis, TORC1, TRPV1, YAP/TAZ

Introduction Skeletal muscle is one of the abundant tissues of the human body, accounting for roughly 40% of the body mass in men [1]. In addition to its primary function in locomotion, there is a growing recognition that skeletal muscle has an important role in whole-body metabolism and protein homeostasis during aging [2, 3]. The clinical importance of skeletal muscle health is highlighted by the diverse patient population reported to show significant losses in skeletal muscle mass including those suffering from systemic diseases (cancer, sepsis or HIV-AIDS), organ failure (cirrhosis, chronic kidney disease and chronic heart failure) or inactivity (as a result of obesity, rheumatoid arthritis or prolonged bed rest) as well as aging (sarcopenia). Currently, the most effective therapy for maintaining or restoring muscle mass is resistance exercise, which is often not a realistic option for the aforementioned patient populations. Given this state of affairs, there is great interest in defining the anabolic and catabolic signaling pathways that regulate skeletal muscle mass as the basis for developing more effective therapies to prevent or restore the loss of muscle mass associated with systemic disease, bed rest, and aging. Moreover, a better understanding of the signaling pathways that regulate skeletal muscle mass will allow for the design of more effective training and nutritional programs aimed at increasing muscle mass in athletes with the goal of improving performance.

History One of the most remarkable qualities of skeletal muscle, that is often taken for granted, is its ability to specifically alter its physical characteristics (phenotype) in response to a particular type of contractile activity. The most obvious example of this phenotypic plasticity is the significant increase in skeletal muscle mass following a progressive, high-resistance exercise training program. The observation that resistance exercise can increase muscle mass dates back to the to the ancient Greeks when it was reputed that Milo of Crotona achieved his great strength by carrying a calf on his back every day until it was a bull. Current resistance exercise training programs have replaced the bull with barbells and dumbbells, prescribing three sets of 8-12 repetitions for each exercise, performed on alternating days, three times a week [4]. The dramatic increase in muscle

size following resistance exercise is primarily the result of an increase in muscle fiber size (hypertrophy). The contribution from an increase in muscle fiber number (hyperplasia) is likely minor under normal conditions [5-9], but appears to become more prevalent under circumstances of extreme loading and/or hypertrophy in both humans and rodents [10-17].

Skeletal muscle mass is primarily dictated by the balance between the rates of protein synthesis and degradation. Increasing protein synthesis, decreasing protein breakdown, or modulating both factors can result in a net increase in the rate of protein synthesis which leads to muscle hypertrophy [18]. Accordingly, resistance exercise has been shown in both humans and rodents to cause a net increase in the rate of protein synthesis [18-24], and this is primarily dictated by intrinsic cellular processes, and not necessarily by transient alterations in systemic hormones such as testosterone [25]. Recent work in humans has shown that the initial myofibrillar protein synthetic response to resistance exercise in untrained muscle is not necessarily predictive of the hypertrophic response to training [26, 27]. However, once muscle damage characteristic of the first few weeks of a resistance training program has subsided, myofibrillar protein synthesis rates do reflect hypertrophic potential [27].

Baar and Esser (1999) provided the first mechanistic data linking high-force contractions to muscle hypertrophy via the prolonged activation of mTOR (mechanistic target of rapamycin) signaling, the cell’s master regulator of protein synthesis [28-31]. Bodine and colleagues (2001) followed up with the seminal study demonstrating mTOR function was absolutely necessary for skeletal muscle hypertrophy [32]. The importance of mTOR activation was subsequently linked to hypertrophic growth in humans by Mayhew and co-workers (2009), showing that changes in translational signaling following a bout of unaccustomed resistance exercise was predictive of the hypertrophic response after 16 weeks of resistance exercise training [33]. While mTOR signaling is considered to be the primary pathway regulating skeletal muscle mass, other signaling pathways are shown to be capable of regulating skeletal muscle hypertrophy. The focus of this chapter will be to briefly discuss the role of mTOR signaling in muscle

hypertrophy (for an in-depth review of mTOR signaling, refer to the recent review by Saxton and Sabatini [34]), followed by a review of other anabolic signaling pathways involved in the regulation of skeletal muscle mass. The chapter will conclude with a review of catabolic signaling pathways that promote muscle atrophy by inhibiting protein synthesis and/or increasing the rate of protein degradation.

Anabolic signaling

Mechanotransduction: translating force to a hypertrophic signal During resistance exercise, muscle tension is a primary impetus for the growth response. As such, the first step in initiating an intracellular signaling cascade that results in hypertrophy is tension sensing. Skeletal muscle fibers perceive tension via a variety of elements within the muscle fiber membrane and intracellular cytoskeleton. Structures linking the muscle fiber membrane to the extracellular matrix, such as integrins, cadherins, and focal adhesion complex proteins, undergo conformational changes when force is applied to them, which affects intracellular behavior. For instance, when the extracellular matrix interacts with membrane-bound integrins, focal adhesion complexes form that mobilize ribosomes and mRNAs which facilitates translation [35]. Muscle cell culture experiments show that disruption of focal adhesion complex proteins blunts intracellular hypertrophic signaling [36]. It is also known that these focal adhesion complexes can directly activate ribosomal proteins to facilitate protein translation [37].

The intracellular actin cytoskeleton is also connected to integrins. Physical interactions between actin and intermediate filaments within skeletal muscle such as desmin and muscle ankryn repeat proteins (MARPs, which are associated with the giant scaffold protein titin) also play mechanosensing roles via these connections. Titin is now recognized as a central mediator of hypertrophic signaling [38, 39], and titin-associated MARPs can migrate away from titin and affect various signaling pathways as well as gene transcription [40]. The Z-disc, which titin and many other myofilaments are anchored to within the sarcomere, is also emerging as a central node for translating

mechanical tension to intracellular signaling [39, 41]. Furthermore, the primary intermediate filament, desmin, deforms myonuclei via direct force transmission from the cell membrane (which can affect transcription) and facilitates intracellular stress signaling [42].

Mechanotransduction in skeletal muscle is a relatively recent area of inquiry, so mechanosensing protein interactions and the consequences are not yet fully elucidated. Despite the relative immaturity of this area, significant progress has been made regarding mechanotransduction and activation of mTOR. Recent evidence indicates that phosphatidic acid (PA), a lipid second messenger, is responsive to mechanical load and directly activates mTOR independent from intermediate signaling [43-45]. This PA is generated from mechanical-sensitive diacylglycerol kinases found in the membrane structures of skeletal muscle [45]. Furthermore, PA supplementation increases muscle protein synthesis in rodents [46], and enhances hypertrophy from resistance exercise training in humans [47]. More work is needed to fully elucidate how mechanical tension is ultimately translated to skeletal muscle growth, but mTOR signaling appears to be a focal point in this process.

mTOR signaling mTOR (mechanistic target of rapamycin) is a member of the phosphoinositide 3-kinase (PI3K)-related kinase family that has been shown to form two separate multi-protein complexes designated mTOR complex 1 (TORC1) and 2 (mTORC2) [48]. mTORC1 is a master regulator of protein synthesis, integrating a number of upstream signals including growth factors (insulin, IGF-1), nutrients (amino acids) and mechanical strain; TORC2 is primarily involved in the regulation of cytoskeleton dynamics and cell proliferation and survival [48, 49].

Although it had been known for some time that insulin-like growth factor-1 is capable of inducing muscle fiber hypertrophy, the underlying mechanism remained unknown until Rommel and colleagues (2001) provided evidence showing that IGF-1 activated TORC1 via the PI3K/AKT pathway [50, 51]. Activation of the PI3K/AKT pathway by IGF-1

increases protein synthesis through AKT-mediated phosphorylation of tuberous sclerosis protein 2 (TSC2) protein, the primary inhibitor of TORC1 activity [52-55]. The phosphorylation of TSC2 by AKT inhibits its activity, resulting in the accumulation of the active form of Rheb (Ras homolog enriched in brain), a potent activator TORC1 [56, 57]. Rheb localizes with the late endosome/lysomal (LEL), along with other key modulators of mTOR such as PA and TSC2, indicating that the LEL is the key site of integration for mTOR signaling during hypertrophy [44]. Indeed, mechanical stimulation increases the association of mTOR with the LEL [58]. Furthermore, amino-acid-mediated stimulation of mTOR is Rheb-dependent, and mTORs association with the LEL is facilitated by amino-acid availability [59]. Once activated by Rheb, TORC1 stimulates protein synthesis by phosphorylating ribosomal S6 kinase (p70S6k1) and eukaryotic initiation factor 4E binding protein (4E-BP1), two factors that enhance the initiation of mRNA translation (thus promoting protein synthesis). In addition to these factors, TORC1 activation has been shown to increase the expression of the ε-subunit of eIF2B holoenzyme, thereby promoting the recruitment of the initiator methionine tRNA to the start codon and increasing protein synthesis [60]. The expression of eIF2Bε was shown to increase following resistance exercise training, with its abundance appearing to be an important determinant of the hypertrophic response [33, 61].

In a search for proteins that were targeted for degradation by the muscle-specific ubiquitin ligase Fbxo32 (commonly known as MAFbx or atrogin), Lagirand-Cantaloube and co-workers (2008) identified the eukaryotic initiation factor 3, subunit 5 (eIF3f) [62]. As the largest of the initiation factors, evidence indicates that the eIF3 complex is involved in nearly all aspects of translation initiation; most notable for the current discussion is the finding that eIF3f can serve as a docking site for TORC1 and p70S6k1 [63-65]. Upon activation, TORC1 is recruited to the eIF3f-p70S6K1 complex, leading to the phosphorylation and subsequent release of active p70S6K1, which is then capable of increasing protein synthesis by phosphorylating ribosomal protein S6 (rpS6) and eIF4B [64, 65]. In a series of studies, it was demonstrated that the in vivo and in vitro overexpression of eIF3f induced myotube and muscle fiber hypertrophy, respectively, by stimulating protein synthesis through the phosphorylation of p70S6K1, rpS6 and 4E-BP1

[62, 66]. It will be important to determine if the regulation of muscle hypertrophy by eIF3f is conserved in humans and what role it has in the hypertrophic response following resistance exercise.

