6
J Phys Fitness Sports Med, 2(4): 423-428 (2013) DOI: 10.7600/jpfsm.2.423 JPFSM: Review Article How β2-adrenergic agonists induce skeletal muscle hypertrophy? Takashi Kitaura Received: September 2, 2013 / Accepted: September 10, 2013 Abstract Some β 2 -adrenergic agonists (β 2 -agonists) can strongly induce muscular hypertrophy, and are prohibited to use as doping drugs for athletes. The pharmacological mechanism for such induced hypertrophy is not clear. These agonists affect many organs via the cAMP-PKA system. Muscular hypertrophy is most likely induced by way of protein synthesis via the IGF-1/PI3K/ PKB system and negative myostatin pathway. There are some reports indicating that β 2 -agonists might stimulate protein synthesis via the IGF-1/PI3K/PKB system. Furthermore, it inhibits proteolysis via the ATP-dependent ubiquitin-proteasome system (UPS), autophagy-lysosome system and calcium-calpain system. In this review, some discrepancies are introduced between a basic hypertrophic mechanism and inhibited atrophic mechanism using β 2 -agonists. Surfaces of muscle fiber might have some various receptors, and the β 2 -adrenoceptor might be activated di- rectly by PI3K via s to trigger skeletal muscle hypertrophy. β 2 -agonists might be stimulated to synthesize follistatin of the myostatin inhibitor, as well as to produce satellite cells. Furthermore, increased myostatin may work to determine the size of each muscle fiber, and an increased number of quiescent satellite cells serve as myonuclei donors for hypertrophied muscle fibers. Recently, it was reported that follistatin synthesis was regulated by microRNAs. The effect of microRNA on β 2 -agonists is not clear. In the cAMP/PKA/CREB pathway, β 2 -agonists might inhibit various proteolytic systems, resulting in an increase of structural proteins. But β 2 -agonists have more pharmacological functions like lipolytic action and stimulation of the central nervous system; thus, more research and analysis is needed. Keywords : β 2 -adrenergic agonists, muscle hypertrophy, clenbuterol, protein synthesis, protein degradation Introduction Muscular hypertrophy is one of the most interesting phenomena for athletes and people with muscle-wasting disorders. There are many methods to induce muscular hypertrophy like anaerobic weight training and hormonal treatment. Some β2-adrenergic agonists (β2-agonists) like clenbuterol, fenoterol, formoterol and salbutamol are well known as bronchodilators to treat asthma, and act as decongestants or as powerful anabolic agents. Therefore, they are limited in use for doping in athletic competitions. In particular, the anabolic action is the most intrinsic property in sports. Over the past 30 years many investiga- tors have reported about skeletal and/or cardiac muscle hypertrophy with clenbuterol administration 1-5) . But the mechanism of the action of drugs is complicated, and still under discussion in some reviews 6-9) . In this review, I propose a way to clarify the problem of the mechanism. The basic idea to explain muscular hypertrophy is due to the increased structural proteins. It was caused by the increased protein synthesis and the decreased proteolysis. The homeostatic protein content is maintained by the balance between synthesis and break- down. The grade of change is summarized into hypertro- phic grades (H1-H4), atrophic grades (A1-A4) or keeping the situation (K) with no change (Table 1). If both the increasing grade and the degradation grade are the same high level (I5 and D5), protein content shows no change (K). Therefore, we need a few analyses to determine atro- phy or hypertrophy in two different directions. But most research was done in one direction for the various reasons of time, technique, and so on. In many cases, increasing grades (I1-I5) depend on the activity of the IGF-1/PI3K/Akt signaling pathway 10-12) and negative myostatin pathway for protein synthesis 13-15) . On the other hand, degradation grades depend on the activ- ity of the ATP dependent ubiquitin-proteosome system (UPS) 16,17) , autophagy-lysosome system 18-20) and calcium sensitive calpain pathway 21-23) . They are well documented by Ito et al. 24) . Increments of structural proteins There are some reports that β2-agonists increase protein Correspondence: [email protected] Laboratory of Exercise Biochemistry, Division of Sports Education, Health Service Center, Kanazawa University, Kakuma, Kanazawa, Ishikawa 920-1192, Japan

How β2-adrenergic agonists induce skeletal muscle hypertrophy?

