G Model

ARTICLE IN PRESS

YPHRS 2837 1–16

Pharmacological Research xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Pharmacological Research journal homepage: www.elsevier.com/locate/yphrs

Review

1

Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action

2

3

4 5 6 7

Q1

Vikas Dutt a , Sanjeev Gupta a , Rajesh Dabur b , Elisha Injeti c , Ashwani Mittal a,∗ a b c

Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, Haryana 136119, India Biochemistry Department, MD University, Rohtak, Haryana 124001, India Pharmaceutical Sciences Department, Cedarville University, OH 45314, USA

8

9 45

a r t i c l e

i n f o

a b s t r a c t

10 11 12 13 14 15

Article history: Received 6 March 2015 Received in revised form 24 May 2015 Accepted 24 May 2015 Available online xxx

16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44

Keywords: Atrophy Cachexia Eicosapentaenoic acid Resveratrol Cox2 inhibitor Histone decetylase inhibitor Phosphodiesterase inhibitor ␤-Adrenoceptor agonists Megestrol acetate Anti-cytokines Chemical compounds studied in this article: EPA (PubChem CID: 446284) ␤-Hydroxy-␤-methylbutyrate (PubChem CID: 69362) Cis-resveratrol (PubChem CID: 1548910) Trans-resveratrol (PubChem CID: 445154) Ghrelin (PubChem CID: 44576256) Celecoxib (PubChem CID: 2662) Meloxicam (PubChem CID: 54677470) Trichostatin A (PubChem CID: 444732) Torbafylline (PubChem CID: 65888) Pentoxifylline (PubChem CID: 4740) Formoterol (PubChem CID: 3410) Clenbuterol (PubChem CID: 2783) Enobosarm (PubChem CID: 11326715) Thalidomide (PubChem CID: 5426) Megestrol acetate (PubChem CID: 11683)

Over the last two decades, new insights into the etiology of skeletal muscle wasting/atrophy under diverse clinical settings including denervation, AIDS, cancer, diabetes, and chronic heart failure have been reported in the literature. However, the treatment of skeletal muscle wasting remains an unresolved challenge to this day. About nineteen potential drugs that can regulate loss of muscle mass have been reported in the literature. This paper reviews the mechanisms of action of all these drugs by broadly classifying them into six different categories. Mechanistic data of these drugs illustrate that they regulate skeletal muscle loss either by down-regulating myostatin, cyclooxygenase2, pro-inflammatory cytokines mediated catabolic wasting or by up-regulating cyclic AMP, peroxisome proliferator-activated receptor gamma coactivator-1␣, growth hormone/insulin-like growth factor1, phosphatidylinositide 3-kinases/protein kinase B(Akt) mediated anabolic pathways. So far, five major proteolytic systems that regulate loss of muscle mass have been identified, but the majority of these drugs control only two or three proteolytic systems. In addition to their beneficial effect on restoring the muscle loss, many of these drugs show some level of toxicity and unwanted side effects such as dizziness, hypertension, and constipation. Therefore, further research is needed to understand and develop treatment strategies for muscle wasting. For successful management of skeletal muscle wasting either therapeutic agent which regulates all five known proteolytic systems or new molecular targets/proteolytic systems must be identified. © 2015 Elsevier Ltd. All rights reserved.

Abbreviations: Cox, cyclooxygenase; CREB, cAMP response element binding protein; CACS, cancer-related anorexia and cachexia syndrome; CRP, C-reactive protein; COPD, chronic obstructive pulmonary disease; DAG, des-acyl ghrelin; ERK, extracellular signal-regulated kinase; EPA, eicosapentaenoic acid; FoxO, forkhead box O; Epac, exchange protein directly activated by cAMP; eIF4G and eIF4E, eukaryotic translation initiation factor 4G and 4E; HAT, histone acetyltransferase; HDAC, histone deacetylase; HETE, hydroxyeicosatetraenoic acid; HMB, ␤-hydroxy-␤-methylbutyrate; LLC, Lewis lung carcinoma; MAC, murine adeno-carcinoma; NY, neuropeptide Y; NF␬B, nuclear factor kappa; PUFA, polyunsaturated fatty acid; PIF, proteolysis-inducing factor; PPAR␥, peroxisome proliferator-activated receptor; PGE, prostaglandin; PDE, phosphodiesterase; PKA, protein kinase A; PTX, pentoxifylline; ROS, reactive oxygen species; STAT, signal transducer and activator of transcription; SARM, selective androgen receptor modulator; SOD, superoxide dismutase; TNF␣, tumor necrosis factor; TWEAK, TNF␣ like weak inducer of apoptosis; TLR, toll-like receptor; TPA, 12-0-tetradecanoylphorbol-13-acetetate; TGF␤1, transforming growth factor beta 1; TSA, trichostatin A; CKD, chronic kidney disease; Activin type II receptor, ActRIIB, ActRIIA; Activinlike kinase, ALK4, ALK5; MuRF1, muscle-specific RING-finger 1. ∗ Corresponding author. Tel.: +91 01744 238049; fax: +91 01744 238008. E-mail address: [email protected] (A. Mittal). http://dx.doi.org/10.1016/j.phrs.2015.05.010 1043-6618/© 2015 Elsevier Ltd. All rights reserved.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

Q2

G Model YPHRS 2837 1–16

V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

2 46

47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75

ARTICLE IN PRESS

Contents 1. 2.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Classification and mechanisms of actions of drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1. Natural compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1.1. Eicosapentanoic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1.2. ␤-Hydroxy-␤-methylbutyrate (HMB) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1.3. Resveratrol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1.4. Ghrelin and its receptor agonist . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.2. Enzyme inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.2.1. Cox2 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.2.2. Histone decetylase inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.2.3. PDE inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3. ␤-Adrenoceptor agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3.1. Formoterol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3.2. Clenbuterol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4. Anti-cytokines agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4.1. Anti-TNF␣ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4.2. Thalidomide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.5. Other investigational drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.5.1. Enobosarm . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.5.2. GLPG0492 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.5.3. OHR/AVR118 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.5.4. APD209 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.5.5. Anti-myostatin/activin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.6. Miscellaneous agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.6.1. Megestrol acetate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

76

77

1. Introduction

In the current literature, three factors that can initiate loss of 79 skeletal muscle mass have been identified. They are (1) chronic 80 diseases (cachexia) like diabetes, cancer, chronic obstructive pul81 monary disease (COPD), acquired immune deficiency syndrome 82 (AIDS), and renal/cardiac failure, (2) disuse conditions (atrophy) 83 like denervation, immobolization, and microgravity (3) aging 84 (sarcopenia). These factors gradually lead to distinct phenotypic 85 changes in the skeletal muscle by accelerating protein degrada86 tion [1–5]. Muscle wasting and weakness generated in each of 87 these conditions (cachexia, atrophy, and sarcopenia) is a complex 88 and highly regulated phenomenon, characterized by substantial 89 decrease in muscle fiber cross-sectional area, myonuclear number, 90 protein content and muscle strength while increasing in fatigabil91 ity and resistance to insulin [2,6–8]. In addition, it is also associated 92 with an increased risk of death. 93 Beyond a reduced survival rate, wasting is also linked to poor 94 functional status and quality of life. Up to one-third of all can95 cer patients die due to direct consequences of cachexia and 96 not from cancer, while in AIDS, more than 5% weight loss over a period of 4-months are associated with an increased risk of 97 death and opportunistic infections. Studies have revealed that 98 different types of molecular triggers/catabolic players such as 99 myostatin, pro-inflammatory cytokines i.e. tumor necrosis fac100 tor alpha (TNF␣), interleukin-1 beta (IL-1␤), interleukin-6 (IL-6), 101 TNF-like weak inducer of apoptosis (TWEAK), interferon gamma 102 (IFN␥) are involved in muscle wasting under above mentioned 103 clinical settings [7,9,10]. These cytokines on binding to their 104 respective receptor results in activation of the NF␬B, a common 105 transcription factor in most of the protein catabolic pathways 106 Q4 leading to proteolysis in skeletal muscles (Supplementary Fig. 107 1). This makes nuclear factor kappa B (NF␬B) an attractive 108 pharmaceutical target for therapeutic interventions. Other impor109 tant targets include anabolic pathways (phosphatidylinositide 78Q3

110

3-kinases/protein kinase B; PI3K/Akt) and five major proteolytic machineries like Ca2+ -dependent calpain, Ca2+ -independent cathepsin L, autophagy, ubiquitin (Ub)–proteasome system and the caspase system [3,11,12]. Caspases are critical not only for muscle atrophy but also for the apoptotic process. The majority of the muscles wasting studies have demonstrated that autophagy system uses lysosomal enzymes (like cathepsin L) for the processing of damaged cellular components [12]. There are some studies which have reported enhanced cathepsin L activity without any change in the level of autophagy related markers under atrophic conditions [13–15]. These studies highlight the possibility of cathepsin L dependent and independent autophagy systems. Moreover, several reports in literature indicate that cathepsin L may be present in extracellular compartment independent to lysosomal fraction which further illustrates the probable special role of this protease during atrophy [16,17]. Supplementary material related to this article can be found, in the online version, at http://dx.doi.org/10.1016/j.phrs.2015.05.010 In spite of many promising therapeutic targets for treating muscle wasting, not even a single drug is clinically proven to be safe. In this review, we classify nineteen potential drugs (that are in use at laboratory/preclinical level of investigation using different atrophic models) into six categories and then discuss their mechanism of action (Fig. 1). Q5

111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134

2. Classification and mechanisms of actions of drugs

135

2.1. Natural compounds

136

The mechanisms of action of four natural compounds are discussed in this section. These compounds are (A) eicosapentanoic acid, (B) ␤-hydroxy-␤-methylbutyrate, (C) resveratrol, and (D) ghrelin and its receptor agonists.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

137 138 139 140

G Model

ARTICLE IN PRESS

YPHRS 2837 1–16

V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

X

Inhibition Activator Down-regulation Up-regulation Mediated Inhibition

Q8

Fig. 1. Symbols used in the figures.

2.1.1. Eicosapentanoic acid Eicosapentaenoic acid (EPA) is a naturally occurring long-chain Q6 142 polyunsaturated fatty acid (PUFA) of omega-3 (␻-3) family mainly 143 found in deep-sea fish and certain algae. EPA exhibits diverse phar144 macological actions including anti-genotoxic, anti-oxidant and 145 chemo-preventive effects. Numerous studies showed that in can146 cer patients there is a progressive muscle wasting (75% decrease in 147 skeletal muscle protein mass) which makes it a key reason for the 148 cause of cachexia. This is due to up-regulation in the level of various 149 pro-inflammatory cytokines and their mediated activation of skele150 tal muscle specific proteolytic system. One of those systems is the 151 NF␬B-dependent ubiquitin–proteasome pathway which is respon152 sible for degradation of myofibrillar proteins and thus skeletal 153 muscle wasting [18–20]. Studies have shown that EPA suppresses 154 the production of pro-inflammatory cytokines such as IL-1, TNF␣ 155 and IL-6 in cancer induced cachexia [21–23]. EPA also inhibits the 156 NF␬B activation by suppressing IB kinase activity (IKK, an enzyme 157 that phosphorylate I␬B and promotes its degradation). This stops 158 the translocation of NF␬B from cytosol into nucleus and prevents 159 the expression of various components of the proteolytic machinery 160 such as muscle-specific E3 ubiquitin ligase (muscle-specific RING161 finger 1, MuRF1). This process tremendously helps in preserving 162 the skeletal muscle mass [24,25]. 163 In vivo studies have shown that murine adeno-carcinoma-16 164 (MAC16) tumor bearing mice when treated with EPA displayed 165 decreased proteasome activity (i.e. decrease in expression of 166 20S proteasome protein and the p42 regulator) in gastrocne167 mius muscle [26]. EPA also controls the proteolysis-inducing 168 factor (PIF)/15-lipoxygenase (15LOX)/15-hydroxyeicosatetraenoic 169 acid (15HETE)-mediated activation of NF␬B pathway in cancer 170 cachexia. This study shows that EPA down-regulates the protein 171 catabolism process by inhibiting the release of arachidonic acid 172 (AA) from muscle cells in the presence of the PIF and its conver173 sion into 15LOX metabolite i.e. 15HETE. This is the only metabolite 174 formed from arachidonic acid that has the capability of inducing 175 protein degradation through NF␬B/Ub–proteasome system by acti176 vating IKK and protein kinase C (PKC). It can be concluded that 177 EPA regulates the cancer cachexia by targeting the production of 178 15HETE and stabilizing the I␬B/NF␬B complex [27,28]. 179 A large number of studies have shown that carcinoma bear180 ing mice (such as Colon-26 and Lewis lung) can induce cancer 181 cachexia by up-regulating the levels of expression of paired box7 182 (Pax7), which inhibits the myogenin transcription factor thereby 183 blocking the myogenesis program [29–32]. However, EPA treat184 ment in association with endurance exercise for short periods can 185 partially prevent this increase in Pax7 levels in Lewis lung carci186 noma (LLC) bearing mice and thus improving the muscle wasting 187 141