Wnt/β-catenin signaling and ribosome biogenesis In their original description of IGF-1 mediated muscle hypertrophy, Rommel and colleagues (2001) also identified glycogen synthase kinase 3β (GSK3β) as a downstream target of PI3K/AKT signaling [51]. In contrast to TORC1, phosphorylation by AKT leads to GSK3β inactivation, which prevents further inhibition via phosphorylation of the translation initiation factor eIF2Bε [51]. The inhibition of GSK3β activity by IGF-1 or lithium (a known inhibitor of GSK3β) treatment, or over-expression of a dominant-negative form of GSK3β, all resulted in a significant myotube hypertrophy, thus confirming the importance of GSK3β inhibition by IGF-1/PI3K/AKT signaling in the regulation of muscle mass [51, 67].

In addition to eIF2Bε, the inactivation of GSK3β by IGF-1 via AKT phosphorylation also results in the stabilization of β-catenin protein [68]. β-catenin is known to exist in two separate intracellular pools; an actin cytoskeleton pool involved in the formation of adherens junctions and a cytoplasmic pool that is the downstream mediator of the Wnt signaling pathway involved in the regulation of gene expression [69]. Under resting conditions, cytoplasmic β-catenin is phosphorylated by GSK3β, targeting it for degradation by the proteasome [70]. Upon GSK3β inhibition, via phosphorylation of Ser9 by AKT or sequestration by Dishevelled following Wnt activation, β-catenin accumulates in the cytoplasm. β-catenin ultimately translocates to the nucleus where it regulates the expression of target genes such as c-Myc and cyclin D [71, 72]. These studies show that β-catenin levels can be regulated by a Wnt-dependent and independent mechanism. Following seven days of mechanical overload induced by synergist ablation, nuclear βcatenin levels increased by over 4-fold in the mouse plantaris muscle through a Wnt-

independent mechanism [73]. In a follow-up study, Armstrong and colleagues (2006) reported that the skeletal muscle-specific inactivation of β-catenin completely prevented muscle fiber hypertrophy, demonstrating the necessity of β-catenin in the regulation of muscle hypertrophy [74]. Associated with the increase in nuclear β-catenin levels during hypertrophy were increased myonuclear c-Myc expression (an established target gene of β-catenin) and a 3-fold increase in total RNA [73]. The increase in the total RNA pool is indicative of ribosome biogenesis given that ~85% of total RNA consists of ribosomal RNA [75]. A greater ribosomal content of the cell results in an increase in protein synthesis as a consequence of greater translational capacity. An increase in the translational capacity of the cell represents an additional mechanism for increasing the rate of protein synthesis in response to a hypertrophic stimulus. While the necessity of enhanced translational efficiency through TORC1 activation is well-established, the importance of increased translational capacity to skeletal muscle hypertrophy is an emerging area of interest [76]. Interestingly, in a review of the literature, Hannan et al., (2003) concluded that increased translational capacity was required for cardiac hypertrophy and that increased translational efficiency alone was not sufficient to promote muscle hypertrophy [77].

Although the exact mechanism(s) involved in the regulation of ribosome biogenesis during muscle hypertrophy have not been fully elucidated, evidence from a number of different studies supports a model in which increased β-catenin expression up-regulates c-Myc expression, which subsequently drives transcription of ribosomal DNA through the regulation of RNA polymerase I and components of the preinitiation complex [73, 78, 79]. Transcription of ribosomal DNA (rDNA) to rRNA by polymerase I is considered the major rate-limiting step in ribosome biogenesis [80]. Both the mitogen-activated protein kinase/extracellular signal-regulated pathway (MAPK/ERK), as well as the mTOR pathway, regulate Pol I formation and activation [81], indicating that these signaling cascades are also mediators of ribosome biogenesis. Pol I regulation via mTORC1 is controlled by upstream binding factor (UBF) [81], which dictates the number of active ribosomal genes (of which there are hundreds in the human genome) [82]. In culture, blocking mTORC1 in myotubes decreases UBF and rRNA abundance, thereby

preventing myotube hypertrophy [83]. Specifically blocking Pol I in muscle cell cultures has the same effect [84]. mTOR also has nuclear localization in muscle, directly interacting with the rDNA promoter and influencing ribosomal gene transcription [85]. The mechanistic evidence suggesting that translational capacity via ribosome biogenesis is a primary mediator of muscle hypertrophic potential is compelling. Observational studies in both rodents and humans also indicate that robust upregulation of ribosome biogenesis is characteristic of, and likely necessary for, successful muscle hypertrophy [84, 86-93]. Future loss- and gain-of-function studies will determine whether ribosome biogenesis and increased translational capacity are indispensable for muscle hypertrophy.

A non-canonical Wnt signaling pathway has been described that is also capable of inducing muscle hypertrophy independent of GSK3β or β-catenin [94]. Experiments in myotubes revealed that Wnt7a binding to the frizzled homolog 7 (Fzd7) receptor activated AKT via a PI3K mechanism that did not involve IGF-1 signaling [94]. As expected, AKT activation by Wnt7a resulted in an increase in TORC1 activity, as assessed by rpS6 phosphorylation, and an increase in myotube diameter [94]. Consistent with these findings, over-expression of Wnt7a in skeletal muscle fibers increased cross-sectional area 40-55%; however, whether or not this hypertrophy was caused by TORC1 activation and/or the increase satellite cells associated with Wnt7a over-expression, remains to be determined [94, 95]. β-adrenergic receptor signaling The β-adrenergic receptors (β-AR) are members of the guanine nucleotide-binding Gprotein-coupled receptor family with the β2 subtype being the most abundant in skeletal muscle [96-98]. Signaling through the β2-AR occurs primarily via coupling with the G protein Gαs in skeletal muscle, though there are reports describing signaling events involving Gαi and, more recently, Gβγ [99, 100]. β2-AR coupling to Gαs activates adenylyl cyclase production of cyclic-AMP (cAMP), which in turn activates downstream signaling through protein kinase A (PKA) [101]. Alternatively, β2-AR signaling involving

the Gβγ dimer initiates signaling through a PKA-independent pathway involving PI3K activation of AKT [102-104]. Historically, β-adrenergic receptor agonists (β-agonists) have been used to treat patients suffering from bronchial ailments such as asthma; however, Emery and coworkers (1984) made the fortuitous discovery that administration of the β-agonist clenbuterol caused skeletal muscle hypertrophy in rats and was associated with a 34% increase in muscle protein synthesis [105]. The ability of β-agonists to increase skeletal muscle mass has since been confirmed by numerous studies and shown to be effective in humans as well (reviewed in Lynch GS, 2008) [101]. The increase in muscle mass following β-agonist administration is the result of hypertrophy (an increase in cell size) and not hyperplasia (an increase in cell number) or satellite cell activity [106-108]. The mechanism through which β-agonists exert their anabolic effect has been attributed to enhanced protein synthesis, though studies have reported a decrease in protein degradation as a contributing factor [105, 109-114]. The increase in skeletal muscle mass following 14 days of clenbuterol treatment was completely blocked by coadministration of rapamycin, a potent inhibitor of TORC1 signaling, indicating the anabolic action of β-agonists is the result of an increase in protein synthesis via TORC1 activation [115]. In support of this mechanism, the same authors reported that clenbuterol activated TORC1 signaling, as evidenced by increased phosphorylation of AKT, p70S6K1 and 4E-BP1 [115]. Moreover, these findings suggest that in skeletal muscle, β-agonists function through a β2-AR/ Gβγ complex that activates PI3K/AKT signaling. Interestingly, rapamycin or triciribine (an AKT inhibitor) was unable to block the muscle-sparing effects of clenbuterol following muscle denervation suggesting a different mode of action, possibly involving cAMP/PKA pathway downstream of β2-AR/ Gαs [115, 116]. Upstream in β2-AR signaling, GRK2, a G-protein coupled receptor kinase, plays an integral role in desensitization of adrenergic receptors following stimulation. Eventual desensitization of β2-ARs is of critical importance specifically in heart muscle, since continued activation could lead to pathology and loss of function. In

skeletal muscle, however, loss of GRK2 leads to enhanced hypertrophy following clenbuterol treatment through increased AKT signaling [117]. Worth noting is that β2AR-mediated hypertrophy is not always functional, resulting in reduced normalized strength [117, 118], which is most noticeable in fast-twitch predominant muscles [117, 119]. Impaired relative force production via β2-AR stimulation highlights that successful skeletal muscle hypertrophy is a complex and interrelated process that requires coordination at multiple levels to ultimately facilitate proper function.

In addition to Gαs and Gβγ, lysophosphotidic acid (LPA)-induced muscle hypertrophy was uncovered that involved the Gαi2 isoform [120]. In a series of loss- and gain-offunction experiments, Minetti and colleagues (2011) showed that LPA-induced myotube hypertrophy was mediated by a 40% increase in protein synthesis via TORC1 activation, as indicated by increased phosphorylation of its downstream targets p70S6K1 and rpS6 [120]. TORC1 activation by LPA, however, was independent of AKT but required protein kinase C (PKC) inhibition of GSK3β, though the mechanism linking PKC to TORC1 activation remains to be identified [120]. Furthermore, in vivo upregulation of β2-AR via adeno-associated virus induces hypertrophy independent from mTOR activation, suggesting an alternative pathway can contribute to β2-AR-mediated hypertrophy [121].

Emerging pathways Unlike PI3K/AKT/TORC1 and β2-AR signaling pathways, there are less well-established signaling pathways that have been shown to be capable of regulating muscle hypertrophy. Although it is still too early to know the importance of these “new” pathways in human skeletal muscle hypertrophy, an understanding of how each of these pathways function will provide a more comprehensive knowledge of the signaling pathways that regulate skeletal muscle hypertrophy.