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: How β2-adrenergic agonists induce skeletal muscle hypertrophy?

J Phys Fitness Sports Med, 2(4): 423-428 (2013)DOI: 10.7600/jpfsm.2.423

JPFSM: Review Article

How β2-adrenergic agonists induce skeletal muscle hypertrophy?Takashi Kitaura

Received: September 2, 2013 / Accepted: September 10, 2013

Abstract Some β2-adrenergic agonists (β2-agonists) can strongly induce muscular hypertrophy, and are prohibited to use as doping drugs for athletes. The pharmacological mechanism for such induced hypertrophy is not clear. These agonists affect many organs via the cAMP-PKA system. Muscular hypertrophy is most likely induced by way of protein synthesis via the IGF-1/PI3K/PKB system and negative myostatin pathway. There are some reports indicating that β2-agonists might stimulate protein synthesis via the IGF-1/PI3K/PKB system. Furthermore, it inhibits proteolysis via the ATP-dependent ubiquitin-proteasome system (UPS), autophagy-lysosome system and calcium-calpain system. In this review, some discrepancies are introduced between a basic hypertrophic mechanism and inhibited atrophic mechanism using β2-agonists. Surfaces of muscle fiber might have some various receptors, and the β2-adrenoceptor might be activated di-rectly by PI3K via Gαs to trigger skeletal muscle hypertrophy. β2-agonists might be stimulated to synthesize follistatin of the myostatin inhibitor, as well as to produce satellite cells. Furthermore, increased myostatin may work to determine the size of each muscle fiber, and an increased number of quiescent satellite cells serve as myonuclei donors for hypertrophied muscle fibers. Recently, it was reported that follistatin synthesis was regulated by microRNAs. The effect of microRNA on β2-agonists is not clear. In the cAMP/PKA/CREB pathway, β2-agonists might inhibit various proteolytic systems, resulting in an increase of structural proteins. But β2-agonists have more pharmacological functions like lipolytic action and stimulation of the central nervous system; thus, more research and analysis is needed.Keywords : β2-adrenergic agonists, muscle hypertrophy, clenbuterol, protein synthesis, protein

degradation

Introduction

Muscular hypertrophy is one of the most interesting phenomena for athletes and people with muscle-wasting disorders. There are many methods to induce muscular hypertrophy like anaerobic weight training and hormonal treatment. Some β2-adrenergic agonists (β2-agonists) like clenbuterol, fenoterol, formoterol and salbutamol are well known as bronchodilators to treat asthma, and act as decongestants or as powerful anabolic agents. Therefore, they are limited in use for doping in athletic competitions. In particular, the anabolic action is the most intrinsic property in sports. Over the past 30 years many investiga-tors have reported about skeletal and/or cardiac muscle hypertrophy with clenbuterol administration1-5). But the mechanism of the action of drugs is complicated, and still under discussion in some reviews6-9). In this review, I propose a way to clarify the problem of the mechanism. The basic idea to explain muscular hypertrophy is due to the increased structural proteins. It was caused by the increased protein synthesis and the

decreased proteolysis. The homeostatic protein content is maintained by the balance between synthesis and break-down. The grade of change is summarized into hypertro-phic grades (H1-H4), atrophic grades (A1-A4) or keeping the situation (K) with no change (Table 1). If both the increasing grade and the degradation grade are the same high level (I5 and D5), protein content shows no change (K). Therefore, we need a few analyses to determine atro-phy or hypertrophy in two different directions. But most research was done in one direction for the various reasons of time, technique, and so on. In many cases, increasing grades (I1-I5) depend on the activity of the IGF-1/PI3K/Akt signaling pathway10-12) and negative myostatin pathway for protein synthesis13-15). On the other hand, degradation grades depend on the activ-ity of the ATP dependent ubiquitin-proteosome system (UPS)16,17), autophagy-lysosome system18-20) and calcium sensitive calpain pathway21-23). They are well documented by Ito et al.24).