3

condition [32]. EPA also stimulates mitochondrial biogenesis by up-regulating peroxisome proliferator-activated receptor gamma coactivator-1␣ (PGC-1␣) protein levels in LLC-bearing mice and carries out fiber-types switching from glycolytic fibers to oxidative fibers which makes the fiber less susceptible to atrophy [32]. EPA administration can also increase the reactive oxygen species (ROS) scavenging activity of superoxide dismutase (SOD), and thus increased the weight gain in pancreatic cancer patients [33]. Like cancer cachexia, EPA also impacts sepsis induced muscle wasting by down-regulating the muscle protein catabolism [34]. Additionally, EPA treatments in C2C12 cells induce skeletal muscle differentiation and prevent apoptosis and necrosis by reverting TNF␣ inhibitory effects. Studies show that TNF␣ inhibits the muscle differentiation either by blocking myogenesis in differentiating myoblasts and inducing apoptosis of myoblast/myotubes or by blocking peroxisome proliferator-activated receptor (PPAR␥; a major transcriptional regulator of lipid metabolism) activity. It is also evident that altered expression of PPAR␥ inhibits not only myotubes formation but also the expression of muscle-specific myogenic proteins (myogenin and MyoD). These findings illustrate the fact that EPA can promote muscle cells differentiation under pro-inflammatory cytokines mediated catabolic conditions [24,35]. Similar to EPA, fish oil (abundant in ␻3 PUFAs such as EPA and docosahexaenoic acid) is also very effective in regulating muscle pathophysiology. Studies have shown that dietary supplementation of fish oil prevents the LPS-induced inhibition of Akt signaling by decreasing forkhead box O1 (FoxO1) and FoxO4 abundance along with decreasing mRNA expression of muscle-specific E3 ubiquitin ligases i.e. muscle-specific F-box protein (MAFbx; atrogin1) and MuRF1, markers of the Ub–proteasome system in gastrocnemius and latissimus dorsi muscles of the pigs. This treatment reduced the TNF␣ and cyclooxygenase2 (Cox2) mRNA abundance in myofibers via regulation of muscle toll-like receptor (TLR) and nucleotide-binding oligomerization domain protein (NOD) signaling pathways, leading to improved skeletal muscle mass [25]. Studies also illustrate that fish oil or EPA inhibits the muscle degeneration and inflammation and ameliorates muscle physiological function as evaluated by grip test in dystrophindeficient (mdx) mice model [36]. Despite the above mentioned anti-cachectic effects of EPA such as gain in lean body mass, improvement in grip strength, reductions in pro-inflammatory cytokines (TNF␣, IL-6) in various cachectic conditions, few studies also reported the ineffectiveness of EPA in treating these conditions. Clinical results from randomized trials have suggested that EPA administration may not be effective in treating cancer cachexia. Likewise in another clinical trial, impact of EPA supplement was compared with megestrol acetate (standard cachexia treatment drug) in cancer cachexia where it was found that megestrol acetate is more effective than EPA in increasing body weight [22,37,38]. Overall these studies show that EPA regulates cachexia by reducing the expression level of large number of pro-inflammatory molecules such as TNF␣, IL-1, IL-6, PIF, Cox2, 15HETE, IKK, NF␬B, FoxO along with increasing the level of PPAR␥ which resulted in correcting the metabolic abnormalities as well as modulate the immune function (Fig. 2). However contradictory claims of other studies regarding ineffectiveness of EPA in cancer cachexia may limit its use. Further elaborate studies are needed to confirm its anti-cachectic role [39]. 2.1.2. ˇ-Hydroxy-ˇ-methylbutyrate (HMB) HMB a leucine metabolite is formed by trans-amination of leucine to ␣-ketoisocaproate in muscle followed by its oxidation in the cytosol of the liver. HMB is used as a therapeutic supplement to treat various muscle disorders [40–43]. HMB

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247

248 249 250 251 252

G Model YPHRS 2837 1–16 4

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

the loss of muscle mass [40]. The administration of HMB in combination with arginine and glutamine amino-acids to stage IV cancer patients, increased their body mass when compared to placebo [44]. Furthermore, HMB exerts protective effects against dexamethasone-induced atrophy by down-regulating the activated Akt/FoxO and MuRF1 levels which controls autophagy and ubiquitin dependent proteolytic systems [42,45]. HMB is also involved in the regulation of satellite cell proliferation probably through reduction of nuclear apoptosis [46]. Overall, HMB treatments regulate skeletal muscle wasting by stimulating protein anabolic and inhibiting protein catabolic pathways (Fig. 3).

Fig. 2. Schematic representation of EPA therapeutic action in tumor/cancer/sepsis cachexia.

253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271

stimulates protein anabolism and inhibits protein catabolism under PIF-induced cachectic condition [41]. This suggests that HMB facilitates hyper-phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) resulting in the formation of active translation initiation complex (eIF4G:eIF4E). Besides this, HMB also up-regulates the phosphorylation of mammalian target of rapamycin (mTOR)/p70S6K pathway which is required for the formation of translation apparatus and elongation factors. Additionally, HMB reverses the PIF-induced auto-phosphorylation of double strand RNA-dependent protein kinase (PKR), leading to inhibition of phosphorylation of ␣-subunit of eIF2 resulting in accelerated translation process. In other words, HMB stimulates protein synthesis in muscle by activating multiple pathways leading to increased translation thereby compensating the loss of protein. In an experimental animal model of cancer-induced cachexia, HMB treatment not only down-regulated the expression of key regulatory components of 19S and 20S proteasome–proteolytic pathway but also stimulated protein synthesis which further regulates

2.1.3. Resveratrol Resveratrol (3,5,4 -trihydroxystilbene) is a natural polyphenol present mainly in peanuts, pines, skin of grapes, especially in red wine and exists both as cis- and trans-isomers. But the trans-isomer is the biologically active form [47–49]. Resveratrol is known to possess diverse therapeutic activities including anti-inflammatory, anti-atherosclerotic, anti-tumoral, cardioprotective, anti-diabetic and anti-oxidative effects [48,50–52]. A large number of studies performed in vitro and in vivo have confirmed that resveratrol treatment can prevent protein degradation induced by PIF, Angiotensin I and II, phorbal ester, 12-O-tetradecanoylphorbol-13acetetate (TPA), dexamethasone. In addition, resveratrol has shown its protective effect on muscle wasting under diverse catabolic conditions including cachexia and disuse [48,49,53]. Besides this, resveratrol treatment shows its anti-cachectic role by inhibiting NF␬B activity and MuRF1 expression in MAC16 tumor and C26 adenocarcinoma bearing mice [54,55]. Studies have shown that resveratrol regulates TNF␣ induced atrophy by up-regulating the phosphorylation of Akt, p70S6K, mTOR and 4E-BP1 and protects the dexamethasone-induced atrophy by inhibiting the increase in atrogin1 and MuRF1 expression via activation of SIRT1 (sirtuin, a histone deacetylase) and PGC-1␣ [48,49]. In streptozotocin induced diabetic rat model, resveratrol administration enhanced the insulin sensitivity, AMP-activated protein kinase (AMPK) and SIRT1 activities, and mitochondrial genesis and down-regulated the mRNA expression of pro-inflammatory cytokines (IL-1␤ and IL-6) and NF␬B expression [52,55]. Furthermore, resveratrol treatment for COPD decreases the levels of TNF␣ and increases the expression of SIRT1 which ultimately activates AMPK and regulates the

Fig. 3. Schematic representation of HMB therapeutic action in PIF/cancer/dexamethasone-induced atrophy.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

272 273 274 275 276 277 278 279 280 281 282

283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

5

Fig. 5. Schematic representation of ghrelin/its mimetic therapeutic action in cancer, COPD, CKD, unloading induced muscle wasting.

Fig. 4. Schematic representation of reserveratrol COPD/diabetes/TNF␣/dexamethasone-induced atrophy.

312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328

329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344

therapeutic

action

in

muscle wasting [56]. Pharmacokinetic and toxicological studies have revealed that higher doses of resveratrol (100 mg/kg/day) in humans and up to 700 mg/kg in rats do not show any adverse effects [51,57]. But there are conflicting data to show that resveratrol treatment is unable to reverse or block the loss of muscle mass and hence lacks anti-cachectic property as studied in rats bearing the Yoshida AH130 ascites hepatoma and in mice bearing the Lewis lung carcinoma [50]. Overall, various studies have indicated that resveratrol treatments control the muscle wasting and increase the muscle insulin sensitivity by down-regulating the level of pro-inflammatory cytokines and up-regulating the level of Akt, AMPK and SIRT1 in COPD/diabetes/TNF␣/dexamethasone-induced atrophy (Fig. 4). However, as Visioli rightfully cautions, these results from cell and animal studies may not always translate into similar results in human studies [58]. 2.1.4. Ghrelin and its receptor agonist Ghrelin (Growth Hormone Release-Inducing), a growth hormone (GH)-releasing polypeptide, is made up of 28 amino acids and mainly secreted from entero-endocrine cells of the stomach. This is found in both acylated and non-acylated forms. It binds to GH secretagogue receptor (formerly known as GHSR-1␣) and stimulates appetite via activation of neuropeptide Y (NY) in the hypothalamus and helps in regulation of body weight [59–61]. In skeletal muscles, it checks muscle wasting by down-regulating the TNF␣ mRNA expression without showing any significant effects on insulin-like growth factor-1 (IGF-1) mRNA in gastrocnemius muscle in a burninduced injury model. Ghrelin precursor peptide (des-acyl ghrelin, DAG) also has regulatory effect on TNF and IFN-induced skeletal muscle atrophy. This study shows that DAG attenuates the cytokine induced reduction in phosphorylation of Akt, FoxO1 and glycogen synthase kinase-3 beta (GSK3␤) in C2C12 myotubes. Besides

this, DAG also inhibits the activation of NF␬B and down-regulates atrogin1/MuRF1 mRNA expression [62,63]. In the lung adenocarcinoma induced cachexia, ghrelin treatment suppressed the inflammation by preventing the induction of TNF␣, IL-1␤, IL-6 and C-reactive protein (CRP). In addition, it also ameliorated skeletal muscle wasting by reducing the expressions of phosphorylated-p38 mitogen-activated protein kinase (p-p38MAPK), activated-NF␬B, FoxO1, MuRF1, and atrogin in the lysates of skeletal muscle of tumor-bearing mice [64]. Similar to its effect in cancer cachexia, ghrelin has shown positive results in regulating loss of skeletal muscle mass in chronic kidney disease (CKD) by enhancing mitochondrial oxidative capacity and Akt phosphorylation. These changes not only enhance insulin sensitivity but also favor skeletal muscle anabolism [60]. A clinical study which involved three week ghrelin treatment of cachectic patients with COPD and chronic heart failure (CHF), an increase in lean body mass and muscle strength pointed toward its anti-cachectic effect [59,65]. In another model of muscle wasting (unloadinginduced muscle atrophy), ghrelin administration acutely increased plasma GH and amplified phosphorylation of signal transducer and activator of transcription (STAT5), which increases IGF-1 mRNA expression in the plantaris muscle. This study illustrates that ghrelin alleviates muscle atrophy by stimulating GH-STAT5-IGF-1 axis in the atrophied muscle [66]. Although ghrelin plays a critical role in stimulating the appetite, increasing body mass and preventing muscle catabolism, its clinical effectiveness is limited due to its half life (30 min) and route of administration (intravenous) [67,68]. Additionally, a ghrelin receptor agonist, anamorelin HCl, has also demonstrated its positive effect in cancer anorexia–cachectic patients by increasing the level of GH, IGF-1, and insulin-like growth factor-binding protein 3 (IGFBP-3) which leads to increased appetite and body mass. Furthermore, it has better half life (7–12 h) compared to ghrelin (0.5 h) [69–71]. Though results of this drug are quite promising for cachexia treatment, further controlled studies with large sample size are still required to understand its efficacy and safety. From various reports it can be concluded that ghrelin has positive effect in controlling skeletal muscle wasting under diverse clinical settings such as cancer, COPD, CKD by up-regulating anabolic molecules (NY, GH, IGF-1, IGFBP-3, STAT5) and downregulating proteolytic systems (activated-NF␬B, FoxO1, MuRF1, and atrogin levels) (Fig. 5). In addition, by reducing the expression of inflammatory molecules including CRP, TNF␣, IL-1␤, and IL-6, this peptide shows systemic anti-inflammatory effect. Due to these promising features this drug is under Phase III clinical study.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

6

Fig. 6. Schematic representation of Cox2 inhibitors therapeutic action in cancer and arthritis induced atrophy.