Nitric oxide (NO) signaling Inhibition of nitric oxide synthase (NOS) by NG-nitro-L-arginine methyl ester (L-NAME) administration significantly blunted muscle hypertrophy induced by mechanical overload

of the rat plantaris muscle [122, 123]. Building on these early studies, Ito and colleagues (2013) uncovered a signaling pathway by which neuronal NOS (nNOS) production of nitric oxide mediates muscle hypertrophy [124]. Upon mechanical loading, nNOS derived nitric oxide reacts with superoxide to generate peroxynitrite. Peroxynitrite then activates the TRPV1 (transient receptor potential cation channel, subfamily V, member 1) channel causing an increase in intracellular Ca2+ levels via release from the sarcoplasmic reticulum. Elevation of intracellular Ca2+ causes an increase in protein synthesis through TORC1 activation by an unknown mechanism; though speculative at this time, the authors proposed a similar Ca2+/calmodulin mechanism as that used by amino acids to stimulate TORC1 activity [124, 125]. In agreement with these findings, the administration of the TRPV1 agonist capsaicin activated TORC1 signaling to a comparable level as that observed with mechanical overload [124]. Germline deletion of NOS production elicits a mitochondrial unfolded protein response as well as an upregulation in protein degradative pathways, resulting in impaired muscle growth during development [126]. These findings allude to the important but often overlooked relationship between mitochondrial function (which provide ATP for biosynthetic processes like protein synthesis and ribosome biogenesis) and hypertrophic potential. NOS scavenges reactive oxygen species (ROS) such as superoxides, which cause oxidative stress and can be damaging to the cell. However, a physiological balance of ROS seem to promote hypertrophy [127]. More work is needed to fully elucidate the mechanisms whereby NOS, either directly or indirectly, contributes to hypertrophy. PGC-α4 signaling PGC-1α (peroxisome proliferative activated receptor, gamma, coactivator 1 alpha) was originally identified as a coactivator of PPARγ in brown adipose tissue, but has since been shown to have an important role in skeletal muscle adaptation to exercise [128, 129]. Although the over-expression of PGC-1α in skeletal muscle was found to ameliorate the loss of muscle mass caused by denervation, there was no evidence to suggest PGC-1α might have a role in regulating skeletal muscle hypertrophy [130]. Recently, Ruas et al., (2012) identified a splice variant of PGC-1α, PGC-1α4, capable of inducing muscle fiber hypertrophy when over-expressed both in vitro and in vivo [131].

The mechanism through which PGC-1α4 promotes hypertrophic growth appears to be through the down-regulation of myostatin expression while simultaneously increasing IGF-1 expression [131]. Importantly, in humans, PGC-1α4 expression was found to be dramatically increased in response to a 8-week training program consisting of both resistance and endurance exercises [131]. Moreover, leg press performance (number of repetitions) following the training program correlated with the increase in PGC-1α4 expression, suggesting PGC-1α4 expression might serve as a readout for optimizing a strength training program [131]. Further investigation showed that PGC-1α4 transcription is dependent on G protein-coupled receptor 56 (GPR56); over-expression of this receptor induces myotube hypertrophy while knockdown attenuates hypertrophy in mice, and receptor regulation is enhanced during hypertrophy in mice and humans [132]. Since the original work showing a connection between PGC-1α4 and hypertrophy with resistance exercise, others have shown that this PGC-1α splice variant is similarly up-regulated with endurance exercise [133, 134] and hypoxia [135]. More work is needed to determine whether PGC-1α4 drives hypertrophy, or is part of the global remodeling response to exercise in humans.

microRNAs MicroRNAs (miRs) are small (~ 22 nucleotides), noncoding RNAs that regulate gene expression through a post-transcriptional mechanism [136]. A small family of musclespecific miRs, referred to as myomiRs, have been shown to have an important role in muscle development and disease [137]. In response to mechanical overload in mice, myomiR expression is drastically altered, suggesting a regulatory role in the hypertrophic process [138]. For instance, miR-1 and -133a are known to target growth factor transcripts for degradation (specifically IGF1); these two myomiRs are downregulated during overload, which may allow for up-regulation of growth factor synthesis in skeletal muscle to promote hypertrophy [138]. The expression of myomiRs has also been found to change in response to both resistance and endurance exercise following a single bout or with training in humans [139]. Davidsen and co-workers (2011) identified a small group of miRs that were differentially expressed between low- and high-responders following a 12-week resistance exercise program [140]. In particular,

the change in miR-378 expression in response to training was positively correlated with gains in skeletal muscle mass [140]. Most recently, it has been reported that myomiRs are stabilized via packaging into extracellular vesicles (e.g. exosomes) or via chaperone proteins and effluxed from skeletal muscle under various conditions [141, 142]. Trafficking of miRNAs appears to be a method of intercellular regulation between and amongst tissues. For example, muscle stem cells (known as satellite cells) release miR206 into the microenvironment which specifically regulates proper extracellular matrix (ECM) deposition during hypertrophy [143]. ECM remodeling is necessary for coordinating the hypertrophic process, but excessive ECM can inhibit muscle hypertrophy [143, 144]. Release of miRNAs from muscle fibers into the extracellular space could also serve to clear specific myoMirs from muscle in order to facilitate growth processes [145]. To have a better understanding of the molecular mechanisms through which myomiRs regulate skeletal muscle hypertrophy, future studies will need to focus on identifying target genes and their connection to anabolic signaling.

Catabolic signaling As dramatic as the increase in skeletal muscle mass can be as the result of resistance exercise training, so the loss of skeletal muscle mass can be following periods of disuse. Muscle atrophy can also occur as the consequence of certain systemic diseases (cancer, sepsis or HIV-AIDS), organ failure (cirrhosis, chronic kidney disease and chronic heart failure), inactivity (as a result of rheumatoid arthritis or prolonged bed rest) and aging. In contrast to muscle hypertrophy, muscle atrophy comes about when the rate of protein degradation exceeds the rate of protein synthesis, either through an increased degradation and/or decreased protein synthesis.

In an effort to determine if a common mechanism might underlie muscle atrophy brought about by differing catabolic conditions, Bodine and colleagues (2001) searched for genes that shared a similar pattern of expression in response to denervation, immobilization, and hind limb unloading [146]. Two genes, MAFbx (Muscle Atrophy Fbox) and MuRF-1 (Muscle RING Finger 1), were identified that were significantly upregulated in all three models of muscle atrophy [146]. Concurrently, Gomes and co-

workers reported the identification of atrogin-1 (also known as MAFbx) in atrophying muscle caused by food deprivation, diabetes, cancer and renal failure [147]. Further analysis revealed that both MAFbx and MuRF-1 are skeletal muscle-specific ubiquitin ligases involved in protein degradation through the ubiquitin-proteosome (UPP) system [146, 147]. The central importance of these two genes to the skeletal muscle atrophy process was confirmed by experiments showing that inactivation of either MAFbx or MuRF-1 resulted in a blunted atrophic response under catabolic conditions [146]. Recently, UPP activation independent from MAFBx and MuRF-1 has been observed during skeletal muscle remodeling [148]. Additional ubiquitin ligases, such as MUSA1 [149] and Nedd4-1 [150, 151], have also been implicated in the UPP system and could emerge as important regulators of muscle catabolism.

Shifting protein balance via UPP activation to favor breakdown generally results in atrophy, but it is important to highlight that a certain degree of breakdown is important for maintaining protein integrity and an optimal hypertrophic response [152, 153]. For instance, muscle-specific knockout of a protein that is essential for proper proteasome activity impairs muscle growth and function during post-natal development [154]. Studies in humans also report acute up-regulation of MAFBx and MuRF-1 after acute resistance exercise [155], which highlights that muscle protein breakdown is a component of the hypertrophic process. A controlled level of breakdown likely supports amino acid-reallocation during times of growth, and also prevents the aggregation of mis-folded and non-functional proteins. More work is needed to understand the complex interplay between protein breakdown and synthesis in mediating muscle fiber size.

AKT/Foxo signaling The ability of IGF-1 to block MAFbx expression under catabolic conditions induced by the synthetic glucocorticoid dexamethasone indicated, paradoxically, that the PI3K/AKT signaling pathway was involved in the regulation of muscle atrophy [156]. A downstream target of PI3K/AKT signaling through which IGF-1 could inhibit MAFbx expression was the Forkhead box O (FoxO) class of transcription factors; phosphorylation by AKT causes the Foxo protein to remain sequestered in the cytoplasm, unable to activate

transcription of target genes such as MAFbx and MuRF-1 [157, 158]. Subsequent studies confirmed such a mechanism by providing evidence demonstrating that IGF-1 activation of AKT leads to repression of MAFbx and MuRF-1 expression through FoxO phosphorylation [159, 160]. Further, over-expression of a constitutively active form of FoxO3 isoform caused a significant increase in MAFbx expression and reduced myotube diameter by 50% [159]. Conversely, Southgate and colleagues (2007) reported that FoxO1 was also able to promote muscle atrophy by inhibiting TORC1 signaling through the up-regulation of 4E-BP1 expression [161]. In addition to describing a key signaling pathway regulating muscle atrophy, these studies reveal the degree of crosstalk between anabolic and catabolic pathways in skeletal muscle.

Further evidence of the degree of functional overlap between anabolic and catabolic signaling is highlighted by the importance of FoxO3 in mediating autophagy. Separate from the UPP system, autophagy is another mechanism of protein breakdown that utilizes autophagosomes and lysosomes to facilitate systematic degradation and recycling of cellular components. In skeletal muscle, FoxO3 directly controls autophagy through the transcription of autophagy-related genes [162]. Although autophagy is a protein degradative process, and excessive autophagy contributes to atrophy in various catabolic conditions [163], inhibition of autophagy also results in atrophy and myopathy and is required to maintain muscle mass [162, 164]. This again highlights that a fine balance between protein synthesis and breakdown is required in order to undergo optimal muscle growth.

Given that the UPP is the major contributor to muscle protein degradation, and both MAFbx and MuRF-1 are E3 ubiquitin ligases, identifying the proteins each one targets for degradation is important for understanding their respective catabolic action and how it promotes muscle atrophy. MAFbx had been shown to target eIF3f and MyoD for degradation whereas thick myofilament proteins, such as myosin-binding protein C, myosin light chains 1 and 2 and myosin heavy chain, are targeted for degradation by MuRF-1 [62, 165, 166]. These findings suggest MAFbx and MuRF-1 have distinct functions in muscle atrophy; MAFbx modulates the expression level of genes involved in

regulating protein synthesis while MuRF-1 influences the level of protein degradation [167].