Increments of structural proteins

There are some reports that β2-agonists increase protein Correspondence: [email protected]

Laboratory of Exercise Biochemistry, Division of Sports Education, Health Service Center, Kanazawa University, Kakuma, Kanazawa, Ishikawa 920-1192, Japan

Page 2: How β2-adrenergic agonists induce skeletal muscle hypertrophy?

424 JPFSM : Kitaura T

synthesis via the IGF-1/PI3K/Akt pathway in striated muscles25-29). Although β2-agonists do not bind to IGF receptors directly, clenbuterol causes a transient increase of IGF-I in cells30) and of IGF-II in rat soleus29) and mas-seter muscle31). The IGF-1/PI3K/Akt pathway might be stimulated by increased IGFs. The peptide hormone ghrelin32), which increases before meals and decreases after meals, is secreted from the stomach and the hypo-thalamus, where it stimulates the secretion of growth hormone (GH) from the anterior pituitary gland. Then GH promotes IGF-1 synthesis and muscle growth. However, the increasing mechanism of IGFs by clenbuterol remain to be resolved. Spurlock et al.33) showed that clenbuterol promotes the synthesis of IGF-1 mRNA-related muscle growth in mouse gracilis muscle. β2-agonists activate the guanine nucleotide exchange factor domain of the β2-adrenoceptor causing the exchange of GDP for GTP at the α stimulatory subunit of the guanine nucleotide-binding regulatory protein (Gαs) and the subsequent dissociation of Gαs from the tightly associated β and γ subunits (Gβγ).

It was shown by von Maltzahn et al that Gαs could di-rectly activate PI3K34). And activation of frizzled (Fzd) 7, another member of the G protein coupled receptor family, induced hypertrophy of rodent myotubes via the activation of the PI3K-Akt-mTOR pathway. Gαs-coupled receptors expressed in skeletal muscle were summarized and reported by Berdeaux and Stewart8) (Fig. 1). It may suggest that β2-adrenoceptor might be able to directly activate PI3K via Gαs to trigger skeletal muscle hyper-trophy. Skeletal muscle hypertrophy was explained by hypertrophy of individual muscle fibers and hyperplasia increasing the number of cells. The former was supported by the increased diameter of muscle fibers; the latter by the increased number of fibers. Increased mRNA and protein content of muscle structural proteins and growth factor like IGF-1, and decreased mRNA and protein con-tent of proteolytic enzymes may explain the hypertrophy of muscle. Table 1 explains high hypertrophy (e.g. H4) by the high increasing grade (e.g. I5) and low degradation grade (e.g. D1) in muscular change. Myostatin is a member of transforming growth fac-tor beta (TGF-β) and inhibits muscle differentiation and growth in myogenesis (Fig. 2). Therefore, decreased myo-statin content by varoius stimulations also will explain increased protein synthesis. However, β2-agonists surpris-ingly showed increased myostatin contents. Therefore, skeletal muscle hypertrophy by β2-agonists should be explained by increased IGF, not by increased myostatin, which may have other functions. Myostatin is expressed in satellite cells and regulates satellite cell quiescence and self-renewal35). The adult muscle cells are post-mitotic and unable to divide, they must acquire external DNA. Satellite cells retain their mitotic capacity36). Higher lev-els of myostatin have been observed in quiescent satellite cells37). It may mean that an increasing number of qui-escent satellite cells serve as myonuclei donors for hy-pertrophied muscle fibers and support preferential incre-ments of DNA content in the latter phase of hypertrophy with β-agonist Cimaterol38).

K H H H H

K H H H

K H H

K H

Deg

rada

tion

grad

e

K

Table 1. Grade of muscular change

Capital letters I and D represent increments and decrements in muscular proteins, respectively. Capital letters H and A represent hypertrophy and atrophy of muscle, respectively. Capital letter K represents no change. The additional numbers indicate the intensity of each item.

Fig. 1 GPCRs that induce myofiber hypertrophy. Four GPCRs (Fzd7, β2-AR, CRFR2, and LPA receptor) have been shown to stimulate hypertrophy in myotubes and/or myofibers. Cognate ligands are italicized. A partial view of the known signaling mediators is shown. CRFR2 stimulates myofiber growth by an uncharacterized effector pathway. β2-AR also induces fiber type transitions to fast-twitch fibers (not shown). Data are cited from Berdeaux and Stewart8).