391

392 393 394

395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414

415 416 417 418 419 420 421 422 423 424 425 426 427

2.2. Enzyme inhibitors The mechanism of action of three different enzyme inhibitors will be discussed in this section. They are Cox2 inhibitors, histone decetylase inhibitors and phosphodiesterase (PDE) inhibitors. 2.2.1. Cox2 inhibitors Cyclooxygenase exists in multiple isoforms i.e. Cox1, Cox2 and Cox3. Of all these isoforms, Cox2 has pro-inflammatory actions and is induced by cytokines and mitogens not only in immune cells but also in other tissues including skeletal muscles. Cox2, a bifunctional enzyme, has both cyclooxygenase and peroxidase activities. The cyclooxygenase activity is responsible for the synthesis of prostaglandins (PGE2) from arachidonic acid while peroxidase activity can generate proximate carcinogens. These prostaglandins perform various actions via specific G-proteincoupled receptors and nuclear peroxisome proliferator-activated receptors (PPARs). Cox2 and PGE2 both are the downstream effectors of cytokine activity and mediate cachexia [72–75]. Preclinical and clinical trials strongly support the potential role for Cox2 inhibitors in the treatment of cancer [76]. So far a number of Cox2 inhibitors have been reported which inhibit PGE2 activity in diverse tumor bearing mice models and showed attenuating impact on the cachectic condition. Out of these inhibitors only two (celecoxib and meloxicam) have been widely used to study Cox2 mediated muscle loss [73,74,77]. 2.2.1.1. Celecoxib. Lai et al. performed a placebo-controlled study with celecoxib on 11 cachectic patients with either head and neck or gastrointestinal cancer. Interestingly, celecoxib treated patients showed a significant increase in body mass index and their quality of life improved as compared to placebo [78]. This reflects its effectiveness in treating cancer cachexia. To understand the efficacy and safety of celecoxib, Mantovani et al. performed a Phase II non-randomized study on cancer cachectic patients. The treatment group showed a significant increase in lean body mass and a decrease in TNF␣ along with improvement of grip strength and quality of life [76]. Not only in cancer cachexia, celecoxib showed its anti-cachectic role in diverse pathophysiological conditions. Rheumatoid arthritis cachectic rabbits when treated with celecoxib

showed reduction in the weight loss as well as in the level of inflammatory molecules (such as IL-6 and NF␬B) [79]. 2.2.1.2. Meloxicam. Meloxicam is another Cox2 inhibitor that inhibits the growth of murine adeno-carcinoma tumors (MAC 13, MAC16). Studies have shown that meloxicam treatment inhibits the LPS induced expression of Cox2, atrogin1 and MuRF1 and regulates the loss in muscle mass of rats by attenuating protein catabolism [75,77]. Similar to cancer, chronic arthritis also induces muscle wasting by mediating the Cox2 pathway. Studies show that Cox2 pathway not only inhibits the GH-IGF1 axis but also increases the TNF␣ and IGFBP-5 mRNA expression that contributes to the activation of ubiquitin–proteasome system and ultimately carry out degradation of muscle protein. However, administration of meloxicam ameliorated muscle loss by preventing arthritisinduced up-regulation of atrogin1 and MuRF1 in arthritic rats [80]. Fig. 6 illustrates the mechanism of action of Cox2 inhibitors in regulation of muscle wasting in cancer and arthritis patients 2.2.2. Histone decetylase inhibitors 2.2.2.1. Trichostatin A (TSA). TSA is a well known inhibitor of class I and II histone deacetylase. Just like phosphorylation/dephosphorylation, acetylation/deacetylation of cellular proteins is also involved in the regulation of muscle mass which is maintained by histone acetyltransferases (HATs) and histone deacetylases (HDACs) activities. Under atrophic condition, this process gets perturbed and causes de-gradation of muscle specific proteins by activation of proteolytic machineries [81,82]. In the denervation-induced muscle wasting, HDAC4 level is up-regulated which further increases the expression level of Ub–proteasome markers i.e. atrogin1 and MuRF1 by downregulating Dach2 level (a transcription factor and a repressor of the myogenin promoter) and accelerate the level of myogenin, a muscle-specific transcription factor essential for muscle development. Myogenin, in turn, creates a positive feedback loop and regulates HDAC4 expression [83,84]. Published data shows that under denervation condition (neuromuscular disorders), TSA modulates the level of atrogenes and control the muscle mass by reducing the HDAC4 activity and myogenin level, and increasing

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

428 429

430 431 432 433 434 435 436 437 438 439 440 441 442 443 444

445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505

506 507 508 509 510 511 512 513 514 515 516 517

518 519 520 521 522 523 524 525 526 527 528

the expression level of Dach2 [85]. TSA treatment improves body weight, myofiber number and myofiber cross-sectional area [86]. Forkhead box O (FoxO), one of the transcription factors, necessary for muscle atrophy in various pathological conditions including muscle disuse and cancer, is directly regulated by acetylation and deacetylation process. A recent report shows that lysine acetylation of FoxO prevents its nuclear localization and makes it inactive while deacetylation process activates FoxO in disuse of skeletal muscles. Data illustrates that TSA inactivates FoxO by inhibiting the HDAC activity, which results in attenuation of skeletal muscle atrophy and contractile dysfunction [82]. Furthermore, TSA treatments to C2C12 myotubes under nutrient deprived atrophy, results in repression of FoxO target genes including Lc3 (a marker of autophagy), atrogin1 and MuRF1 [82]. Similarly, TSA treatment in the dexamethasone induced atrophic mice regulates muscle mass wasting by inhibiting atrogin1 and MuRF1 levels in C2C12 myoblast cells [85]. TSA treatment leads to increased muscle size and function in normal and dystrophic mice by inducing the expression of follistatin, an antagonist of myostatin (a negative regulator of skeletal muscle development). But TSA treatment to tumor-bearing mice increased the expression level of follistatin without preserving or increasing skeletal muscle mass. These studies show that alteration of myostatin/follistatin axis is not sufficient to protect muscle mass specifically in cancer-induced cachexia [87–89]. Such findings point to the fact that the outcome of TSA treatment is not same under diverse clinical settings. In studies on mdx mice model, Colussi et al. (2008, 2009), reported the role of nitric oxide (NO) as an epigenetic regulator in duchenne muscular dystrophy (DMD) and explained that NO countered the progression of muscular dystrophy by blocking the HDAC. Moreover, like TSA impact on dystrophic mice, NO also enhance the follistatin level and increase the myofiber length in mdx mice. In other words, the data thus depict that both (NO and HDAC inhibitor) can block the common target in DMD i.e. HDAC and up-regulate the expression of follistatin [90,91]. Overall published data illustrate that TSA regulates the muscle mass and contractile function by inhibiting HDAC mediated FoxO activation in different muscle wasting conditions (Fig. 7). Due to contradictory findings, further studies are needed to confirm the use of HDAC blocker to regulate atrophy [81,82,85]. 2.2.3. PDE inhibitors Cyclic adenosine 3 ,5 -monophosphate (cAMP), acts as an intracellular second messenger in response to diverse extracellular signals including hormones, neurotransmitters, growth factors and regulates a wide range of biological processes such as cell proliferation, differentiation, growth through its two receptor proteins i.e. PKA (cAMP-dependent protein kinase A) and Epac (exchange protein directly activated by cAMP). Phosphodiesterase (PDE) blocks the cAMP-dependent signaling by cleaving the diester ring of cAMP and decreases its level. Therefore use of PDE inhibitors is quite promising in regulation of diverse pathophysiological conditions including skeletal muscle atrophy. 2.2.3.1. Torbafylline (HWA 448). Torbafylline is a xanthine derivative which acts as a PDE inhibitor [92]. During burn injury, increase in the level of PDE4 plays prominent role in skeletal muscle proteolysis through activation of Ub–proteasome dependent proteolytic pathway. Various studies have highlighted that in skeletal muscles inhibition of PDE by torbafylline stimulates cAMP production and activates Epac which further catalyzes the activation of G-proteins i.e. Rap (Ras family of small GTPases), by facilitating the replacement of guanosine diphosphate (GDP) with guanosine triphosphate (GTP). The activated Epac-Rap GTPase pathway increases the level of phosphorylated-Akt and stimulates the protein anabolic

7

Fig. 7. Schematic representation of TCS therapeutic action in denervation, muscle disuse, cancer, nutrient deprived induced atrophy.

pathways by activating the PI3K/Akt axis. This axis shows its positive effects on protein synthesis and inhibitory effects on protein degradation by elevating the level of phospho-GSK3␤ and phosphomTOR and decreasing the level of phospho-FoxO1 [13,93,94]. In addition, torbafylline treatment down-regulates mRNA expression of E3 ligases, cathepsin L, and calpain and regulates the proteolytic pathways in burn-induced injury. Besides this, torbafylline regulates muscle proteolysis by down-regulating the level of proinflammatory cytokines (IL-6 and TNF␣) probably by stimulating both (Epac and PKA) cAMP dependent receptor proteins [13,95,96]. In addition to injury induced atrophy, torbafylline attenuates inflammatory cytokine levels in a large number of muscle wasting models such as diabetes, cancer and sepsis [92,97,98]. These findings illustrate the anti-inflammatory properties of this compound. Furthermore, a torbafylline anti-atrophic effect has also been proved in denervation, casting or cancer induced cachexia models [92,97,99]. Overall it can be said that inhibition of PDE activity with torbafylline leads to stimulation of cAMP/Epac-Rap/PI3K/Akt pathway mediated anti-proteolytic effects in skeletal muscle atrophy (Fig. 8). 2.2.3.2. Pentoxifylline (PTX). Like torbafylline, PTX is another xanthine derivative that non-selectively inhibits PDE [92,99]. PTX is also known as suppressor of TNF␣ production [100,101] and this property makes it helpful in prevention of skeletal muscle wasting in cancer, sepsis, trauma, and AIDS models [92]. Published data show that PTX administration under diverse pathological conditions (diabetes, tumor, sepsis) in animal models stimulates cAMP formation and reduces the total rate of protein breakdown by down-regulating Ca2+ -dependent (calpain), Ca2+ -independent (cathepsin L) and proteasome (subunits of 20S and 26S) proteolytic system activities (Fig. 8) [98,101,102]. PTX seems to be effective in regulating the muscle wasting in diverse atrophic models while under septic condition torbafylline treatments attenuate proteasome activity with greater potency as compared to PTX [92]. Similar to these non-selective PDE inhibitors, selective inhibitors of PDE i.e.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548

549 550 551 552 553 554 555 556 557 558 559 560 561 562 563

G Model

ARTICLE IN PRESS

YPHRS 2837 1–16

V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

8

Fig. 8. Schematic representation of PDE inhibitors therapeutic action in tumor/diabetes/sepsis/burn induced atrophy.