Myostatin signaling Myostatin is a member of the transforming growth factor-β (TGF-β) superfamily that acts as a potent negative regulator of skeletal muscle mass [168]. Myostatin is a “myokine”, or a hormone-like protein (cytokine) released from muscle, that can act in an autocrine or paracrine fashion. Myostatin binds the activin type IIB receptor, which in turn activates activin receptor-like kinase 4 (ALK4) or ALK5 receptor and initiates downstream signaling through the Smad2/Smad3 transcription factor complex [169]. While the evidence is clear that inactivation of myostatin expression or activity promotes skeletal muscle hypertrophy, the mechanism by which myostatin induces muscle atrophy remains to be fully defined [168, 170, 171]. Studies have shown that myostatin is capable of inhibiting AKT/TORC1 signaling through a Smad2/3-dependent mechanism [170, 172, 173]. In vivo, Smad3 activation induces MAFbx, inhibits mTOR and protein synthesis, and promotes atrophy [174]. Recent muscle cell culture evidence indicates that Smad 3 synergistically increases FoxO-mediated expression of MAFbx and MuRF-1 via DNA binding at promoter regions for these genes [175], suggesting myostatin may function via this mechanism. Regardless, these findings provide yet another example of the crosstalk between the signaling pathways that regulate skeletal muscle mass.

AMPK signaling The AMP-activated protein kinase (AMPK) is a multi-protein kinase composed of a catalytic subunit (α) and two regulatory subunits (βγ) [176]. In response to cellular stress that causes a decrease in cellular energy levels (as sensed by an increase in the AMP:ATP ratio), AMPK is activated and functions to restore the energy status of the cell by turning on catabolic pathways and turning off ATP-consuming anabolic pathways such as protein synthesis [176]. As such, AMPK is called the “energy sensor” of the cell. Chronic activation of AMPK ultimately results in mitochondrial biogenesis [177], making AMPK a primary mediator of endurance exercise adaptations. The central role that

AMPK plays in the regulation of cell metabolism, and protein synthesis in particular, suggest that it could also be involved in regulating skeletal muscle mass.

Inoki and colleagues (2003) provided evidence that AMPK was able to inhibit protein synthesis through phosphorylation of TSC2, a potent inhibitor of TORC1 activity [178]. Various in vivo experiments in rodents subsequently showed that AMPK and mTOR are inversely regulated with hypertrophic stimuli [179-181]. In AMPK knockout mice, muscle mass and fiber size was increased as a result of TORC1 activation [182]. Further, AMPK inactivation accelerated the rate of muscle hypertrophy induced by mechanical overload, thus confirming the idea that AMPK acts to limit skeletal muscle growth [183]. AMPK down-regulation also attenuates atrophy during muscle disuse [184].

In addition to inhibiting TORC1 through TSC2 activation, AMPK has also been shown to restrict muscle growth by inducing expression of FoxO transcription factors and expression of target genes MAFbx and MuRF-1 [185]. In myotubes, activation of AMPK by the AMP analog AICAR (5-Aminoimidazole-4-carboxamide ribonucleotide) caused enhanced myofibrillar protein degradation that was likely the result of FoxO mediated up-regulation of MAFbx and MuRF-1 expression [185]. Accordingly, when AMPK activation is suppressed in the presence of AICAR, MuRF-1 is not up-regulated [186].

The reciprocal relationship between AMPK and mTOR forms the basis of the “molecular interference” phenomenon, which stipulates that optimal hypertrophic adaptation from resistance training (i.e. maximal mTOR activation) cannot be realized in circumstances when endurance training adaptations are occurring simultaneously (i.e. AMPK will interfere with mTOR when training “concurrently”). While chronic and excessive AMPK activation will likely inhibit maximal hypertrophic potential, it is too simplistic to reduce endurance and resistance exercise adaptations to two distinct and competing pathways. For instance, resistance exercise increases AMPK activation [187-192] while aerobic exercise stimulates mTOR [193-195] in humans. In fact, mechanical strain has recently been linked directly to AMPK activation and energy homeostasis [196]. It follows that the evidence for molecular interference inhibiting hypertrophy in humans is quite limited

[197]. There is likely a balance between AMPK and mTOR activation that still allows for maximal hypertrophic adaptation in vivo, but more work is needed to elucidate the limits of this interplay.

NF-κB signaling NF-κB is a dimeric transcription factor involved in a range of biological processes including inflammatory and immune responses, and is rapidly activated by inflammatory cytokines such as TNF-α [198]. In skeletal muscle, however, NF-κB expression was reported to dramatically increase during muscle atrophy that was independent of TNF-α [199]. Subsequent studies showed that NF-κB signaling was in fact necessary and sufficient for mediating the hypertrophic response [200, 201]. The mechanism through which NF-κB signaling promoted atrophy remained unknown until recently when Jackman and colleagues (2012) showed that NF-κB directly regulates the expression of MuRF-1 through a Bcl-3 dependent mechanism [202]. Furthermore, NF-κB sites are required for MuRF-1 activation during disuse atrophy [203]. Alternatively, a member of the TNF family of inflammatory cytokines, TWEAK (TNF-like weak inducer of apoptosis), has been shown to induce skeletal muscle atrophy through up-regulation of MuRF-1 via NF-κB activation [204]. Indeed, high levels of TWEAK promote muscle atrophy in vivo via activation of the “canonical”, or traditional NF-κB signaling pathway [205, 206]. However, through the TWEAK receptor fibroblast growth factor-inducible 14 (Fn14), TWEAK (or potentially some other ligand) can also signal through the “noncanonical” NF-κB pathway which involves synthesis of NF-κB inducible kinase (NIK). Skeletal muscle hypertrophy in humans is associated with Fn14 up-regulation [207, 208] and resistance exercise causes non-canonical NF-κB activation without TWEAK induction [209], suggesting a complex regulation of this pathway that is likely dependent on TWEAK levels and the nature of the stimulus [210].

Hippo pathway and YAP/TAZ signaling The Hippo pathway got its name when a group of researchers noticed that a mutation of one of the key kinase components (Hpo) resulted in tissue overgrowth in the fruit fly,

specifically around the head, that was reminiscent of how a hippopotamus looked [211]. The Hippo pathway has since been recognized as a key mediator of organ size control [212]. In brief, Hippo signaling involves phosphorylation of MST1 and MST2, which acts on the downstream kinases large tumor suppressor kinases 1 and 2 (LATS1 and LATS2), which in turn phosphorylates and represses the transcriptional cofactor Yesassociated protein (YAP) and/or its mammalian paralogue TAZ (transcriptional coactivator with PDZ-binding motif). When YAP/TAZ is de-phosphorylated, it acts as a transcriptional co-activator and affects gene transcription. In various tissues, a high degree of cross-talk is also apparent between the Hippo pathway and defined muscle mass-regulating pathways, such as Akt/mTOR, Myostatin/SMAD, and AMPK, and the Hippo pathway may be activated by a variety of receptors [213-215]. However, the mechanisms of Hippo regulation on muscle fiber size were not explored until recently. YAP was first shown to promote hypertrophy via TEAD transcription factors in skeletal muscle, independent from mTOR activity or MAFbx and MuRF-1 transcription [216]. Indeed, YAP expression increases with a different time course than mTORC1 activation during mechanical overload, and may also regulate muscle mass via ribosome biogenesis and/or altered SMAD2/3 activity [217]. The Hippo pathway and YAP/TAZ regulation are exciting new targets for muscle mass regulation and merit further interrogation.

Summary There have been significant advancements during the last decade in our understanding of the anabolic and catabolic signaling pathways involved in the regulation of skeletal muscle mass. These advances have uncovered the mechanistic details of such signaling pathways and demonstrated the complex crosstalk that occurs between pathways. Future studies will certainly continue to enhance our understanding of the aforementioned pathways as well as better define the importance of emerging pathways not often considered in the field of skeletal muscle plasticity. While there still remains much to be learned, one conclusion that is clear is that the regulation of skeletal muscle mass represents the orchestrated output of multiple anabolic and catabolic signaling pathways.

Figure legend Figure 1. Anabolic and catabolic signaling pathways that regulate skeletal muscle mass. Clenbuterol, IGF-1 and Wnt 7a are able to activate PI3K/AKT signaling through binding to β-AR, IGFR-1 and Fzd7 receptor, respectively. AKT inhibits TSC2 activity which results in Rheb activation of TORC1, localized at the late endosome-lysosome (LEL). TORC1 increases protein synthesis through phosphorylation of S6K1 (p70S6K1) and 4EBP1. AKT is also able to increase protein synthesis through inhibition of GSK3β. Inactivation of GSK3β leads to stabilization of β-catenin resulting in c-Myc induction of ribosome biogenesis. Protein synthesis can be inhibited through AMPK activation of TSC2 or mTOR. AMPK can also increase protein degradation through FoxO regulation of MAFbx and MuRF-1 expression. MAFbx and MuRF-1 are muscle-specific ubiquitin ligases that target proteins for degradation, such as eIF3f and myosin, respectively. FoxO up-regulation of MAFbx and MuRF-1 expression can also be mediated through myostatin binding to the ActIIRβ receptor with the downstream activation of Smad 2 and Smad 3 transcription factors. Canonical activation of NF-κB through an unknown mechanism, that is independent of inflammatory cytokines such as TNF-α, increases MuRF-1 expression through activation of Bcl-3 and facilitates protein degradation. Noncanonical activation of NF-κB via Fn14 may result in muscle hypertrophy. YAP/TAZ is regulated via the Hippo pathway, of which the core components are MST1 and 2 and LATS 1 and 2. Activated YAP/TAZ can promote myogenic gene transcription via TEADs or augment protein synthesis via other mechanisms. Arrow indicates activation where as a crossbar indicates inhibition.

References 1. 2. 3. 4. 5. 6. 7.

8.

9. 10. 11.

12.

13.

14. 15.

16.

17. 18. 19.