Page 3: How β2-adrenergic agonists induce skeletal muscle hypertrophy?

425JPFSM : Muscle hypertrophy with β2 agonists

In the cAMP/PKA/CREB pathway, the major effector of cAMP in skeletal muscle is protein kinase A (PKA). β2-agonists bind their own receptors and increase cAMP on PKA regulatory subunits, leading to the release of PKA catalytic subunits, which then drive cAMP response ele-ment binding protein (CREB) phosphorylation on Ser133. CREB phosphorylation occurs rapidly in skeletal muscle in response to β2--agonist administration27). Many genes contain cAMP response elements, and are thus potential targets of CREB39). CREB stimulates retinoblastoma protein promoter activity, together with MyoD, to induce the differentiation of myoblasts into myotubes40). CREB interacts with MyoD on follistatin promoter, thus promot-ing follistatingene expression41) (Fig. 2). Follistatin also known as an activin-binding protein is a protein that in humans is encoded by the FSTgene and inhibits excessive muscle growth. Follistatin has been shown to induce skel-etal muscle hypertrophy, notably by inhibiting myostatin activity42,43). Follistatin-mediated skeletal muscle hyper-trophy is regulated by Smad3 and mTOR independently

of myostatin. Modulation of myostatin activity by fol-listatin could therefore contribute to the hypertrophy and anti-atrophy effects of β2-agonists. Gilson et al.44) showed that the IGF-IR/Akt/mTOR pathway plays a critical role in follistatin-induced muscle hypertrophy. There are vari-ous kinds of follistatin, which have different functions. It is said that only follistatin-344 is active in muscle tissue. Recently, it was reported that a few microRNAs (miR-NAs or miRs) regulate skeletal muscle hypertrophy45-49). For example, Clop et al.50) introduced that miR-1 and miR-206 were important in muscular hypertrophy caus-ing translational inhibition of myostatin gene. Sun Y et al.51) showed that mTOR regulated miR-1 and follistatin, i.e. the mTOR-miR-1-HDAC4-follistatin pathway. These might bring a novel explanation of muscular hypertrophy by β2-agonists.

Decrement of structural proteins

On the ubiquitin-proteosome system, two typical E3

Fig. 2 A model for Gαs-, Gαi-, and Gβγ-mediated signalling in skeletal muscle. See text for details. Dotted lines describe pathways whose molecular mechanisms and/or role in adult skeletal muscle have yet to be completely defined. Open arrows indicate a transcriptional regulation. CREB: cAMP response element binding protein; Epac: exchange protein activated by cAMP; FoxO: forkhead transcription factor; G: guanine nucleotide-binding regulatory protein; Gαi: α inhibitory subunit of G protein; Gαs: α stimulatory subunit of G protein; Gβ: β subunit of G protein. Gγ: γ subunit of G protein; GSK: glycogen synthase kinase; HDAC: histone deacetylase; IGF: insulin-like growth factor; MAFbx: muscle atrophy F-box/atrogin; MEF: myocyte enhancer factor; mTOR: mammalian target of rapamycin; MuRF: muscle RING-finger protein; PI3K: phosphoinositide 3-kinase; NR: nuclear receptor; PGC: peroxisome proliferator-activated receptor γ coactivator; PKA: cAMP-dependent protein kinase A; PKC: protein kinase C; Rap: Ras-related protein; SIK: salt induced kinase. Data are cited from Joassard et al.9).

Follistatin

Myostatin IGF-1 β2-agonists

Gβγ GαiGαs

PKA-induced Gαs

to Gαi switching

cAMP

PKA

CREB SIK1

EpacRap

HDAC Myogenin PKC

MyoD

Myf5

MEF2 MuRF1 p70S6K GSK-3β

NR4A3Follistatin PGC-1α4

PGC-1α1FoxOmTOR GSK-3β

PI3K

AKt

MAFbx/Atrogin-1 IGF-1 Myostatin

Activin type IIBreceptor

MuRF1

Protein synthesis Protein degradation

Adenylate cyclase

Muscle gene expression Protein degradation Protein synthesis

Page 4: How β2-adrenergic agonists induce skeletal muscle hypertrophy?