565

rolipram and cilomilast (Ariflo) have also been reported to reduce muscle atrophy in denervation and casting animal models [103].

566

2.3. ˇ-Adrenoceptor agonists

564

567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582

583 584 585 586 587 588 589 590 591 592 593 594 595 596

Skeletal muscles are rich in ␤2-adrenoceptors (␤2-AR) which are G-protein coupled receptors. On activation, this coupled complex activates PKA and cAMP response element binding protein (CREB) through stimulation of adenylate cyclase [104]. PKA phosphorylates CREB leading to its activation and binding to the DNA and inducing the expression of the myogenic transcription factors (Myf5, MyoD and Pax3) which regulates the myogenesis program [105]. In addition, CREB also promotes satellite cell proliferation and skeletal muscle regeneration after muscle injury [106]. In other words, ␤2-AR is involved in the regulation of skeletal muscle proliferation and differentiation programs and these properties make this receptor signaling pathway a novel therapeutic target for controlling the skeletal muscle wasting [107,108]. Some ␤adrenoceptor agonists include formoterol, clenbuterol and others which are under investigation regarding their potential to regulate skeletal muscle atrophy. 2.3.1. Formoterol Formoterol is a ␤2-AR agonist with anti-cachectic property. This compound has shown its therapeutic potential by stimulating skeletal muscle growth/hypertrophy and body weight not only by down-regulating the muscle specific proteolytic systems including Ub–proteasome and caspase-3 dependent apoptosis but also by inducing the protein anabolic response (i.e. Akt-dependent pathway). In addition to skeletal muscle, this compound has shown its powerful protective action on cardiac muscle as well [109,110]. In COPD patients, skeletal muscle dysfunction is usually responsible for limitations on physical exercise. Formoterol treatment enhances skeletal muscle oxidative process by increasing the PGC1␣ and mtDNA content in their skeletal muscles. As a result, the exercise capacity of COPD patients is gradually improved [111,112].

In the tumor-bearing rats, formoterol administration significantly increased the level of follistatin and decreased the levels of the myostatin and its receptor (activin IIB, ActIIB) thereby regulating the loss of muscle mass [108,113,114]. Various studies have demonstrated that myostatin suppresses the anabolic pathways (IGF-1/PI3K/Akt axis) and activates the catabolic pathways (p38MAPK/ERK1/2, Wnt and FoxO1 signaling and Ub–proteasome system) while follistatin binds to myostatin and antagonizes its function [115–119]. Furthermore, formoterol administration in bupivacaine-treated rats ameliorates muscle wasting by increasing Pax7 and decreasing myogenin mRNA content which further stimulates satellite cell proliferation and favors muscle regeneration. In addition, formoterol can induce mitochondrial biogenesis to prevent dexamethasone induced atrophy [120,121]. Beneficial effect of formoterol was also observed under both cancer cachexia (Yoshida AH-130 ascites tumor bearing rat) and sarcopenic conditions. These studies suggest that ␤2-AR agonist treatment leads to reduced muscle wasting by regulating the loss of muscle mass [110,122].

2.3.2. Clenbuterol Clenbuterol is another ␤2-AR agonist which stimulates the cAMP level and suppresses the fasting-induced expression of atrogin1 and MuRF1 by inducing Akt/FoxO3 phosphorylation [104]. Clenbuterol administration under denervation induced atrophy, regulates the muscle wasting process through activation of protein anabolism and inhibition of cathepsin L and Ub ligases dependent proteolytic system. Studies have shown that these effects of clenbuterol are possibly mediated through cAMP/PKA signaling pathway without involving Akt [14]. Similar regulatory role of clenbuterol has also been demonstrated in other atrophic experimental animal models such as hind-limb suspension, immobilization, and spinal cord injury. As shown in Fig. 9(a) and (b), overall these drugs regulate muscle wasting through diverse mechanisms that include down-regulation of apoptosis, rate of protein catabolism,

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615

616 617 618 619 620 621 622 623 624 625 626 627 628 629 630

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

9

Fig. 9. (a) Schematic representation of ␤-agonist therapeutic action in tumor/dexamethasone/sarcopenic induced atrophy. (b) Schematic representation of ␤-agonist therapeutic action in fasting/denervation induced atrophy.

632

up-regulation of rate of protein anabolism, and muscle regeneration processes.

633

2.4. Anti-cytokines agents

631

634 635 636 637 638 639 640 641 642 643 644 645

646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661

662 663 664 665 666 667 668

Cytokines (such as TNF␣, TWEAK, IL-1, IL-6) are the major culprits in almost any kind of pathophysiological condition that leads to chronic inflammation and ultimately results in loss of skeletal muscle. Therefore, use of antibodies against such cytokines could be beneficial in regulating loss of skeletal muscle. There are many studies that have highlighted the beneficial effect of some of these antibodies such as anti-TNF␣, anti-IL-1 and anti-IL-6 on halting the muscle loss and restoring their function. Some of these antibodies are under Phase I/II/III trials (Table 1). More detailed information about other anti-cytokines except anti-TNF␣ that are beneficial in treating cancer cachexia can be found in the review article by Ma et al. [123]. 2.4.1. Anti-TNF˛ Studies have shown that administration of an anti-murine TNF IgG in rats bearing the Yoshida AH-130 ascites hepatoma down-regulates not only the level of circulatory TNF␣ and muscle ubiquitin gene expression, but also controls the protein degradation in skeletal muscles [124]. Likewise, injection of soluble-TNF receptor (sTNFR1, specific inhibitors of TNF␣), in monocratiline (MCT) treated Sprague–Dawley rats prevent the interaction of TNF␣ with its receptors and attenuates the MAFbx and ubiquitin transcription. This study illustrates that anti-TNF␣ treatment reduces the skeletal muscle wasting in cardiac cachexia and preserves the body mass [102]. Interestingly, Granado et al. reported the opposite effect of sTNFR1 on arthritic rat. They observed that administration of polyethylene glycol linked-sTNFRI to arthritic rats does not alter the gastrocnemius muscle mass and MuRF1/MAFbx gene expression [125]. 2.4.2. Thalidomide Thalidomide (a glutamic acid derivative) was first introduced in 1951 as a sedative compound and later as an anti-emetic. It was widely used by pregnant women for treating morning sickness and became notorious for birth defects. As a result thalidomide was pulled out of market. Later this compound gained intense interest because of its anti-TNF␣ activity. As a result Food and Drug

Administration (FDA) has approved this drug in 1998 for treatment of erythema nodosum leprosum (Leprosy reaction variant) [126,127]. Presently, there is renewed interest in thalidomide for its extensive array of actions such as anti-inflammatory, anti-angiogenic, immuno-modulator along with anti-emetic and sedative activities. Additionally, this compound has shown its anticachectic property in cancer patients by down-regulating the TNF␣ mRNA expression. Like other drugs mentioned above, thalidomide also prevents the nuclear translocation of NF␬B by inhibiting the activity of I␬B kinase and preserves the skeletal muscle mass in cachexia [28]. Besides this, thalidomide reduces the serum levels of IL-6 and CRP in the cancer-cachexia patients [128–130]. Liu et al. [131] reported that thalidomide can preserve the fast-twitch type skeletal muscle fibers in tumor induced cachexia by decreasing the expression of TNF␣ and transforming growth factor beta1 (TGF␤1) in the soleus muscle of cholangiocarcinoma rats. TGF␤1 is a negative regulator of muscle mass which inhibits Akt-dependent pathway (protein anabolic pathway) via Smad2/Smad3 and enhances the proteolysis of muscle specific proteins [132]. Kaplan and co-workers observed the anti-cachectic effect of thalidomide in AIDS associated muscle wasting patients. Such patients when treated with thalidomide (100 mg/day and 200 mg/day) showed a significant improvement in weight compared to the placebo treated group [133]. Similarly, another research group worked with pancreatic cancer cachexia patients and observed significant weight gain in thalidomide treated patients [19]. Tumor-associated macrophages express Cox2 that acts as a source for prostaglandins (PGE) production. These PGE impairs immune surveillance by inhibiting several T-cells and natural killer cell functions. Report shows that thalidomide and its derivatives inhibit the lipopolysaccharide (LPS) induced Cox2 and PGE2 synthesis in murine macrophages [134,135] and control the systemic inflammation. In addition to anti-inflammation and anti-cachectic activity, thalidomide treatment (Phase II trial) has also shown its impact on appetite in 64% of the patients who are in advanced stage of cancer [130]. Overall thalidomide inhibits the activity/expression of various pro-inflammatory cytokines and transcription factors such as TNF␣, TGF␤1, IL-6, CRP, Cox2 and NF␬B and shows its tremendous therapeutic potential by preventing the loss of muscle mass in tumor/cancer/AIDS induced cachexia (Fig. 10). Thalidomide also been reported to cause several side effects including peripheral neuropathy, dizziness, constipation, and rashes [132].

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

10

Table 1 Molecules effected by drugs used under diverse clinical settings. S. no

Name of drug

Model used for study

1. Natural compounds A.

Eicosapentanoic acid

Tumor/cancer/sepsis induced atrophy

B.

␤-Hydroxy-methyl butyrate (HMB)

PIF/cancer/ dexamethasone-induced atrophy

C.

Reserveratrol

COPD/diabetes/TNF␣/ dexamethasone-induced atrophy

D.

Ghrelin and des acyl ghrelin (appetite stimulant)

Cancer, COPD, CKD, unloading induced muscle wasting

Celecoxib (Cox2 inhibitor)

Cancer and arthritis induced cachexia Cancer and arthritis induced cachexia

2. Enzyme inhibitors A. Cox2 inhibitor I. II.

B. Histone deacetylase inhibitor I.

C. PDE inhibitor I.

II.

3. ␤-Adrenoceptor agonists A.

B.

4. Anti-cytokine agents A.

Meloxicam (Cox2 inhibitor)

Molecules affected Inhibits: IL-1, TNF␣, IL-6, NF␬B, 15LOX, caspase-8, FoxO Increases: p-Akt, PGC-1␣/PPAR␥ Inhibits: FoxO, MuRF1 Ub–proteasome (regulatory subunits of 19S and 20S) Increases: eIF4G:eIF4E (translation initiation complex), PKR, mTOR and p70S6K Inhibits: TNF␣, IL-1␤, IL-6, NF␬B, MuRF1 Increases: Akt, mTOR, p70S6K, 4E-BP1 AMPK and SIRT1, Inhibits: NF␬B, FoxO1, MuRF1, atrogin1, TNF␣, IL-1␤, and IL-6 Increases: growth hormone, IGF-1, IGFBP-3, neuropeptide Y

Phase II trial completed

Inhibits: Cox2, TNF␣, IL-6, NF␬B, PGE2 Inhibits: Cox2, TNF␣, IGFBP-5, atrogin-1, MuRF1 Increases: GH-IGF1 axis

Trichostatin A (HDAC inhibitor)

Denervation, muscle disuse, cancer, nutrient deprived induced atrophy

Inhibits: class I and II histone deacetylases, FoxO, atrogin-1/MAFbx, MuRF1 and Lc3, myogenin Increases: Dach2 (negative myogenin regulator),

Torbafylline (PDE inhibitor)

Tumor/diabetes/sepsis/burn induced muscle wasting

Pentoxifylline (PDE inhibitor)

Tumor/diabetes/sepsis/burn induced muscle wasting

Inhibits: PDE, IL-6, TNF␣, ubiquitin, E3-ligase, Cathepsin L and Calpain Increases: cAMP, Akt Inhibits: TNF␣, cathepsin L, Calpain, 20S and 26S proteasome

Formoterol

Tumor/dexamethasone/ sarcopenic induced atrophy

Clenbuterol

Denervation and fasting induced atrophy

Anti-TNF

Rats bearing the Yoshida AH-130 ascites hepatoma Non-small cell lung cancer (NSCLC)-related anemia and cachexia Cancer cachexia

B.