Clarys, J.P., et al., Human body composition: A review of adult dissection data. Am J Hum Biol, 1999. 11(2): p. 167-174. Peppa, M., et al., Skeletal muscle insulin resistance in endocrine disease. J Biomed Biotechnol, 2010. 2010: p. 527850. Demontis, F. and N. Perrimon, FOXO/4E-BP signaling in Drosophila muscles regulates organism-wide proteostasis during aging. Cell, 2010. 143(5): p. 813-25. Hass, C.J., M.S. Feigenbaum, and B.A. Franklin, Prescription of resistance training for healthy populations. Sports Med, 2001. 31(14): p. 953-64. Snow, M.H. and B.S. Chortkoff, Frequency of bifurcated muscle fibers in hypertrophic rat soleus muscle. Muscle Nerve, 1987. 10(4): p. 312-7. MacDougall, J.D., et al., Muscle fiber number in biceps brachii in bodybuilders and control subjects. J Appl Physiol, 1984. 57(5): p. 1399-403. Larsson, L. and P.A. Tesch, Motor unit fibre density in extremely hypertrophied skeletal muscles in man. Electrophysiological signs of muscle fibre hyperplasia. Eur J Appl Physiol Occup Physiol, 1986. 55(2): p. 130-6. Giddings, C.J. and W.J. Gonyea, Morphological observations supporting muscle fiber hyperplasia following weight-lifting exercise in cats. Anat Rec, 1992. 233(2): p. 178-95. Kelley, G., Mechanical overload and skeletal muscle fiber hyperplasia: a metaanalysis. J Appl Physiol, 1996. 81(4): p. 1584-8. Eriksson, A., et al., Skeletal muscle morphology in power-lifters with and without anabolic steroids. Histochemistry and cell biology, 2005. 124(2): p. 167-175. Eriksson, A., et al., Hypertrophic muscle fibers with fissures in power-lifters; fiber splitting or defect regeneration? Histochemistry and cell biology, 2006. 126(4): p. 409-417. Kadi, F., et al., Effects of anabolic steroids on the muscle cells of strength-trained athletes. Medicine and science in sports and exercise, 1999. 31(11): p. 15281534. MacDougall, J., et al., Muscle ultrastructural characteristics of elite powerlifters and bodybuilders. European journal of applied physiology and occupational physiology, 1982. 48(1): p. 117-126. Yu, J.-G., et al., Effects of long term supplementation of anabolic androgen steroids on human skeletal muscle. PloS one, 2014. 9(9): p. e105330. Soffe, Z., et al., Effects of loaded voluntary wheel exercise on performance and muscle hypertrophy in young and old male C57Bl/6J mice. Scandinavian journal of medicine & science in sports, 2016. 26(2): p. 172-188. Tamaki, T., et al., Characteristics of compensatory hypertrophied muscle in the rat: I. Electron microscopic and immunohistochemical studies. The Anatomical Record, 1996. 246(3): p. 325-334. Ho, K.W., et al., Skeletal muscle fiber splitting with weight‐ lifting exercise in rats. American Journal of Anatomy, 1980. 157(4): p. 433-440. Goldberg, A.L., Protein synthesis during work-induced growth of skeletal muscle. J Cell Biol, 1968. 36(3): p. 653-8. Chesley, A., et al., Changes in human muscle protein synthesis after resistance exercise. J Appl Physiol, 1992. 73(4): p. 1383-8.

20. 21. 22.

23. 24.

25.

26.

27.

28.

29. 30.

31. 32.

33.

34. 35.

36.

Phillips, S.M., et al., Mixed muscle protein synthesis and breakdown after resistance exercise in humans. Am J Physiol, 1997. 273(1 Pt 1): p. E99-107. Noble, E.G., Q. Tang, and P.B. Taylor, Protein synthesis in compensatory hypertrophy of rat plantaris. Can J Physiol Pharmacol, 1984. 62(9): p. 1178-82. MacDougall, J.D., et al., Changes in muscle protein synthesis following heavy resistance exercise in humans: a pilot study. Acta Physiol Scand, 1992. 146(3): p. 403-4. MacDougall, J.D., et al., The time course for elevated muscle protein synthesis following heavy resistance exercise. Can J Appl Physiol, 1995. 20(4): p. 480-6. Biolo, G., et al., Increased rates of muscle protein turnover and amino acid transport after resistance exercise in humans. Am J Physiol, 1995. 268(3 Pt 1): p. E514-20. West, D.W., et al., Human exercise-mediated skeletal muscle hypertrophy is an intrinsic process. The international journal of biochemistry & cell biology, 2010. 42(9): p. 1371-1375. Mitchell, C.J., et al., Acute post-exercise myofibrillar protein synthesis is not correlated with resistance training-induced muscle hypertrophy in young men. PloS one, 2014. 9(2): p. e89431. Damas, F., et al., Resistance training‐ induced changes in integrated myofibrillar protein synthesis are related to hypertrophy only after attenuation of muscle damage. The Journal of physiology, 2016. 594(18): p. 5209-5222. Baar, K. and K. Esser, Phosphorylation of p70(S6k) correlates with increased skeletal muscle mass following resistance exercise. Am J Physiol, 1999. 276(1 Pt 1): p. C120-7. Aronson, D., et al., Exercise stimulates the mitogen-activated protein kinase pathway in human skeletal muscle. J Clin Invest, 1997. 99(6): p. 1251-7. Sherwood, D.J., et al., Differential regulation of MAP kinase, p70(S6K), and Akt by contraction and insulin in rat skeletal muscle. Am J Physiol, 1999. 276(5 Pt 1): p. E870-8. Hayashi, T., et al., Skeletal muscle contractile activity in vitro stimulates mitogenactivated protein kinase signaling. Am J Physiol, 1999. 277(4 Pt 1): p. C701-7. Bodine, S.C., et al., Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol, 2001. 3(11): p. 1014-9. Mayhew, D.L., et al., Translational signaling responses preceding resistance training-mediated myofiber hypertrophy in young and old humans. J Appl Physiol, 2009. 107(5): p. 1655-62. Saxton, R.A. and D.M. Sabatini, mTOR signaling in growth, metabolism, and disease. Cell, 2017. 168(6): p. 960-976. Chicurel, M.E., et al., Integrin binding and mechanical tension induce movement of mRNA and ribosomes to focal adhesions. Nature, 1998. 392(6677): p. 730733. Crossland, H., et al., Focal adhesion kinase is required for IGF-I-mediated growth of skeletal muscle cells via a TSC2/mTOR/S6K1-associated pathway. Am J Physiol Endocrinol Metab, 2013. 305(2): p. E183-93.

37. 38.

39. 40. 41. 42. 43.

44.

45.

46.

47. 48. 49.

50. 51.

52.

53.

Klossner, S., et al., Mechano-transduction to muscle protein synthesis is modulated by FAK. Eur J Appl Physiol, 2009. 106(3): p. 389-98. Krüger, M. and S. Kötter, Titin, a central mediator for hypertrophic signaling, exercise-induced mechanosignaling and skeletal muscle remodeling. Frontiers in physiology, 2016. 7. Potts, G.K., et al., A map of the phosphoproteomic alterations that occur after a bout of maximal-intensity contractions. J Physiol, 2017. Belgrano, A., et al., Multi-tasking role of the mechanosensing protein Ankrd2 in the signaling network of striated muscle. PLoS One, 2011. 6(10): p. e25519. Frank, D., et al., The sarcomeric Z-disc: a nodal point in signalling and disease. Journal of molecular medicine, 2006. 84(6): p. 446. Palmisano, M.G., et al., Skeletal muscle intermediate filaments form a stresstransmitting and stress-signaling network. J Cell Sci, 2015. 128(2): p. 219-24. Hornberger, T.A., Mechanotransduction and the regulation of mTORC1 signaling in skeletal muscle. The international journal of biochemistry & cell biology, 2011. 43(9): p. 1267-1276. Jacobs, B.L., C.A. Goodman, and T.A. Hornberger, The mechanical activation of mTOR signaling: an emerging role for late endosome/lysosomal targeting. Journal of muscle research and cell motility, 2014. 35(1): p. 11-21. You, J.-S., et al., The role of diacylglycerol kinase ζ and phosphatidic acid in the mechanical activation of mammalian target of rapamycin (mTOR) signaling and skeletal muscle hypertrophy. Journal of Biological Chemistry, 2014. 289(3): p. 1551-1563. Mobley, C.B., et al., Effects of oral phosphatidic acid feeding with or without whey protein on muscle protein synthesis and anabolic signaling in rodent skeletal muscle. Journal of the International Society of Sports Nutrition, 2015. 12(1): p. 32. Joy, J.M., et al., Phosphatidic acid enhances mTOR signaling and resistance exercise induced hypertrophy. Nutrition & metabolism, 2014. 11(1): p. 29. Laplante, M. and D.M. Sabatini, mTOR signaling in growth control and disease. Cell, 2012. 149(2): p. 274-93. Hornberger, T.A., K.B. Sukhija, and S. Chien, Regulation of mTOR by mechanically induced signaling events in skeletal muscle. Cell Cycle, 2006. 5(13): p. 1391-6. Vandenburgh, H.H., et al., Mechanically induced alterations in cultured skeletal muscle growth. J Biomech, 1991. 24 Suppl 1: p. 91-9. Rommel, C., et al., Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol, 2001. 3(11): p. 1009-13. Dan, H.C., et al., Phosphatidylinositol 3-kinase/Akt pathway regulates tuberous sclerosis tumor suppressor complex by phosphorylation of tuberin. J Biol Chem, 2002. 277(38): p. 35364-70. Inoki, K., et al., TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol, 2002. 4(9): p. 648-57.

54.

55. 56. 57. 58.

59. 60. 61.

62.

63. 64.

65. 66.

67. 68.

69. 70.

71.