426 JPFSM : Kitaura T

ubiquitin (Ub) ligases, atrogin-1 and muscle ring-finger protein 1 (MuRF-1), are important in regulating the con-tents of MyoD and myosin, respectively, and are activated by the FOXO transcription factors (Fig. 2). Bodin et al.16) and Sandri et al.17) showed Akt-mTOR pathway protein degradation via inhibition of the ubiquitin proteasome system (UPS). Costelli et al.52) demonstrated that clen-buterol treatment suppresses the expression of ubiquitin (Ub) mRNA in rats. And Gonçalves et al.53) suggested that clenbuterol inhibited atrogin-1 expression. Recently Ijiri et al.54) reported that clenbuterol induced muscle hyper-trophy of neonatal chicks owing to decreased atrogin-1 mRNA. In the autophagy system, the breakdown of pro-teins is necessary for amino acid turnover for maintain-ing normal cellular function. Zhao et al.55) showed that inhibition of FOXO3 by activated Akt promotes skeletal muscle loss via the coordinate activation of both UPS and autophagy. Furthermore, the calcium-calpain system also plays important roles for protein turnover. Gonçalves et al.19) showed clenbuterol suppresses proteasomal and lysosomal proteolysis in rat soleus muscles. With clen-buterol, Douillard et al.56) showed the inhibition of calpain proteinase in rat skeletal muscles, and Higgins et al.21) showed the inhibited activity of calpain in lambs. Even if proteolysis is activated by β2-agonists, more activated protein synthesis will produce more structural proteins with their own fragmented amino acids and amino acids from foods. It is just like I4 and D3 representing hyper-trophy (H1) in grade of muscular change (Table 1).

Conclusion

It is true that muscular hypertrophy is induced by β2-agonists. The medical use of drugs may bring many ben-efits for many patients. The pharmacological mechanisms and effects of β2-agonists on skeletal muscle are becom-ing clearer, in part, due to new analyzing methods for analysis of signaling pathways and downstream effectors regulating protein homeostasis. However, undesirable side effects like cardiac muscle damage and hypertension from the drugs must be taken into account. More novel trials like miRNA function analysis may help to elucidate the mechanism of muscular hypertrophy and other phar-macological functions.

Acknowledgments

This work was supported by Grant-in-Aid for Scientific Re-search (No. 21500628 to T.K.) from the Ministry of Education, Science, Sports, and Culture of Japan.

References

1) Ryall JG, Sillence MN and Lynch GS. 2006. Systemic ad-ministration of β2-adrenoceptor agonists, formoterol and sal-meterol, elicit skeletal muscle hypertrophy in rats at micro-

molar doses. Br J Pharmacol 147: 587-595. 2) Sato S, Nomura S, Kawano F, Tanihata J, Tachiyashiki K and

Imaizumi K. 2008. Effects of the β2-agonist clenbuterol on β1- and β2-adrenoceptor mRNA expressions of rat skeletal and left ventricle muscles. J Pharmacol Sci 107: 393-400.

3) Kitaura T, Tsunekawa N and Kraemer WJ. 2002. Inhibited longitudinal growth of bones in young male rats by clen-buterol. Med Sci Sports Exerc 34: 267-273.

4) Sneddon AA, Delday MI and Maltin CA. 2000. Amelioration of denervation-induced atrophy by clenbuterol is associated with increased PKC-alpha activity. Am J Physiol Endocrinol Metab 279: E188-E195.

5) Petrou M, Wynne DG, Boheler KR and Yacoub MH. 1995. Clenbuterol induces hypertrophy of the latissimus dorsi muscle and heart in the rat with molecular and phenotypic changes. Circulation 92(9 Suppl): II483-489.

6) Lynch GS and Ryall JG. 2008. Role of β-adrenoceptor signal-ing in skeletal muscles: implications for muscle wasting and disease. Physiol Rev 88: 729-767.

7) Sato S, Shirato K, Kizaki T, Ohno H, Tachiyashiki K and Imaizumi K. 2012. Effects of β2-agonists and exercise on β2-adrenergic receptor signaling in skeletal muscles. J Phys Fit-ness Sports Med 1: 139-144.