ALD518 (anti-IL6)

C.

E.

MABp1 (anti-IL-1␣) IP-1510 (IL-1 receptor antagonist) sTNFR1

F.

Thalidomide

D.

Trial phase

Cancer cachexia Monocratiline (MCT) treated Sprague-Dawley rats Tumor/cancer/AIDS induced cachexia

Inhibits: myostatin, caspase-3, Wnt, FoxO1, atrogin, MuRF1 Increases: follistatin, IGF1/PI3K/Akt, Pax7 Inhibits: FoxO3, atrogin, MuRF1, cathepsin L Increases: cAMP, phospho-Akt, PGC-1␣

TNF␣, ubiquitin and MAFbx Phase I and II completed

IL-6

Phase I complete, Phase III under trial Phase I and II complete

IL-1␣ IL-1 receptor antagonist TNF␣, MAFbx/MuRF1, ubiquitin

Phase II trial

Inhibits: TNF␣, IL-6, Cox2, PGE2, NF␬B, TGF␤1 Increases: Akt

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

G Model

ARTICLE IN PRESS

YPHRS 2837 1–16

V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

11

Table 1 (Continued) S. no

Name of drug

Model used for study

Trial phase

5. Other investigational drugs A. B.

Ostarine GLPG0492

Cancer cachexia Hindlimb immobilization, DMD Cancer cachexia Cancer cachexia

Phase II trial

Healthy adults individuals

Phase II under trial

Sporadic inclusion body myositis Cancer cachexia

Phase II, III under trial

C. D.

OHR/AVR118 APD209

E. F.

REGN1033 (anti-myostatin) BYM338

G.

ACVR2B

6. Miscellaneous agents A.

712

713 714 715 716 717 718 719 720 721 722 723 724 725

726 727 728

Megesterol acetate (MA)

Tumor/cancer/AIDS cachexia

2.5. Other investigational drugs 2.5.1. Enobosarm Enobosarm (also called ostarine, GTx-024) is a non-steroidal, orally bio-available and a selective androgen receptor modulator (SARM). It has shown a dose-dependent improvement in total lean body mass and physical function in healthy elderly men and postmenopausal women. Besides this, enobosarm is also useful in the prevention of muscle wasting associated with cancer cachexia. Data from Phase II clinical trial where cancer patients were administrated with ostartine showed improvement in muscle performance (measured by stair climb) and insulin resistance without any visible side effect and toxicity. Hence enobosarm possesses a strong efficacy and safety in cancer cachexia conditions to regulate muscle loss [136–138]. 2.5.2. GLPG0492 GLPG0492 (galapagos) is another non-steroidal SARM that has shown its efficacy on muscle by increasing muscle fiber size and

Fig. 10. Schematic representation tumor/cancer/AIDS cachexia.

of

thalidomide

therapeutic

action

in

Phase IIb trial Phase II pilot trial

Molecules affected

Inhibits TNF␣ and IL-6. ␤-Adrenoceptor agonist, act like megesterol and formoterol Myostatin (MSTN) Blocking the activin type II B receptor Inhibit signaling of MSTN/activin

Inhibits: IL-1, IL-6, and TNF␣, Ub–proteasome system, E2 ligase and atrogin-1 Increases: neuropeptide Y secretion

skeletal muscle function in the hindlimb of immobilized mice and in duchenne muscular dystrophy (DMD) patients respectively. This drug is in preclinical trial to treat DMD [139,140]. 2.5.3. OHR/AVR118 It is a broad-spectrum peptide-nucleic acid immune-modulator with anti-inflammatory activity that targets both cellular proinflammatory chemokine and cytokine synthesis (such as TNF␣ and IL-6). A Phase II trial of this drug on patients with advanced cancer and cachexia achieved stabilization of body weight, body fat and muscle mass with a significant increase in appetite without showing any adverse effect. This study shows that OHR/AVR118 drug diminishes the harmful effects of such cytokines/chemokines, whose activation has a direct effect on muscle metabolism. Besides cancer cachexia patients, this drug has also been used for treatment of AIDS cachexia patients, where it helped in alleviating multiple symptoms of the disease [141,142]. 2.5.4. APD209 APD209 is an oral, fixed-dose combination of megestrol (a progestin which is used off-label in cachexia) and formoterol (a selective ␤-adrenoceptor agonist) for use in treatment of cancer cachexia. A Phase IIa trial study with this drug on patients with cancer-related anorexia and cachexia syndrome (CACS) demonstrated improved muscle size and strength [143,144]. Hence it may be a potential new drug for treating cachexia in the future. 2.5.5. Anti-myostatin/activin Available evidences suggest that members of the TGF-␤ superfamily such as myostatin (MSTN) and activin A are powerful catabolic stimuli that inhibit muscle growth and promote muscle protein loss in various disease states [145,146]. Both these ligands bind initially with activin type II receptor (ActRIIB and ActRIIA) and then with activin type I receptor (activin-like kinase; ALK-4 or ALK5) to activate cytosolic SMAD2/3 proteins which leads to proteolysis and cause muscle fiber atrophy. It has been reported that inhibition of the myostatin ameliorates the dystrophic phenotype in the mdx mouse model, sarcopenia in an aging mouse model and muscle wasting in tumor-bearing mice [88,147–149]. Bimagrumab (BYM338) and REGN1033 are two human monocloncal antibodies against myostatin. The treatment with BYM338 enhances differentiation of primary human skeletal myoblasts and increases skeletal muscle mass in mice by blocking the activin type II B receptors (ActRIIB). However REGN1033 administration prevents loss of muscle mass in dexamethasone or immobilization

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

729 730 731

732 733 734 735 736 737 738 739 740 741 742 743 744

745 746 747 748 749 750 751 752

753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770

G Model YPHRS 2837 1–16 12

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

within 2 weeks. This soluble receptor is also very effective in preventing cardiac muscle atrophy in tumor-bearing mice [88,145]. During a Phase II clinical trial of ACE031 with DMD patients and healthy volunteers, some participants experienced gum bleeding, nosebleeds as well as dilation in skin blood vessels which led to discontinuation of the trials. Overall BYM338, REGN1033 and soluble ActRIIB/Fc are promising drugs that have the ability to regulate muscle wasting/cachexia by blocking MSTN/activin signaling under diverse pathological conditions including cancer and may further help in improving the rate of mortality and morbidity.

2.6. Miscellaneous agents

Fig. 11. Schematic representation of megesterol acetate therapeutic action in tumor/cancer/AIDS cachexia.

771 772 773 774 775 776 777 778 779 780 781 782 783

induced muscle wasting by directly antagonizing the myostatin [123]. Presently, BYM338 is under Phase II/III trial to treat sporadic inclusion body myositis patients (a rare life-threatening musclewasting condition) while REGN1033 is in Phase II to treat patients with sarcopenia [150,151]. Though various endogenous regulators including follistatin usually blocks MSTN signaling and maintains the muscle mass, some studies have suggested that even increase in the level of follistatin is ineffective in preserving muscle in cancer cachexia [87]. However, systemic administration of a soluble form of the activin type IIB receptor (ActRIIB/Fc; ACVR2B-ACE031) in mice bearing cancer cachexia can inhibit MSTN signaling and its related ligands like activin and significantly increases muscle growth (40–60%)

2.6.1. Megestrol acetate In 1993, FDA approved megestrol acetate (MA) for the treatment of cancer and AIDS induced cachexia. More than 15 clinical trials have demonstrated that at 160–1600 mg/day, this drug significantly improves appetite and the lean body mass. This drug is being used either alone or as a supplement along with meloxicam for cachectic cancer patients where it has shown positive effects in regulating the loss of body mass [23,152,153]. MA is used to improve appetite and weight gain in AIDS/cancer induced anorexia–cachexia syndrome [154–156]. Though the mechanism of stimulation of appetite/increased body mass is not clear, studies have shown the involvement of neuropeptide Y and suppression of pro-inflammatory cytokines such as IL-1, IL-6, and TNF␣ [23,157,158]. These studies have shown that in MA-treated rats, the food and water intake significantly increased when compared to untreated group. The levels of NY in the lateral hypothalamic area increased indicating that NY may be responsible for this orexigenic (appetite-stimulating) effect. Another study have shown that administration of MA in tumor (Yoshida AH-130 ascites hepatoma)-bearing rats results in reversal of muscle wasting process by decreasing the mRNA level of ubiquitin, E2 and atrogin1 which are the key biomarkers of Ub–proteasome proteolytic system [159]. These studies suggest that MA, either alone or as a supplement with meloxicam, can be used to stimulate appetite in cancer/AIDS induced cachectic patients. It causes down-regulation of inflammation mediated molecules such as IL-1, IL-6, TNF␣, ubiquitin, E2, atrogin1 and up-regulation of NY level. This leads to increased appetite, body mass, and decreased muscle protein degradation

Fig. 12. Drugs and their overall inhibitory effect on skeletal muscle specific proteolytic system.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

784 785 786 787 788 789 790 791 792 793

794

795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

829

by suppressing the Ub–proteasome mediated proteolytic system (Fig. 11). MA treatment is also associated with some adverse effects. If the treatment is discontinued abruptly it can lead to thromboembolic phenomena, peripheral edema, hyperglycemia, hypertension, adrenal suppression and adrenal insufficiency [22].

830

3. Conclusions

824 825 826 827 828

849

Up-regulation of skeletal muscle protein breakdown is hallmark of atrophy and as a result all potential drugs target the proteolytic systems to cure or prevent the skeletal muscle atrophy. As shown in Table 1 and Fig. 12, all the drugs have displayed positive effects in diverse atrophic models either by inhibiting particular molecules/cytokines/proteolytic systems involved in protein catabolic pathway or by improving the satellite cell function during muscle injury and aging. In spite of this, except MA, no other drug has yet been recommended by FDA to prevent or treat muscle atrophy. This underlines the fact that the reported drugs are not efficiently targeting every proteolytic system. It can be safely inferred that skeletal muscle atrophy being a multi-factorial syndrome, needs a multi-targeted approach to yield success. There are reports in literature regarding use of some non-pharmacological therapies (such as nutritional supplement and rehabilitation) to delay the onset of disease and ease its symptoms at least up to some extent and improve the quality of life [160]. Thus there is the need for combinational treatment and developing a novel approach to treat skeletal muscle wasting.

850

Conflict of interest

831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848

852

All authors (VD, SG, RD, EI, AM) declare that they have no conflicts of interest involving this work.

853

Acknowledgments

851

Q7 854 855 856

857

858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887

This work is fully supported by UGC-MRP grant (F. 411266/2012 (SR)) and ICMR-BMS grant (IRIS Cell No. 2012-2656), India. References [1] D.J. Glass, Skeletal muscle hypertrophy and atrophy signalling pathways, Int. J. Biochem. Cell Biol. 37 (2005) 1974–1984. [2] E.E. Dupont-Versteegden, Apoptosis in muscle atrophy: relevance to sarcopenia, Exp. Gerontol. 40 (2005) 473–481. [3] P. Zhang, X. Chen, M. Fan, Signaling mechanisms involved in disuse muscle atrophy, Med. Hypotheses 69 (2007) 310–321. [4] A.J. Murton, D. Constantin, P.L. Greenhaff, The involvement of the ubiquitin proteasome system in human skeletal muscle remodelling and trophy, Biochim. Biophys. Acta 1782 (2008) 730–743. [5] J.J. McCarthy, K.A. Esser, C.A. Peterson, E.E. Dupont-Versteegden, Evidence of MyomiR network regulation of beta-myosin heavy chain gene expression during skeletal muscle atrophy, Physiol. Genomics 39 (2009) 219–226. [6] A. Kumar, S. Bhatnagar, P.K. Paul, TWEAK and TRAF6 regulate skeletal muscle atrophy, Curr. Opin. Clin. Nutr. Metab. Care 15 (2012) 233–239. [7] A. Mittal, S. Bhatnagar, A. Kumar, E. Lach-Trifilieff, S. Wauters, H. Li, D.Y. Makonchuk, D.J. Glass, A. Kumar, The TWEAK-Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice, J. Cell Biol. 188 (2010) 833–849. [8] H. Li, S. Malhotra, A. Kumar, Nuclear factor-kappa B signaling in skeletal muscle atrophy, J. Mol. Med. (Berl.) 86 (2008) 1113–1126. [9] M. Llovera, N. Carbó, J. López-Soriano, C. García-Martínez, S. Busquets, B. Alvarez, N. Agell, P. Costelli, F.J. Lopez-Soriano, A. Celada, J.M. Argiles, Different cytokines modulate ubiquitin gene expression in rat skeletal muscle, Cancer Lett. 133 (1) (1998) 83–87. [10] R.A. Frost, C.H. Lang, Protein kinase B/Akt: a nexus of growth factor and cytokine signalling in determining muscle mass, J. Appl. Physiol. 103 (2007) 378–387. [11] A. Bonetto, F. Penna, M. Muscaritoli, V.G. Minero, F. Rossi Fanelli, F.M. Baccino, et al., Are antioxidants useful for treating skeletal muscle atrophy? Free Radic. Biol. Med. 47 (2009) 906–916.