Manning, B.D., et al., Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol Cell, 2002. 10(1): p. 151-62. Potter, C.J., L.G. Pedraza, and T. Xu, Akt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol, 2002. 4(9): p. 658-65. Garami, A., et al., Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2. Mol Cell, 2003. 11(6): p. 1457-66. Inoki, K., et al., Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev, 2003. 17(15): p. 1829-34. Jacobs, B.L., et al., Eccentric contractions increase the phosphorylation of tuberous sclerosis complex‐ 2 (TSC2) and alter the targeting of TSC2 and the mechanistic target of rapamycin to the lysosome. The Journal of physiology, 2013. 591(18): p. 4611-4620. Sancak, Y., et al., The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science, 2008. 320(5882): p. 1496-1501. Webb, B.L. and C.G. Proud, Eukaryotic initiation factor 2B (eIF2B). Int J Biochem Cell Biol, 1997. 29(10): p. 1127-31. Kubica, N., et al., Resistance exercise increases muscle protein synthesis and translation of eukaryotic initiation factor 2Bepsilon mRNA in a mammalian target of rapamycin-dependent manner. J Biol Chem, 2005. 280(9): p. 7570-80. Lagirand-Cantaloube, J., et al., The initiation factor eIF3-f is a major target for atrogin1/MAFbx function in skeletal muscle atrophy. EMBO J, 2008. 27(8): p. 1266-76. Hinnebusch, A.G., eIF3: a versatile scaffold for translation initiation complexes. Trends Biochem Sci, 2006. 31(10): p. 553-62. Holz, M.K., et al., mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events. Cell, 2005. 123(4): p. 569-80. Harris, T.E., et al., mTOR-dependent stimulation of the association of eIF4G and eIF3 by insulin. EMBO J, 2006. 25(8): p. 1659-68. Csibi, A., et al., The translation regulatory subunit eIF3f controls the kinasedependent mTOR signaling required for muscle differentiation and hypertrophy in mouse. PLoS One, 2010. 5(2): p. e8994. Vyas, D.R., et al., GSK-3beta negatively regulates skeletal myotube hypertrophy. Am J Physiol Cell Physiol, 2002. 283(2): p. C545-51. Satyamoorthy, K., et al., Insulin-like growth factor-1 induces survival and growth of biologically early melanoma cells through both the mitogen-activated protein kinase and beta-catenin pathways. Cancer Res, 2001. 61(19): p. 7318-24. Valenta, T., G. Hausmann, and K. Basler, The many faces and functions of betacatenin. EMBO J, 2012. 31(12): p. 2714-36. Ikeda, S., et al., Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J, 1998. 17(5): p. 1371-84. He, T.C., et al., Identification of c-MYC as a target of the APC pathway. Science, 1998. 281(5382): p. 1509-12.

72. 73.

74.

75.

76.

77. 78.

79.

80.

81. 82. 83.

84.

85.

86.

87.

Shtutman, M., et al., The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci U S A, 1999. 96(10): p. 5522-7. Armstrong, D.D. and K.A. Esser, Wnt/beta-catenin signaling activates growthcontrol genes during overload-induced skeletal muscle hypertrophy. Am J Physiol Cell Physiol, 2005. 289(4): p. C853-9. Armstrong, D.D., V.L. Wong, and K.A. Esser, Expression of beta-catenin is necessary for physiological growth of adult skeletal muscle. Am J Physiol Cell Physiol, 2006. 291(1): p. C185-8. Ecker, R.E. and G. Kokaisl, Synthesis of protein, ribonucleic acid, and ribosomes by individual bacterial cells in balanced growth. J Bacteriol, 1969. 98(3): p. 121926. Miyazaki, M. and K.A. Esser, Cellular mechanisms regulating protein synthesis and skeletal muscle hypertrophy in animals. J Appl Physiol, 2009. 106(4): p. 1367-73. Hannan, R.D., et al., Cardiac hypertrophy: a matter of translation. Clin Exp Pharmacol Physiol, 2003. 30(8): p. 517-27. von Walden, F., et al., Mechanical loading induces the expression of a Pol I regulon at the onset of skeletal muscle hypertrophy. Am J Physiol Cell Physiol, 2012. 302(10): p. C1523-30. Nader, G.A., T.J. McLoughlin, and K.A. Esser, mTOR function in skeletal muscle hypertrophy: increased ribosomal RNA via cell cycle regulators. Am J Physiol Cell Physiol, 2005. 289(6): p. C1457-65. Mayer, C. and I. Grummt, Ribosome biogenesis and cell growth: mTOR coordinates transcription by all three classes of nuclear RNA polymerases. Oncogene, 2006. 25(48): p. 6384-6391. Kusnadi, E.P., et al., Regulation of rDNA transcription in response to growth factors, nutrients and energy. Gene, 2015. 556(1): p. 27-34. Sanij, E., et al., UBF levels determine the number of active ribosomal RNA genes in mammals. J Cell Biol, 2008. 183(7): p. 1259-1274. Nader, G.A., T.J. McLoughlin, and K.A. Esser, mTOR function in skeletal muscle hypertrophy: increased ribosomal RNA via cell cycle regulators. American Journal of Physiology-Cell Physiology, 2005. 289(6): p. C1457-C1465. Stec, M.J., et al., Ribosome biogenesis may augment resistance training-induced myofiber hypertrophy and is required for myotube growth in vitro. American Journal of Physiology-Endocrinology and Metabolism, 2016. 310(8): p. E652E661. von Walden, F., et al., mTOR signaling regulates myotube hypertrophy by modulating protein synthesis, rDNA transcription, and chromatin remodeling. American Journal of Physiology-Cell Physiology, 2016. 311(4): p. C663-C672. von Walden, F., et al., Mechanical loading induces the expression of a Pol I regulon at the onset of skeletal muscle hypertrophy. American Journal of Physiology-Cell Physiology, 2012. 302(10): p. C1523-C1530. Goodman, C.A., et al., The role of skeletal muscle mTOR in the regulation of mechanical load‐ induced growth. The Journal of physiology, 2011. 589(22): p. 5485-5501.

88.

89.

90.

91.

92.

93.

94.

95. 96. 97. 98.

99.

100.

101.

102.

West, D.W., et al., Acute resistance exercise activates rapamycin‐ sensitive and‐ insensitive mechanisms that control translational activity and capacity in skeletal muscle. The Journal of physiology, 2016. 594(2): p. 453-468. Figueiredo, V.C., et al., Ribosome biogenesis adaptation in resistance traininginduced human skeletal muscle hypertrophy. American Journal of PhysiologyEndocrinology And Metabolism, 2015. 309(1): p. E72-E83. Figueiredo, V.C., et al., Impact of resistance exercise on ribosome biogenesis is acutely regulated by post‐ exercise recovery strategies. Physiological reports, 2016. 4(2): p. e12670. Wen, Y., A.P. Alimov, and J.J. McCarthy, Ribosome Biogenesis is Necessary for Skeletal Muscle Hypertrophy. Exercise and sport sciences reviews, 2016. 44(3): p. 110-115. Kirby, T.J., et al., Blunted hypertrophic response in aged skeletal muscle is associated with decreased ribosome biogenesis. Journal of Applied Physiology, 2015. 119(4): p. 321-327. Stec, M.J., D.L. Mayhew, and M.M. Bamman, The effects of age and resistance loading on skeletal muscle ribosome biogenesis. Journal of Applied Physiology, 2015. 119(8): p. 851-857. von Maltzahn, J., C.F. Bentzinger, and M.A. Rudnicki, Wnt7a-Fzd7 signalling directly activates the Akt/mTOR anabolic growth pathway in skeletal muscle. Nat Cell Biol, 2011. 14(2): p. 186-91. Le Grand, F., et al., Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. Cell Stem Cell, 2009. 4(6): p. 535-47. Kim, Y.S., et al., Characterization of beta 1- and beta 2-adrenoceptors in rat skeletal muscles. Biochem Pharmacol, 1991. 42(9): p. 1783-9. Rattigan, S., et al., Alpha-adrenergic receptors in rat skeletal muscle. Biochem Biophys Res Commun, 1986. 136(3): p. 1071-7. Williams, R.S., M.G. Caron, and K. Daniel, Skeletal muscle beta-adrenergic receptors: variations due to fiber type and training. Am J Physiol, 1984. 246(2 Pt 1): p. E160-7. Gosmanov, A.R., J.A. Wong, and D.B. Thomason, Duality of G protein-coupled mechanisms for beta-adrenergic activation of NKCC activity in skeletal muscle. Am J Physiol Cell Physiol, 2002. 283(4): p. C1025-32. Yamamoto, D.L., D.S. Hutchinson, and T. Bengtsson, Beta(2)-Adrenergic activation increases glycogen synthesis in L6 skeletal muscle cells through a signalling pathway independent of cyclic AMP. Diabetologia, 2007. 50(1): p. 15867. Lynch, G.S. and J.G. Ryall, Role of beta-adrenoceptor signaling in skeletal muscle: implications for muscle wasting and disease. Physiol Rev, 2008. 88(2): p. 729-67. Murga, C., et al., Activation of Akt/protein kinase B by G protein-coupled receptors. A role for alpha and beta gamma subunits of heterotrimeric G proteins acting through phosphatidylinositol-3-OH kinasegamma. J Biol Chem, 1998. 273(30): p. 19080-5.

103.

104.

105. 106.

107. 108.

109.

110.

111. 112. 113.

114.

115. 116.

117.

118.

Murga, C., S. Fukuhara, and J.S. Gutkind, A novel role for phosphatidylinositol 3kinase beta in signaling from G protein-coupled receptors to Akt. J Biol Chem, 2000. 275(16): p. 12069-73. Schmidt, P., F. Holsboer, and D. Spengler, Beta(2)-adrenergic receptors potentiate glucocorticoid receptor transactivation via G protein beta gammasubunits and the phosphoinositide 3-kinase pathway. Mol Endocrinol, 2001. 15(4): p. 553-64. Emery, P.W., et al., Chronic effects of beta 2-adrenergic agonists on body composition and protein synthesis in the rat. Biosci Rep, 1984. 4(1): p. 83-91. Maltin, C.A., M.I. Delday, and P.J. Reeds, The effect of a growth promoting drug, clenbuterol, on fibre frequency and area in hind limb muscles from young male rats. Biosci Rep, 1986. 6(3): p. 293-9. Maltin, C.A. and M.I. Delday, Satellite cells in innervated and denervated muscles treated with clenbuterol. Muscle Nerve, 1992. 15(8): p. 919-25. Roberts, P. and J.K. McGeachie, The effects of clenbuterol on satellite cell activation and the regeneration of skeletal muscle: an autoradiographic and morphometric study of whole muscle transplants in mice. J Anat, 1992. 180 ( Pt 1): p. 57-65. Maltin, C.A., et al., Tissue specific responses to clenbuterol; temporal changes in protein metabolism of striated muscle and visceral tissues from rats. Growth Regul, 1992. 2(4): p. 161-6. Claeys, M.C., et al., Skeletal muscle protein synthesis and growth hormone secretion in young lambs treated with clenbuterol. J Anim Sci, 1989. 67(9): p. 2245-54. Reeds, P.J., et al., Stimulation of muscle growth by clenbuterol: lack of effect on muscle protein biosynthesis. Br J Nutr, 1986. 56(1): p. 249-58. Benson, D.W., et al., Decreased myofibrillar protein breakdown following treatment with clenbuterol. J Surg Res, 1991. 50(1): p. 1-5. Forsberg, N.E., et al., Effects of cimaterol on rabbit growth and myofibrillar protein degradation and on calcium-dependent proteinase and calpastatin activities in skeletal muscle. J Anim Sci, 1989. 67(12): p. 3313-21. Hesketh, J.E., et al., Stimulation of actin and myosin synthesis in rat gastrocnemius muscle by clenbuterol; evidence for translational control. Comp Biochem Physiol C, 1992. 102(1): p. 23-7. Kline, W.O., et al., Rapamycin inhibits the growth and muscle-sparing effects of clenbuterol. J Appl Physiol, 2007. 102(2): p. 740-7. Goncalves, D.A., et al., Clenbuterol suppresses proteasomal and lysosomal proteolysis and atrophy-related genes in denervated rat soleus muscles independently of Akt. Am J Physiol Endocrinol Metab, 2012. 302(1): p. E123-33. Woodall, B.P., et al., Skeletal Muscle-specific G Protein-coupled Receptor Kinase 2 Ablation Alters Isolated Skeletal Muscle Mechanics and Enhances Clenbuterol-stimulated Hypertrophy. J Biol Chem, 2016. 291(42): p. 2191321924. Py, G., et al., Chronic clenbuterol treatment compromises force production without directly altering skeletal muscle contractile machinery. The Journal of physiology, 2015. 593(8): p. 2071-2084.