8) Berdeaux R and Stewart R. 2012. cAMP signaling in skeletal muscle adaptation: hypertrophy, metabolism, and regenera-tion. Am J Physiol Endocrinol Metab 303: E1-E17.

9) Joassard OR, Durieux AC and Freyssenet DG. 2013. β2-Adrenergic agonists and the treatment of skeletal muscle wasting disorders. Int J Biochem Cell Biol 45: 2309-2321.

10) Otto A and Patel K. 2010. Signalling and the control of skeletal muscle size. Experimental Cell Research 316: 3059-3066.

11) Ishii N, Ogasawara R, Kobayashi K and Nakazato K. 2012. Roles played by protein metabolism and myogenic progeni-tor cells in exercise-induced muscle hypertrophy and their relation to resistance training regimens. J Phys Fitness Sports Med 1: 83-94.

12) Miyazaki M. 2012. Growth factor-dependent and indepen-dent regulation of skeletal muscle mass. - Is IGF-1 necessary for skeletal muscle hypertrophy?-. J Phys Fitness Sports Med 2: 101-106.

13) Steelman CA, Recknor JC, Nettleton D and Reecy JM. 2006. Transcriptional profiling of myostatin-knockout mice impli-cates Wnt signaling in postnatal skeletal muscle growth and hypertrophy. FASEB J 20: 580-582.

14) Abo T, Iida RH, Kaneko S, Suga T, Yamada H, Hamada Y and Yamane A. 2012. IGF and myostatin pathways are re-spectively induced during the earlier and the later stages of skeletal muscle hypertrophy induced by clenbuterol, a β-adrenergic agonist. Cell Biochem Funct 30: 671-676.

15) Kim KH, Kim YS and Yang J. 2011. The muscle-hypertro-phic effect of clenbuterol is additive to the hypertrophic ef-fect of myostatin suppression. Muscle Nerve 43: 700-707.

16) Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, Poueymirou WT, Panaro FJ, Na E, Dharmarajan K, Pan ZQ, Valenzuela DM, DeChiara TM, Stitt TN, Yanco-poulos GD and Glass DJ. 2001. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 294: 1704-1708.

17) Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH and Goldberg AL. 2004.

Page 5: How β2-adrenergic agonists induce skeletal muscle hypertrophy?

427JPFSM : Muscle hypertrophy with β2 agonists

Foxo transcription factors induce the atrophy-related ubiqui-tin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117: 399-412.

18) Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, Metzger D, Reggiani C, Schiaffino S and San-dri M. 2009. Autophagy is required to maintain muscle mass. Cell Metab 10: 507-515.

19) Gonçalves DA, Silveira WA, Lira EC, Graça FA, Paula-Gomes S, Zanon NM, Kettelhut IC and Navegantes LC. 2012. Clenbuterol suppresses proteasomal and lysosomal proteolysis and atrophy-related genes in denervated rat so-leus muscles independently of Akt. Am J Physiol Endocrinol Metab 302: E123-E133.

20) Joassard OR, Amirouche A, Gallot YS, Desgeorges MM, Castells J, Durieux AC, Berthon P and Freyssenet DG. 2013. Regulation of Akt-mTOR, ubiquitin-proteasome and autoph-agy-lysosome pathways in response to formoterol admin-istration in rat skeletal muscle. Int J Biochem Cell Biol (In press).

21) Higgins JA, Lasslett YV, Bardsley RG and Buttery PJ. 1988. The relation between dietary restriction or clenbuterol (a se-lective β2 agonist) treatment on muscle growth and calpain proteinase (EC 3.4.22.17) and calpastatin activities in lambs. Br J Nutr 60: 645-652.

22) Navegantes LC, Resano NM, Migliorini RH and Kettelhut IC. 2001. Catecholamines inhibit Ca2+-dependent proteolysis in rat skeletal muscle through β2-adrenoceptors and cAMP. Am J Physiol Endocrinol Metab 281: E449-E454.

23) Kemp CM, Oliver WT and Wheeler TL. 2013. The effects of Capn1 gene inactivation on skeletal muscle growth, de-velopment, and atrophy, and the compensatory role of other proteolytic systems. J Anim Sci 91: 3155-3167.