13

[12] P. Bonaldo, M. Sandri, Cellular and molecular mechanisms of muscle atrophy, Dis. Model. Mech. 6 (2013) 25–39. [13] R. Joshi, N. Kadeer, S. Sheriff, L.A. Friend, J.H. James, A. Balasubramaniam, Phosphodiesterase (PDE) inhibitor torbafylline (HWA 448) attenuates burn-induced rat skeletal muscle proteolysis through the PDE4/cAMP/EPAC/PI3K/Akt pathway, Mol. Cell Endocrinol. 393 (2014) 152–163. [14] D.A. Goncalves, W.A. Silveira, E.C. Lira, F.A. Grac¸a, S. Paula-Gomes, N.M. Zanon, I.C. Kettelhut, L.C. Navegantes, Clenbuterol suppresses proteasomal and lysosomal proteolysis and atrophy-related genes in denervated rat soleus muscles independently of Akt, Am. J. Physiol. Endocrinol. Metab. 302 (2012) E123–E133. [15] S.K. Joshi, H.T. Kim, B.T. Feeley, X. Liu, Differential ubiquitin–proteasome and autophagy signaling following rotator cuff tears and suprascapular nerve injury, J. Orthop. Res. 32 (2014) 138–144. [16] E.M. Duncan, T.L. Muratore-Schroeder, R.G. Cook, B.A. Garcia, J. Shabanowitz, D.F. Hunt, et al., Cathepsin L proteolytically processes histone H3 during mouse embryonic stem cell differentiation, Cell 135 (2008) 284–294. [17] H. Takahashi, K. Ishidoh, D. Muno, A. Ohwada, T. Nukiwa, E. Kominami, S. Kira, Cathepsin L activity is increased in alveolar macrophages and bronchoalveolar lavage fluid of smokers, Am. Rev. Respir. Dis. 147 (1993) 1562–1568. [18] R.B. Hunter, E. Stevenson, A. Koncarevic, H. Mitchell-Felton, D.A. Essig, S.C. Kandarian, Activation of an alternative NF-kappaB pathway in skeletal muscle during disuse atrophy, FASEB J. 16 (2002) 529–538. [19] J.N. Gordon, T.M. Trebble, R.D. Ellis, H.D. Duncan, T. Johns, P.M. Goggin, Thalidomide in the treatment of cancer cachexia: a randomised placebo controlled trial, Gut 54 (2005) 540–545. [20] L.G. Melstrom, K.A. Melstrom Jr., X.Z. Ding, T.E. Adrian, Mechanisms of skeletal muscle degradation and its therapy in cancer cachexia, Histol. Histopathol. 22 (2007) 805–814. [21] V.C. Vaughan, M. Sullivan-Gunn, E. Hinch, P. Martin, P.A. Lewandowski, Eicosapentaenoic acid and oxypurinol in the treatment of muscle wasting in a mouse model of cancer cachexia, PLoS ONE 7 (2012) e45900. [22] I. Ronga, F. Gallucci, F. Riccardi, G. Uomo, Anorexia–cachexia syndrome in pancreatic cancer: recent advances and new pharmacological approach, Adv. Med. Sci. 59 (2014) 1–6. [23] C.R. Tan, P.M. Yaffee, L.H. Jamil, S.K. Lo, N. Nissen, S.J. Pandol, et al., Pancreatic cancer cachexia: a review of mechanisms and therapeutics, Front. Physiol. 5 (2014) 88. [24] P. Magee, S. Pearson, J. Allen, The omega-3 fatty acid, eicosapentaenoic acid (EPA), prevents the damaging effects of tumour necrosis factor (TNF)-alpha during murine skeletal muscle cell differentiation, Lipids Health Dis. 7 (2008) 24. [25] Y. Liu, F. Chen, J. Odle, X. Lin, H. Zhu, H. Shi, Y. Hou, J. Yin, Fish oil increases muscle protein mass and modulates Akt/FOXO, TLR4, and NOD signaling in weanling piglets after lipopolysaccharide challenge, J. Nutr. 143 (2013) 1331–1339. [26] A.S. Whitehouse, H.J. Smith, J.L. Drake, M.J. Tisdale, Mechanism of attenuation of skeletal muscle protein catabolism in cancer cachexia by eicosapentaenoic acid, Cancer Res. 61 (2001) 3604–3609. [27] M.J. Tisdale, Pathogenesis of cancer cachexia, J. Support Oncol. 1 (2003) 159–168. [28] M.J. Tisdale, The ubiquitin–proteasome pathway as a therapeutic target for muscle wasting, J. Support. Oncol. 3 (2005) 209–217. [29] H.C. Olguin, B.B. Olwin, Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self-renewal, Dev. Biol. 275 (2004) 375–388. [30] E. Berardi, P. Aulino, I. Murfuni, A. Toschi, F. Padula, B.M. Scicchitano, Skeletal muscle is enriched in hematopoietic stem cells and not inflammatory cells in cachectic mice, Neurol. Res. 30 (2008) 160–169. [31] M. Tian, K.L. Kliewer, M.L. Asp, M.B. Stout, M.A. Belury, c9t11-Conjugated linoleic acid-rich oil fails to attenuate wasting in colon-26 tumor-induced late-stage cancer cachexia in male CD2F1 mice, Mol. Nutr. Food Res. 55 (2011) 268–277. [32] F. Penna, S. Busquets, F. Pin, M. Toledo, F.M. Baccino, F.J. López-Soriano, P. Costelli, J.M. Argiles, Combined approach to counteract experimental cancer cachexia: eicosapentaenoic acid and training exercise, J. Cachexia Sarcopenia Muscle 2 (2011) 95–104. [33] M. Li, Q. Zhu, C. Hu, J.P. Giesy, Z. Kong, Protective effects of eicosapentaenoic acid on genotoxicity and oxidative stress of cyclophosphamide in mice, Environ. Toxicol. 26 (2011) 217–223. [34] J. Khal, M.J. Tisdale, Downregulation of muscle protein degradation in sepsis by eicosapentaenoic acid (EPA), Biochem. Biophys. Res. Commun. 375 (2008) 238–240. [35] J. Singh, N.K. Verma, S.M. Kansagra, B.N. Kate, C.S. Dey, Altered PPARgamma expression inhibits myogenic differentiation in C2C12 skeletal muscle cells, Mol. Cell Biochem. 294 (2007) 163–171. [36] A.F. Mauricio, E. Minatel, H.S. Neto, M.J. Marques, Effects of fish oil containing eicosapentaenoic acid and docosahexaenoic acid on dystrophic mdx mice, Clin. Nutr. 32 (2013) 636–642. [37] A. Jatoi, K. Rowland, C.L. Loprinzi, J.A. Sloan, S.R. Dakhil, N. MacDonald, B. Gagnon, P.J. Novotny, J.A. Mailliard, T.I. Bushey, S. Nair, B. Christensen, North Central Cancer Treatment G, An eicosapentaenoic acid supplement versus megestrol acetate versus both for patients with cancer-associated wasting: a North Central Cancer Treatment Group and National Cancer Institute of Canada collaborative effort, J. Clin. Oncol. 22 (2004) 2469–2476.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973