119.

120.

121.

122.

123.

124.

125. 126.

127. 128. 129. 130.

131. 132.

133.

134.

135.

Sirvent, P., et al., Effects of chronic administration of clenbuterol on contractile properties and calcium homeostasis in rat extensor digitorum longus muscle. PloS one, 2014. 9(6): p. e100281. Minetti, G.C., et al., Galphai2 signaling promotes skeletal muscle hypertrophy, myoblast differentiation, and muscle regeneration. Sci Signal, 2011. 4(201): p. ra80. Hagg, A., et al., Using AAV vectors expressing the β2-adrenoceptor or associated Gα proteins to modulate skeletal muscle mass and muscle fibre size. Scientific reports, 2016. 6. Sellman, J.E., et al., In vivo inhibition of nitric oxide synthase impairs upregulation of contractile protein mRNA in overloaded plantaris muscle. J Appl Physiol, 2006. 100(1): p. 258-65. Smith, L.W., J.D. Smith, and D.S. Criswell, Involvement of nitric oxide synthase in skeletal muscle adaptation to chronic overload. J Appl Physiol, 2002. 92(5): p. 2005-11. Ito, N., et al., Activation of calcium signaling through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hypertrophy. Nat Med, 2013. 19(1): p. 101-6. Gulati, P., et al., Amino acids activate mTOR complex 1 via Ca2+/CaM signaling to hVps34. Cell Metab, 2008. 7(5): p. 456-65. De Palma, C., et al., Deficient nitric oxide signalling impairs skeletal muscle growth and performance: involvement of mitochondrial dysregulation. Skelet Muscle, 2014. 4(1): p. 22. Makanae, Y., et al., Vitamin C administration attenuates overload‐ induced skeletal muscle hypertrophy in rats. Acta Physiologica, 2013. 208(1): p. 57-65. Puigserver, P., et al., A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell, 1998. 92(6): p. 829-39. Olesen, J., K. Kiilerich, and H. Pilegaard, PGC-1alpha-mediated adaptations in skeletal muscle. Pflugers Arch, 2010. 460(1): p. 153-62. Sandri, M., et al., PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc Natl Acad Sci U S A, 2006. 103(44): p. 16260-5. Ruas, J.L., et al., A PGC-1alpha isoform induced by resistance training regulates skeletal muscle hypertrophy. Cell, 2012. 151(6): p. 1319-31. White, J.P., et al., G protein-coupled receptor 56 regulates mechanical overloadinduced muscle hypertrophy. Proceedings of the National Academy of Sciences, 2014. 111(44): p. 15756-15761. Lundberg, T.R., et al., Truncated splice variant PGC‐ 1α4 is not associated with exercise‐ induced human muscle hypertrophy. Acta Physiologica, 2014. 212(2): p. 142-151. Ydfors, M., et al., The truncated splice variants, NT‐ PGC‐ 1α and PGC‐ 1α4, increase with both endurance and resistance exercise in human skeletal muscle. Physiological reports, 2013. 1(6): p. e00140. Thom, R., et al., Hypoxic induction of vascular endothelial growth factor (VEGF) and angiogenesis in muscle by truncated peroxisome proliferator-activated

136. 137. 138.

139. 140.

141.

142. 143. 144. 145.

146. 147. 148.

149. 150. 151. 152. 153.

154.

receptor γ coactivator (PGC)-1α. Journal of Biological Chemistry, 2014. 289(13): p. 8810-8817. Pasquinelli, A.E., MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship. Nat Rev Genet, 2012. 13(4): p. 271-82. Williams, A.H., et al., MicroRNA control of muscle development and disease. Curr Opin Cell Biol, 2009. 21(3): p. 461-9. McCarthy, J.J. and K.A. Esser, MicroRNA-1 and microRNA-133a expression are decreased during skeletal muscle hypertrophy. Journal of applied physiology, 2007. 102(1): p. 306-313. McCarthy, J.J., The MyomiR network in skeletal muscle plasticity. Exerc Sport Sci Rev, 2011. 39(3): p. 150-4. Davidsen, P.K., et al., High responders to resistance exercise training demonstrate differential regulation of skeletal muscle microRNA expression. J Appl Physiol, 2011. 110(2): p. 309-17. Guescini, M., et al., Muscle releases alpha-sarcoglycan positive extracellular vesicles carrying miRNAs in the bloodstream. PloS one, 2015. 10(5): p. e0125094. Russell, A.P. and S. Lamon, Exercise, Skeletal Muscle and Circulating microRNAs. Prog Mol Biol Transl Sci, 2015. 135: p. 471-96. Fry, C.S., et al., Satellite Cells Regulate Extracellular Matrix Remodelling During Skeletal Muscle Adaptation. Cell Stem Cell, 2016. In Review - 2nd Round. Fry, C.S., et al., Regulation of the muscle fiber microenvironment by activated satellite cells during hypertrophy. FASEB J, 2014. 28(4): p. 1654-65. Hudson, M.B., et al., miR-23a is decreased during muscle atrophy by a mechanism that includes calcineurin signaling and exosome-mediated export. American Journal of Physiology-Cell Physiology, 2014. 306(6): p. C551-C558. Bodine, S.C., et al., Identification of ubiquitin ligases required for skeletal muscle atrophy. Science, 2001. 294(5547): p. 1704-8. Gomes, M.D., et al., Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proc Natl Acad Sci U S A, 2001. 98(25): p. 14440-5. Baehr, L.M., M. Tunzi, and S.C. Bodine, Muscle hypertrophy is associated with increases in proteasome activity that is independent of MuRF1 and MAFbx expression. Front Physiol, 2014. 5: p. 69. Sartori, R., et al., BMP signaling controls muscle mass. Nature genetics, 2013. 45(11): p. 1309-1318. D'Cruz, R., et al., PDLIM7 is a novel target of the ubiquitin ligase Nedd4-1 in skeletal muscle. Biochemical Journal, 2016. 473(3): p. 267-276. Nagpal, P., et al., The ubiquitin ligase Nedd4-1 participates in denervationinduced skeletal muscle atrophy in mice. PLoS One, 2012. 7(10): p. e46427. Bell, R.A., M. Al-Khalaf, and L.A. Megeney, The beneficial role of proteolysis in skeletal muscle growth and stress adaptation. Skelet Muscle, 2016. 6: p. 16. Baehr, L.M., M. Tunzi, and S.C. Bodine, Muscle hypertrophy is associated with increases in proteasome activity that is independent of MuRF1 and MAFbx expression. Frontiers in physiology, 2014. 5: p. 69. Kitajima, Y., et al., Proteasome dysfunction induces muscle growth defects and protein aggregation. J Cell Sci, 2014. 127(24): p. 5204-5217.

155.

156.

157. 158. 159.

160.

161.

162. 163.

164. 165.

166.

167. 168.

169. 170.

171.

Murton, A., D. Constantin, and P. Greenhaff, The involvement of the ubiquitin proteasome system in human skeletal muscle remodelling and atrophy. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 2008. 1782(12): p. 730-743. Sacheck, J.M., et al., IGF-I stimulates muscle growth by suppressing protein breakdown and expression of atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. Am J Physiol Endocrinol Metab, 2004. 287(4): p. E591-601. Tang, E.D., et al., Negative regulation of the forkhead transcription factor FKHR by Akt. J Biol Chem, 1999. 274(24): p. 16741-6. Brunet, A., et al., Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell, 1999. 96(6): p. 857-68. Sandri, M., et al., Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell, 2004. 117(3): p. 399412. Stitt, T.N., et al., The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell, 2004. 14(3): p. 395-403. Southgate, R.J., et al., FOXO1 regulates the expression of 4E-BP1 and inhibits mTOR signaling in mammalian skeletal muscle. J Biol Chem, 2007. 282(29): p. 21176-86. Mammucari, C., et al., FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab, 2007. 6(6): p. 458-71. Zhao, J., et al., FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell metabolism, 2007. 6(6): p. 472-483. Masiero, E., et al., Autophagy is required to maintain muscle mass. Cell Metab, 2009. 10(6): p. 507-15. Cohen, S., et al., During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation. J Cell Biol, 2009. 185(6): p. 1083-95. Lagirand-Cantaloube, J., et al., Inhibition of atrogin-1/MAFbx mediated MyoD proteolysis prevents skeletal muscle atrophy in vivo. PLoS One, 2009. 4(3): p. e4973. Foletta, V.C., et al., The role and regulation of MAFbx/atrogin-1 and MuRF1 in skeletal muscle atrophy. Pflugers Arch, 2011. 461(3): p. 325-35. McPherron, A.C., A.M. Lawler, and S.J. Lee, Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature, 1997. 387(6628): p. 8390. Lee, S.J., Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol, 2004. 20: p. 61-86. Welle, S., K. Burgess, and S. Mehta, Stimulation of skeletal muscle myofibrillar protein synthesis, p70 S6 kinase phosphorylation, and ribosomal protein S6 phosphorylation by inhibition of myostatin in mature mice. Am J Physiol Endocrinol Metab, 2009. 296(3): p. E567-72. Zimmers, T.A., et al., Induction of cachexia in mice by systemically administered myostatin. Science, 2002. 296(5572): p. 1486-8.