24) Ito N, Miyagoe-Suzuki Y and Takeda S. 2013. Molecular ba-sis of muscle hypertrophy and atrophy: Potential therapy for muscular dystrophy. J Phys Fitness Sports Med 2: 179-184.

25) Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence LC, Glass DJ and Yancopoulos GD. 2001. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3: 1014-1019.

26) Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, Yancopoulos GD and Glass DJ. 2001. Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol 3: 1009-1013.

27) Koopman R, Gehrig SM, Leger B, Trieu J, Walrand S, Mur-phy KT and Lynch GS. 2010. Cellular mechanisms under-lying temporal changes in skeletal muscle protein synthesis and breakdown during chronic β-adrenoceptor stimulation in mice. J Physiol 588: 4811-4823.

28) Kline WO, Parano FJ, Yang H and Bodine SC. 2007. Ra-pamycin inhibits the growth and muscle-sparing effects of clenbuterol. J Appl Physiol 102: 740-747.

29) Sneddon AA, Delday MI, Steven J and Maltin CA. 2001. Elevated IGF-II mRNA and phosphorylation of 4E-BP1 and p70S6k in muscle showing clenbuterol-induced anabolism. Am J Physiol Endocrinol Metab 281: E676-E682.

30) Awede BL, Thissen JP and Lebacq J. 2002. Role of IGF-I and IGFBPs in the changes of mass and phenotype induced in rat soleus muscle by clenbuterol. Am J Physiol Endocrinol Metab 282: E31-E37.

31) Matsumoto T, Akutsu S, Wakana N, Morito M, Shimada A and Yamane A. 2006. The expressions of insulin-like growth factors, their receptors, and binding proteins are related to the mechanism regulating masseter muscle mass in the rat. Arch Oral Biol 51: 603-611.

32) Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H and Kangawa K. 1999. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402: 656-660.

33) Spurlock DM, McDaneld TG and McIntyre LM. 2006. Changes in skeletal muscle gene expression following clen-buterol administration. BMC Genomics 7: 320.

34) von Maltzahn J, Bentzinger CF and Rudnicki MA. 2012. Wnt7a-Fzd7 signalling directly activates the Akt/mTOR ana-bolic growth pathway in skeletal muscle. Nat Cell Biol 14: 186-191.

35) Cornelison DDW, Olwin BB, Rudnicki MA and Wold BJ. 2000. MyoD−/− satellite cells in single-fiber culture are dif-ferentiation defective and MRF4 deficient. Developmental Biology 224: 122-137.

36) Mauro A. 1961. Satellite cell of skeletal muscle fibers. Jour-nal of Cell Biology 9: 493-495.

37) McCroskery S, Thomas M, Maxwell L, Sharma M and Kam-badur R. 2003. Myostatin negatively regulates satellite cell activation and self-renewal. Journal of Cell Biology 162: 1135-1147.

38) Beermann DH, Butler WR, Hogue DE, Fishell VK, Dalrym-ple RH, Ricks CA and Scanes CG. 1987. Cimaterol-induced muscle hypertrophy and altered endocrine status in lambs. J Anim Sci 65: 1514-1524.

39) Zhang X, Odom DT, Koo SH, Conkright MD, Canettieri G, Best J, Chen H, Jenner R, Herbolsheimer E, Jacobsen E, Kadam S, Ecker JR, Emerson B, Hogenesch JB, Unterman T, Young RA and Montminy M. 2005. Genome-wide analysis of cAMP-response element binding protein occupancy, phos-phorylation, and target gene activation in human tissues. Proc Natl Acad Sci USA 102: 4459-4464.

40) Magenta A, Cenciarelli C, De Santa F, Fuschi P, Martelli F, Caruso M and Felsani A. 2003. MyoD stimulates RB promot-er activity via the CREB/p300 nuclear transduction pathway. Mol Cell Biol 23: 2893-2906.

41) Iezzi S, Di Padova M, Serra C, Caretti G, Simone C, Maklan E, Minetti G, Zhao P, Hoffman EP, Puri PL and Sartorelli V. 2004. Deacetylase inhibitors increase muscle cell size by pro-moting myoblast recruitment and fusion through induction of follistatin. Dev Cell 6: 673-684.