G Model YPHRS 2837 1–16 14 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

[38] K.C. Fearon, M.D. Barber, A.G. Moses, S.H. Ahmedzai, G.S. Taylor, M.J. Tisdale, G.D. Murray, Double-blind, placebo-controlled, randomized study of eicosapentaenoic acid diester in patients with cancer cachexia, J. Clin. Oncol. 24 (2006) 3401–3407. [39] R.A. Murphy, E. Yeung, V.C. Mazurak, M. Mourtzakis, Influence of eicosapentaenoic acid supplementation on lean body mass in cancer cachexia, Br. J. Cancer 105 (2011) 1469–1473. [40] H.J. Smith, P. Mukerji, M.J. Tisdale, Attenuation of proteasome-induced proteolysis in skeletal muscle by {beta}-hydroxy-{beta}-methylbutyrate in cancer-induced muscle loss, Cancer Res. 65 (2005) 277–283. [41] H.L. Eley, S.T. Russell, J.H. Baxter, P. Mukerji, M.J. Tisdale, Signaling pathways initiated by beta-hydroxy-beta-methylbutyrate to attenuate the depression of protein synthesis in skeletal muscle in response to cachectic stimuli, Am. J. Physiol. Endocrinol. Metab. 293 (2007) E923–E931. [42] K.K. Noh, K.W. Chung, Y.J. Choi, M.H. Park, E.J. Jang, C.H. Park, C. Yoon, N.D. Kim, M.K. Kim, H.Y. Chung, ␤-Hydroxy ␤-methylbutyrate improves dexamethasone-induced muscle atrophy by modulating the muscle degradation pathway in SD rat, PLoS ONE 9 (2014) e102947. [43] A. Rahman, K. Wilund, P.J. Fitschen, K. Jeejeebhoy, R. Agarwala, J.W. Drover, et al., Elderly persons with ICU-acquired weakness: the potential role for ␤-hydroxy-␤-methylbutyrate (HMB) supplementation? J. Parenter. Enteral Nutr. 38 (2013) 567–575. [44] P.E. May, A. Barber, J.T. D’Olimpio, A. Hourihane, N.N. Abumrad, Reversal of cancer-related wasting using oral supplementation with a combination of beta-hydroxy-beta-methylbutyrate, arginine, and glutamine, Am. J. Surg. 183 (2002) 471–479. [45] M.D. Girón, J.D. Vílchez, S. Shreeram, R. Salto, M. Manzano, E. Cabrera, et al., ␤-Hydroxy-␤-methylbutyrate (HMB) normalizes dexamethasone-induced autophagy-lysosomal pathway in skeletal muscle, PLoS ONE 10 (2015) e011752. [46] S.E. Alway, S.L. Pereira, N.K. Edens, Y. Hao, B.T. Bennett, ␤-Hydroxy-␤methylbutyrate (HMB) enhances the proliferation of satellite cells in fast muscles of aged rats during recovery from disuse atrophy, Exp. Gerontol. 48 (2013) 973–984. [47] T.K. Sin, B.Y. Yung, P.M. Siu, Modulation of SIRT1-Foxo1 signaling axis by resveratrol: implications in skeletal muscle aging and insulin resistance, Cell Physiol. Biochem. 35 (2015) 541–552. [48] N. Alamdari, Z. Aversa, E. Castillero, A. Gurav, V. Petkova, S. Tizio, et al., Resveratrol prevents dexamethasone-induced expression of the muscle atrophy-related ubiquitin ligases atrogin-1 and MuRF1 in cultured myotubes through a SIRT1-dependent mechanism, Biochem. Biophys. Res. Commun. 417 (2012) 528–533. [49] D.T. Wang, Y. Yin, Y.J. Yang, P.J. Lv, Y. Shi, L. Lu, L.B. Wei, Resveratrol prevents TNF-␣-induced muscle atrophy via regulation of Akt/mTOR/FoxO1 signaling in C2C12 myotubes, Int. Immunopharmacol. 19 (2014) 206–213. [50] S. Busquets, G. Fuste, E. Ametller, M. Olivan, M. Figueras, P. Costelli, et al., Resveratrol does not ameliorate muscle wasting in different types of cancer cachexia models, Clin. Nutr. 26 (2007) 239–244. [51] L.D. Williams, G.A. Burdock, J.A. Edwards, M. Beck, J. Bausch, Safety studies conducted on high-purity trans-resveratrol in experimental animals, Food Chem. Toxicol. 47 (2009) 2170–2182. [52] K.H. Chen, M.L. Cheng, Y.H. Jing, D.T. Chiu, M.S. Shiao, J.K. Chen, Resveratrol ameliorates metabolic disorders and muscle wasting in streptozotocininduced diabetic rats, Am. J. Physiol. Endocrinol. Metab. 301 (2011) E853–E863. [53] I. Momken, L. Stevens, A. Bergouignan, D. Desplanches, F. Rudwill, I. Chery, et al., Resveratrol prevents the wasting disorders of mechanical unloading by acting as a physical exercise mimetic in the rat, FASEB J. 25 (2011) 3646–3660. [54] S.M. Wyke, S.T. Russell, M.J. Tisdale, Induction of proteasome expression in skeletal muscle is attenuated by inhibitors of NF-kappaB activation, Br. J. Cancer 91 (2004) 1742–1750. [55] S. Shadfar, M.E. Couch, K.A. McKinney, L.J. Weinstein, X. Yin, J.E. Rodríguez, et al., Oral resveratrol therapy inhibits cancer-induced skeletal muscle and cardiac atrophy in vivo, Nutr. Cancer 63 (2011) 749–762. [56] Y. Qi, J.Y. Shang, L.J. Ma, B.B. Sun, X.G. Hu, B. Liu, et al., Inhibition of AMPK expression in skeletal muscle by systemic inflammation in COPD rats, Respir. Res. 15 (2014) 156. [57] D.J. Boocock, G.E. Faust, K.R. Pate, A.M. Schinas, V.A. Brown, M.P. Ducharme, et al., Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent, Cancer Epidemiol. Biomarkers Prev. 16 (2007) 1246–1252. [58] F. Visioli, The resveratrol fiasco, Pharmacol. Res. 90 (2014) 87. [59] N. Nagaya, T. Itoh, S. Murakami, H. Oya, M. Uematsu, K. Miyatake, et al., Treatment of cachexia with ghrelin in patients with COPD, Chest 128 (2005) 1187–1193. [60] R. Barazzoni, X. Zhu, M. Deboer, R. Datta, M.D. Culler, M. Zanetti, et al., Combined effects of ghrelin and higher food intake enhance skeletal muscle mitochondrial oxidative capacity and AKT phosphorylation in rats with chronic kidney disease, Kidney Int. 77 (2010) 23–28. [61] M. Nakazato, N. Murakami, Y. Date, A role for ghrelin in the central regulation of feeding, Nature 409 (2001) 194–198. [62] S. Reano, A. Graziani, N. Filigheddu, Acylated and unacylated ghrelin administration to blunt muscle wasting, Curr. Opin. Clin. Nutr. Metab. Care 17 (2014) 236–240. [63] S. Sheriff, N. Kadeer, R. Joshi, L.A. Friend, J.H. James, A. Balasubramaniam, Des-acyl ghrelin exhibits pro-anabolic and anti-catabolic effects on C2C12

[64]

[65]

[66]

[67]

[68]

[69]

[70]

[71] [72]

[73]

[74]

[75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

[85]

[86]

[87]

myotubes exposed to cytokines and reduces burn-induced muscle proteolysis in rats, Mol. Cell Endocrinol. 351 (2012) 286–295. H. Tsubouchi, S. Yanagi, A. Miura, N. Matsumoto, K. Kangawa, M. Nakazato, Ghrelin relieves cancer cachexia associated with the development of lung adenocarcinoma in mice, Eur. J. Pharmacol. 743 (2014) 1–10. N. Nagaya, J. Moriya, Y. Yasumura, M. Uematsu, F. Ono, W. Shimizu, et al., Effects of ghrelin administration on left ventricular function, exercise capacity, and muscle wasting in patients with chronic heart failure, Circulation 110 (2004) 3674–3679. K. Koshinaka, K. Toshinai, A. Mohammad, K. Noma, M. Oshikawa, H. Ueno, et al., Therapeutic potential of ghrelin treatment for unloading-induced muscle atrophy in mice, Biochem. Biophys. Res. Commun. 412 (2011) 296–301. F. Strasser, T.A. Lutz, M.T. Maeder, Safety, tolerability and pharmacokinetics of intravenous ghrelin for cancer-related anorexia/cachexia: a randomised, placebo-controlled, doubleblind, double-crossover study, Br. J. Cancer 98 (2008) 300–308. T. Akamizu, K. Takaya, T. Irako, Pharmacokinetics, safety, and endocrine and appetite effects of ghrelin administration in young healthy subjects, Eur. J. Endocrinol. 150 (2004) 447–455. J.M. Garcia, J. Friend, S. Allen, Therapeutic potential of anamorelin, a novel, oral ghrelin mimetic, in patients with cancer-related cachexia: a multicenter, randomized, double-blind, crossover, pilot study, Support. Care Cancer 21 (2013) 129–137. C. Pietra, Y. Takeda, N. Tazawa-Ogata, M. Minami, X. Yuanfeng, E.M. Duus, R. Northrup, Anamorelin HCl (ONO-7643), a novel ghrelin receptor agonist, for the treatment of cancer anorexia–cachexia syndrome: preclinical profile, J. Cachexia Sarcopenia Muscle 5 (2014) 329–337. D.C. Currow, A.P. Abernethy, Anamorelin hydrochloride in the treatment of cancer anorexia–cachexia syndrome, Future Oncol. 10 (2014) 789–802. B.A. Bondesen, S.T. Mills, K.M. Kegley, G.K. Pavlath, The COX-2 pathway is essential during early stages of skeletal muscle regeneration, Am. J. Physiol. Cell Physiol. 287 (2004) C475–C483. T.W. Davis, B.S. Zweifel, J.M. O’Neal, D.M. Heuvelman, A.L. Abegg, T.O. Hendrich, et al., Inhibition of cyclooxygenase-2 by celecoxib reverses tumorinduced wasting, J. Pharmacol. Exp. Ther. 308 (2004) 929–934. A.J. Baumgarten, H.H. Fiebig, A.M. Burger, Molecular analysis of xenograft models of human cancer cachexia—possibilities for therapeutic intervention, Cancer Genomics Proteomics 4 (2007) 223–231. A.I. Martin, M.P. Nieto-Bona, E. Castillero, C. Fernandez-Galaz, M. LopezMenduina, A.B. Gomez-Sanmiguel, et al., Effect of cyclooxygenase-2 inhibition by meloxicam, on atrogin-1 and myogenic regulatory factors in skeletal muscle of rats injected with endotoxin, J. Physiol. Pharmacol. 63 (2012) 649–659. G. Mantovani, A. Macció, C. Madeddu, R. Serpe, G. Antoni, E. Massa, Phase II nonrandomized study of the efficacy and safety of COX-2 inhibitor celecoxib on patients with cancer cachexia, J. Mol. Med. 88 (2010) 85–92. H.J. Hussey, M.J. Tisdale, Effect of the specific cyclooxygenase-2 inhibitor meloxicam on tumour growth and cachexia in a murine model, Int. J. Cancer 87 (2000) 95–100. V. Lai, J. George, L. Richey, H.J. Kim, T. Cannon, C. Shores, M. Couch, Results of a pilot study of the effects of celecoxib on cancer cachexia in patients with cancer of the head, neck, and gastrointestinal tract, Head Neck 30 (2008) 67–74. F.I. Romero, M.J. Martínez-Calatrava, O. Sánchez-Pernaute, O. Gualillo, R. Largo, G. Herrero-Beaumont, Pharmacological modulation by celecoxib of cachexia associated with experimental arthritis and atherosclerosis in rabbits, Br. J. Pharmacol. 161 (2010) 1012–1022. M. Granado, A.I. Martín, M.A. Villanúa, A. López-Calderón, Experimental arthritis inhibits the insulin-like growth factor-I axis and induces muscle wasting through cyclooxygenase-2 activation, Am. J. Physiol. Endocrinol. Metab. 292 (2007) E1656–E1665. S.M. Senf, P.B. Sandesara, S.A. Reed, A.R. Judge, p300 acetyltransferase activity differentially regulates the localization and activity of the FOXO homologues in skeletal muscle, Am. J. Physiol. Cell Physiol. 300 (2011) C1490–C1501. A.W. Beharry, P.B. Sandesara, B.M. Roberts, L.F. Ferreira, S.M. Senf, A.R. Judge, HDAC1 activates FoxO and is both sufficient and required for skeletal muscle atrophy, J. Cell Sci. 127 (Pt 7) (2014) 1441–1453. H. Tang, D. Goldman, Activity-dependent gene regulation in skeletal muscle is mediated by a histone deacetylase (HDAC)-Dach2-myogenin signal transduction cascade, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 16977–16982. H. Tang, P. Macpherson, M. Marvin, E. Meadows, W.H. Klein, X.J. Yang, et al., A histone deacetylase 4/myogenin positive feedback loop coordinates denervation-dependent gene induction and suppression, Mol. Biol. Cell 20 (2009) 1120–1131. K.V. Bricceno, P.J. Sampognaro, J.P. Van Meerbeke, C.J. Sumner, H. Fischbeck, B.G. Burnett, Histone deacetylase inhibition suppresses myogenin-dependent atrogene activation in spinal muscular atrophy mice, Hum. Mol. Genet. 21 (2012) 4448–4459. A.M. Avila, B.G. Burnett, A.A. Taye, F. Gabanella, M.A. Knight, P. Hartenstein, et al., Trichostatin A increases SMN expression and survival in a mouse model of spinal muscular atrophy, J. Clin. Invest. 117 (2007) 659–671. A. Bonetto, F. Penna, V.G. Minero, P. Reffo, G. Bonelli, F.M. Baccino, et al., Deacetylase inhibitors modulate the myostatin/follistatin axis without improving cachexia in tumor-bearing mice, Curr. Cancer Drug Targets 9 (2009) 608–616.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145