172. 173.

174.

175.

176. 177.

178. 179.

180.

181.

182. 183. 184.

185.

186.

187. 188.

Sartori, R., et al., Smad2 and 3 transcription factors control muscle mass in adulthood. Am J Physiol Cell Physiol, 2009. 296(6): p. C1248-57. Trendelenburg, A.U., et al., Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Physiol Cell Physiol, 2009. 296(6): p. C1258-70. Goodman, C.A., et al., Smad3 induces atrogin-1, inhibits mTOR and protein synthesis, and promotes muscle atrophy in vivo. Molecular Endocrinology, 2013. 27(11): p. 1946-1957. Bollinger, L.M., et al., SMAD3 augments FoxO3-induced MuRF-1 promoter activity in a DNA-binding-dependent manner. American Journal of PhysiologyCell Physiology, 2014. 307(3): p. C278-C287. Steinberg, G.R. and B.E. Kemp, AMPK in Health and Disease. Physiol Rev, 2009. 89(3): p. 1025-78. Zong, H., et al., AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proceedings of the National Academy of Sciences, 2002. 99(25): p. 15983-15987. Inoki, K., T. Zhu, and K.L. Guan, TSC2 mediates cellular energy response to control cell growth and survival. Cell, 2003. 115(5): p. 577-90. Atherton, P.J., et al., Selective activation of AMPK-PGC-1alpha or PKB-TSC2mTOR signaling can explain specific adaptive responses to endurance or resistance training-like electrical muscle stimulation. FASEB J, 2005. 19(7): p. 786-8. Nader, G.A. and K.A. Esser, Intracellular signaling specificity in skeletal muscle in response to different modes of exercise. J Appl Physiol (1985), 2001. 90(5): p. 1936-42. Thomson, D.M., C.A. Fick, and S.E. Gordon, AMPK activation attenuates S6K1, 4E-BP1, and eEF2 signaling responses to high-frequency electrically stimulated skeletal muscle contractions. J Appl Physiol (1985), 2008. 104(3): p. 625-32. Lantier, L., et al., Coordinated maintenance of muscle cell size control by AMPactivated protein kinase. FASEB J, 2010. 24(9): p. 3555-61. Mounier, R., et al., Important role for AMPKalpha1 in limiting skeletal muscle cell hypertrophy. FASEB J, 2009. 23(7): p. 2264-73. Egawa, T., et al., Involvement of AMPK in regulating slow-twitch muscle atrophy during hindlimb unloading in mice. American Journal of PhysiologyEndocrinology and Metabolism, 2015. 309(7): p. E651-E662. Nakashima, K. and Y. Yakabe, AMPK activation stimulates myofibrillar protein degradation and expression of atrophy-related ubiquitin ligases by increasing FOXO transcription factors in C2C12 myotubes. Biosci Biotechnol Biochem, 2007. 71(7): p. 1650-6. Egawa, T., et al., AICAR-induced activation of AMPK negatively regulates myotube hypertrophy through the HSP72-mediated pathway in C2C12 skeletal muscle cells. Am J Physiol Endocrinol Metab, 2014. 306(3): p. E344-54. Pugh, J.K., et al., Acute molecular responses to concurrent resistance and highintensity interval exercise in untrained skeletal muscle. Physiol Rep, 2015. 3(4). Coffey, V.G., et al., Early signaling responses to divergent exercise stimuli in skeletal muscle from well-trained humans. FASEB J, 2006. 20(1): p. 190-2.

189.

190.

191.

192.

193.

194.

195.

196. 197.

198. 199. 200. 201.

202.

203.

204.

Dreyer, H.C., et al., Resistance exercise increases AMPK activity and reduces 4E-BP1 phosphorylation and protein synthesis in human skeletal muscle. J Physiol, 2006. 576(Pt 2): p. 613-24. Koopman, R., et al., Increase in S6K1 phosphorylation in human skeletal muscle following resistance exercise occurs mainly in type II muscle fibers. Am J Physiol Endocrinol Metab, 2006. 290(6): p. E1245-52. Vissing, K., et al., Differentiated mTOR but not AMPK signaling after strength vs endurance exercise in training-accustomed individuals. Scand J Med Sci Sports, 2013. 23(3): p. 355-66. Wilkinson, S.B., et al., Differential effects of resistance and endurance exercise in the fed state on signalling molecule phosphorylation and protein synthesis in human muscle. J Physiol, 2008. 586(Pt 15): p. 3701-17. Benziane, B., et al., Divergent cell signaling after short-term intensified endurance training in human skeletal muscle. Am J Physiol Endocrinol Metab, 2008. 295(6): p. E1427-38. Mascher, H., et al., Changes in signalling pathways regulating protein synthesis in human muscle in the recovery period after endurance exercise. Acta Physiol (Oxf), 2007. 191(1): p. 67-75. Mascher, H., et al., Enhanced rates of muscle protein synthesis and elevated mTOR signalling following endurance exercise in human subjects. Acta Physiol (Oxf), 2011. 202(2): p. 175-84. Bays, J.L., et al., Linking E-cadherin mechanotransduction to cell metabolism through force-mediated activation of AMPK. Nat Cell Biol, 2017. Murach, K.A. and J.R. Bagley, Skeletal Muscle Hypertrophy with Concurrent Exercise Training: Contrary Evidence for an Interference Effect. Sports Med, 2016. 46(8): p. 1029-39. Hayden, M.S. and S. Ghosh, NF-kappaB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev, 2012. 26(3): p. 203-34. Hunter, R.B., et al., Activation of an alternative NF-kappaB pathway in skeletal muscle during disuse atrophy. FASEB J, 2002. 16(6): p. 529-38. Hunter, R.B. and S.C. Kandarian, Disruption of either the Nfkb1 or the Bcl3 gene inhibits skeletal muscle atrophy. J Clin Invest, 2004. 114(10): p. 1504-11. Van Gammeren, D., et al., The IkappaB kinases IKKalpha and IKKbeta are necessary and sufficient for skeletal muscle atrophy. FASEB J, 2009. 23(2): p. 362-70. Jackman, R.W., C.L. Wu, and S.C. Kandarian, The ChIP-seq-Defined Networks of Bcl-3 Gene Binding Support Its Required Role in Skeletal Muscle Atrophy. PLoS One, 2013. 7(12): p. e51478. Wu, C.-L., et al., NF-κB but not FoxO sites in the MuRF1 promoter are required for transcriptional activation in disuse muscle atrophy. American Journal of Physiology-Cell Physiology, 2014. 306(8): p. C762-C767. Bhatnagar, S., et al., TWEAK causes myotube atrophy through coordinated activation of ubiquitin-proteasome system, autophagy, and caspases. J Cell Physiol, 2012. 227(3): p. 1042-51.

205.

206.

207.

208. 209.

210.

211. 212. 213. 214. 215.

216. 217.

Dogra, C., et al., TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal muscle-wasting cytokine. The FASEB Journal, 2007. 21(8): p. 18571869. Mittal, A., et al., The TWEAK–Fn14 system is a critical regulator of denervationinduced skeletal muscle atrophy in mice. The Journal of cell biology, 2010. 188(6): p. 833-849. Raue, U., et al., Transcriptome signature of resistance exercise adaptations: mixed muscle and fiber type specific profiles in young and old adults. Journal of applied physiology, 2012. 112(10): p. 1625-1636. Murach, K., et al., Single muscle fiber gene expression with run taper. PloS one, 2014. 9(9): p. e108547. Raue, U., et al., TWEAK-Fn14 pathway activation after exercise in human skeletal muscle: insights from two exercise modes and a time course investigation. Journal of Applied Physiology, 2015. 118(5): p. 569-578. Merritt, E.K., et al., Heightened muscle inflammation susceptibility may impair regenerative capacity in aging humans. Journal of applied physiology, 2013. 115(6): p. 937-948. Udan, R.S., et al., Hippo promotes proliferation arrest and apoptosis in the Salvador/Warts pathway. Nature cell biology, 2003. 5(10): p. 914-920. Pan, D., Hippo signaling in organ size control. Genes & development, 2007. 21(8): p. 886-897. Gabriel, B.M., et al., The Hippo signal transduction network for exercise physiologists. J Appl Physiol (1985), 2016. 120(10): p. 1105-17. Fischer, M., et al., YAP-Mediated Mechanotransduction in Skeletal Muscle. Frontiers in physiology, 2016. 7. Gnimassou, O., M. Francaux, and L. Deldicque, Hippo Pathway and Skeletal Muscle Mass Regulation in Mammals: A Controversial Relationship. Frontiers in Physiology, 2017. 8. Watt, K.I., et al., The Hippo pathway effector YAP is a critical regulator of skeletal muscle fibre size. Nat Commun, 2015. 6: p. 6048. Goodman, C.A., et al., Yes-Associated Protein is up-regulated by mechanical overload and is sufficient to induce skeletal muscle hypertrophy. FEBS Lett, 2015. 589(13): p. 1491-7.

clenbuterol

IGF-1

β-AR

IGFR

Wnt7a

myostatin

Fzd7

sarcolemma

?

ActIIRβ

DGK

Fn14

PA

PI3K

canonical NF-𝜅B

AKT

Smad 2/3

GSK3β

Bcl-3

non-canonical

NIK

︎ AMP:ATP ratio

TSC2 β-catenin Rheb

AMPK

mTOR

FoxO

LEL

protein synthesis and/or ➡︎ protein degradation?

c-Myc

ribosome biogenesis

MST 1/2 Myonucleus

eIF3F S6K1

4E-BP1

LATS 1/2 YAP/TAZ

MuRF-1

MAFbx

TEADs

YAP/TAZ ︎ translational capacity

︎ translational efficiency protein synthesis

myogenic gene transcription? protein degradation