42) Amthor H, Nicholas G, McKinnell I, Kemp CF, Sharma M, Kambadur R and Patel K. 2004. Follistatin complexes Myostatin and antagonises Myostatin-mediated inhibition of myogenesis. Dev Biol 270: 19-30.

43) Gilson H, Schakman O, Kalista S, Lause P, Tsuchida K and Thissen JP. 2009. Follistatin induces muscle hypertrophy through satellite cell proliferation and inhibition of both myostatin and activin. Am J Physiol Endocrinol Metab 297: E157-E164.

44) Kalista S, Schakman O, Gilson H, Lause P, Demeulder B, Bertrand L, Pende M and Thissen JP. 2012. The type 1 insu-lin-like growth factor receptor (IGF-IR) pathway is manda-tory for the follistatin-induced skeletal muscle hypertrophy. Endocrinology 153: 241-253.

45) Yuasa K, Hagiwara Y, Ando M, Nakamura A, Takeda S and Hijikata T. 2008. MicroRNA-206 is highly expressed in new-

Page 6: How β2-adrenergic agonists induce skeletal muscle hypertrophy?

428 JPFSM : Kitaura T

ly formed muscle fibers: implications regarding potential for muscle regeneration and maturation in muscular dystrophy. Cell Struct Funct 33: 163-169.

46) Williams AH, Liu N, van Rooij E and Olson EN. 2009. Mi-croRNA control of muscle development and disease. Curr Opin Cell Biol 21: 461-469.

47) van Rooij E, Liu N and Olson EN. 2008. MicroRNAs flex their muscles. Trends Genet 24: 159-166.

48) Townley-Tilson WH, Callis TE and Wang D. 2010. MicroR-NAs 1, 133, and 206: critical factors of skeletal and cardiac muscle development, function, and disease. Int J Biochem Cell Biol 42: 1252-1255.

49) Ge Y and Chen J. 2011. MicroRNAs in skeletal myogenesis. Cell Cycle 10: 441-448.

50) Clop A, Marcq F, Takeda H, Pirottin D, Tordoir X, Bibé B, Bouix J, Caiment F, Elsen JM, Eychenne F, Larzul C, Laville E, Meish F, Milenkovic D, Tobin J, Charlier C and Georges M. 2006. A mutation creating a potential illegitimate microR-NA target site in the myostatin gene affects muscularity in sheep. Nat Genet 38: 813-818.

51) Sun Y, Ge Y, Drnevich J, Zhao Y, Band M and Chen J. 2010. Mammalian target of rapamycin regulates miRNA-1 and fol-listatin in skeletal myogenesis. J Cell Biol 189: 1157-1169.

52) Costelli P, Garcia-Martinez C, Llovera M, Carbo N, Lopez-

Soriano FJ, Agell N, Tessitore L, Baccino FM and Argiles JM. 1995. Muscle protein waste in tumor-bearing rats is ef-fectively antagonized by a β2-adrenergic agonist (clenbuter-ol). Role of the ATP-ubiquitin-dependent proteolytic path-way. J Clin Invest 95: 2367-2372.

53) Gonçalves DA, Lira EC, Baviera AM, Cao P, Zanon NM, Arany Z, Bedard N, Tanksale P, Wing SS, Lecker SH, Kettel-hut IC and Navegantes LC. 2009. Mechanisms involved in 3’,5’-cyclic adenosine monophosphate-mediated inhibition of the ubiquitin-proteasome system in skeletal muscle. Endo-crinology 150: 5395-5404.

54) Ijiri D, Ishitani K, Matsubara T, Hirabayashi M, Kanai Y and Ohtsuka A. 2013. In vivo administration of β2-agonist clen-buterol and subsequent increase in skeletal muscle mass in neonatal chicks. J Poult Sci 50: 150-154.

55) Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, Lecker SH and Goldberg AL. 2007. FoxO3 coordinately ac-tivates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 6: 472-483.

56) Douillard A, Galbes O, Rossano B, Vernus B, Bonnieu A, Candau R and Py G. 2011. Time course in calpain activity and autolysis in slow and fast skeletal muscle during clen-buterol treatment. Can J Physiol Pharmacol 89: 117-125.