G Model YPHRS 2837 1–16

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231

[88] M.E. Benny Klimek, T. Aydogdu, M.J. Link, M. Pons, L.G. Koniaris, T.A. Zimmers, Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia, Biochem. Biophys. Res. Commun. 391 (2010) 1548–1554. [89] G.C. Minetti, C. Colussi, R. Adami, C. Serra, C. Mozzetta, V. Parente, et al., Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors, Nat. Med. 12 (2006) 1147–1150. [90] C. Colussi, C. Mozzetta, A. Gurtner, B. Illi, J. Rosati, C. Gaetano, et al., HDAC2 blockade by nitric oxide and histone deacetylase inhibitors reveals a common target in Duchenne muscular dystrophy treatment, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 19183–19187. [91] C. Colussi, A. Gurtner, J. Rosati, B. Illi, G. Ragone, C. Gaetano, et al., Nitric oxide deficiency determines global chromatin changes in Duchenne muscular dystrophy, FASEB J. 23 (2009) 2131–2141. [92] L. Combaret, T. Tilignac, A. Claustre, L. Voisin, D. Taillandier, C. Obled, et al., Torbafylline (HWA 448) inhibits enhanced skeletal muscle ubiquitin–proteasome-dependent proteolysis in cancer and septic rats, Biochem. J. 361 (2002) 185–192. [93] F.C. Mei, J. Qiao, O.M. Tsygankova, J.L. Meinkoth, L.A. Quilliam, X. Cheng, Differential signalling of cyclic AMP: opposing effects of exchange protein directly activated by cyclic AMP and cAMP-dependent protein kinase on protein kinase B activation, J. Biol. Chem. 277 (2002) 11497–11504. [94] E.O. Brennesvik, C. Ktori, J. Ruzzin, E. Jebens, P.R. Shepherd, J. Jensen, Adrenaline potentiates insulin-stimulated PKB activation via cAMP and Epac: implications for cross talk between insulin and adrenaline, Cell Signal. 17 (2005) 1551–1559. [95] S. Gerlo, P. Verdood, R. Kooijman, Modulation of cytokine production by cyclic adenosine monophosphate analogs in human leukocytes, J. Interferon Cytokine Res. 30 (2010) 883–891. [96] M. Metrich, M. Berthouze, E. Morel, B. Crozatier, A.M. Gomez, F. Lezoualc’h, Role of the cAMP-binding protein Epac in cardiovascular physiology and pathophysiology, Pflugers Arch. 459 (2010) 535–546. [97] D. Breuillé, M.C. Farge, F. Rose, M. Arnal, D. Attaix, C. Obled, Pentoxifylline decreases body weight loss and muscle protein wasting characteristics of sepsis, Am. J. Physiol. 265 (1993) E660–E666. [98] A.M. Baviera, N.M. Zanon, L.C. Carvalho Navegantes, R.H. Migliorini, I. do Carmo Kettelhut, Pentoxifylline inhibits Ca2+ -dependent and ATP proteasome-dependent proteolysis in skeletal muscle from acutely diabetic rats, Am. J. Physiol. Endocrinol. Metab. 292 (2007) E702–E708. [99] L. Combaret, C. Rallière, D. Taillandier, K. Tanaka, D. Attaix, Manipulation of the ubiquitin–proteasome pathway in cachexia: pentoxifylline suppresses the activation of 20S and 26S proteasomes in muscles from tumor-bearing rats, Mol. Biol. Rep. 26 (1999) 95–101. [100] J. Semmler, U. Gebert, T. Eisenhut, J. Moeller, M.M. Schönharting, A. Alléra, et al., Xanthine derivatives: comparison between suppression of tumour necrosis factor-alpha production and inhibition of cAMP phosphodiesterase activity, Immunology 78 (1993) 520–525. [101] C. Deval, S. Mordier, C. Obled, D. Bechet, L. Combaret, D. Attaix, et al., Identification of cathepsin L as a differentially expressed message associated with skeletal muscle wasting, Biochem. J. 360 (2001) 143–150. [102] B.T. Steffen, S.J. Lees, F.W. Booth, Anti-TNF treatment reduces rat skeletal muscle wasting in monocrotaline-induced cardiac cachexia, J. Appl. Physiol. 105 (2008) (1985) 1950–1958. [103] R.T. Hinkle, E. Dolan, D.B. Cody, M.B. Bauer, R.J. Isfort, Phosphodiesterase 4 inhibition reduces skeletal muscle atrophy, Muscle Nerve 32 (2005) 775–781. [104] D.A. Goncalves, E.C. Lira, A.M. Baviera, P. Cao, N.M. Zanon, Z. Arany, et al., Mechanisms involved in 3 ,5 -cyclic adenosine monophosphatemediated inhibition of the ubiquitin–proteasome system in skeletal muscle, Endocrinology 150 (2009) 5395–5404. [105] J.D. Knight, R. Kothary, The myogenic kinome: protein kinases critical to mammalian skeletal myogenesis, Skelet. Muscle 1 (2011) 29. [106] R. Stewart, L. Flechner, M. Montminy, R. Berdeaux, CREB is activated by muscle injury and promotes muscle regeneration, PLoS ONE 6 (2011) e24714. [107] J.G. Ryall, G.S. Lynch, The potential and the pitfalls of beta-adrenoceptor agonists for the management of skeletal muscle wasting, Pharmacol. Ther. 120 (2008) 219–232. [108] Y. Quanjun, Y. Genjin, W. Lili, L. Bin, L. Jin, Y. Qi, et al., Serum metabolic profiles reveal the effect of formoterol on cachexia in tumor-bearing mice, Mol. Biosyst. 9 (2013) 3015–3025. [109] O.R. Joassard, A. Amirouche, Y.S. Gallot, M.M. Desgeorges, J. Castells, A.C. Durieux, et al., Regulation of Akt-mTOR, ubiquitin–proteasome and autophagy–lysosome pathways in response to formoterol administration in rat skeletal muscle, Int. J. Biochem. Cell Biol. 45 (2013) 2444–2455. [110] M. Toledo, J. Springer, S. Busquets, A. Tschirner, F.J. López-Soriano, S.D. Anker, et al., Formoterol in the treatment of experimental cancer cachexia: effects on heart function, J. Cachexia Sarcopenia Muscle 5 (2014) 315–320. [111] B. D’Agostino, M. Polverino, G. Cirino, A. Lombardi, B. Grassi, N. Sullo, et al., Exercise capacity and cytochrome oxidase activity in muscle mitochondria of COPD patients, Respir. Med. 104 (2010) 83–90. [112] N. Sullo, F. Roviezzo, M. Matteis, G. Spaziano, S. Del Gaudio, A. Lombardi, et al., Skeletal muscle oxidative metabolism in an animal model of pulmonary emphysema: formoterol and skeletal muscle dysfunction, Am. J. Respir. Cell Mol. Biol. 48 (2013) 198–203. [113] S. Busquets, M.T. Figueras, G. Fuster, V. Almendro, R. Moore-Carrasco, E. Ametller, et al., Anticachectic effects of formoterol: a drug for potential treatment of muscle wasting, Cancer Res. 64 (2004) 6725–6731.

15

[114] S. Busquets, M. Toledo, E. Marmonti, M. Orpí, E. Capdevila, A. Betancourt, et al., Formoterol treatment downregulates the myostatin system in skeletal muscle of cachectic tumour-bearing rats, Oncol. Lett. 3 (2012) 185–189. [115] C.A. Steelman, J.C. Recknor, D. Nettleton, J.M. Reecy, Transcriptional profiling of myostatin-knockout mice implicates Wnt signaling in postnatal skeletal muscle growth and hypertrophy, FASEB J. 20 (2006) 580–582. [116] C. McFarlane, E. Plummer, M. Thomas, Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-␬B-independent. FoxO1-dependent mechanism, J. Cell Physiol. 209 (2006) 501–514. [117] D. Joulia-Ekaza, G. Cabello, The myostatin gene: physiology and pharmacological relevance, Curr. Opin. Pharmacol. 7 (2007) 310–315. [118] L.R. Rodino-Klapac, A.M. Haidet, J. Kota, C. Handy, B.K. Kaspar, J.R. Mendell, Inhibition of myostatin with emphasis on follistatin as a therapy for muscle disease, Muscle Nerve 39 (2009) 283–296. [119] H. Amthor, G. Nicholas, I. McKinnell, C.F. Kemp, M. Sharma, R. Kambadur, et al., Follistatin complexes myostatin and antagonises myostatin-mediated inhibition of myogenesis, Dev. Biol. 270 (2004) 19–30. [120] E. Ametller, S. Busquets, G. Fuster, M.T. Figueras, M. Olivan, C.C.F. de Oliveira, et al., Formoterol may activate rat muscle regeneration during cancer cachexia, Insci. J. 1 (2011) 1–17. [121] S.R. Jesinkey, M.C. Korrapati, K.A. Rasbach, C.C. Beeson, R.G. Schnellmann, Atomoxetine prevents dexamethasone-induced skeletal muscle atrophy in mice, J. Pharmacol. Exp. Ther. 351 (2014) 663–673. [122] J.G. Ryall, J.D. Schertzer, G.S. Lynch, Attenuation of age-related muscle wasting and weakness in rats after formoterol treatment: therapeutic implications for sarcopenia, J. Gerontol. A: Biol. Sci. Med. Sci. 62 (2007) 813–823. [123] J.D. Ma, S.F. Heavey, C. Revta, E.J. Roeland, Novel investigational biologics for the treatment of cancer cachexia, Expert Opin. Biol. Ther. 14 (2014) 1113–1120. [124] M. Llovera, N. Carbó, C. García-Martínez, P. Costelli, L. Tessitore, F.M. Baccino, et al., Anti-TNF treatment reverts increased muscle ubiquitin gene expression in tumour-bearing rats, Biochem. Biophys. Res. Commun. 221 (1996) 653–655. [125] M. Granado, A.I. Martín, T. Priego, A. López-Calderón, M.A. Villanúa, Tumour necrosis factor blockade did not prevent the increase of muscular muscle RING finger-1 and muscle atrophy F-box in arthritic rats, J. Endocrinol. 191 (2006) 319–326. [126] J.N. Gordon, P.M. Goggin, Thalidomide and its derivatives: emerging from the wilderness, Postgrad. Med. J. 79 (2003) 127–132. [127] S. Joglekar, M. Levin, The promise of thalidomide: evolving indications, Drugs Today (Barc) 40 (2004) 197–204. [128] I. Kedar, W. Mermershtain, H. Ivgi, Thalidomide reduces serum C-reactive protein and interleukin-6 and induces response to IL-2 in a fraction of metastatic renal cell cancer patients who failed IL-2-based therapy, Int. J. Cancer 110 (2004) 260–265. [129] M. Stroud, Thalidomide and cancer cachexia: old problem, new hope? Gut 54 (2005) 447–448. [130] M. Davis, W. Lasheen, D. Walsh, F. Mahmoud, L. Bicanovsky, R. Lagman, A Phase II dose titration study of thalidomide for cancer-associated anorexia, J. Pain Symptom Manage. 43 (2012) 78–86. [131] K.H. Liu, L.M. Liao, L.S. Ro, Y.L. Wu, T.S. Yeh, Thalidomide attenuates tumor growth and preserves fast-twitch skeletal muscle fibers in cholangiocarcinoma rats, Surgery 143 (2008) 375–383. [132] R. Sartori, P. Gregorevic, M. Sandri, TGF␤ and BMP signaling in skeletal muscle: potential significance for muscle-related disease, Trends Endocrinol. Metab. 25 (2014) 464–471. [133] G. Kaplan, S. Thomas, D.S. Fierer, K. Mulligan, P.A. Haslett, W.J. Fesse, et al., Thalidomide for the treatment of AIDS-associated wasting, AIDS Res. Hum. Retroviruses 16 (2000) 1345–1355. [134] J. Fujita, J.R. Mestre, J.B. Zeldis, K. Subbaramaiah, A.J. Dannenberg, Thalidomide and its analogues inhibit lipopolysaccharide-mediated Iinduction of cyclooxygenase-2, Clin. Cancer Res. 7 (2001) 3349–3355. [135] S. Dodson, V.E. Baracos, A. Jatoi, W.J. Evans, D. Cella, J.T. Dalton, et al., Muscle wasting in cancer cachexia: clinical implications, diagnosis, and emerging treatment strategies, Annu. Rev. Med. 62 (2011) 265–279. [136] T. Thum, J. Springer, Breakthrough in cachexia treatment through a novel selective androgen receptor modulator, J. Cachexia Sarcopenia Muscle 2 (2011) 121–123. [137] J.T. Dalton, K.G. Barnette, C.E. Bohl, M.L. Hancock, D. Rodriguez, S.T. Dodson, et al., The selective androgen receptor modulator GTx-024 (enobosarm) improves lean body mass and physical function in healthy elderly men and postmenopausal women: results of a double-blind, placebo-controlled phase II trial, J. Cachexia Sarcopenia Muscle 2 (2011) 153–161. [138] A.S. Dobs, R.V. Boccia, C.C. Croot, N.Y. Gabrail, J.T. Dalton, M.L. Hancock, et al., Effects of enobosarm on muscle wasting and physical function in patients with cancer: a double-blind, randomised controlled phase 2 trial, Lancet Oncol. 14 (2013) 335–345. [139] R. Blanqué, L. Lepescheux, M. Auberval, D. Minet, D. Merciris, C. Cottereaux, et al., Characterization of GLPG0492, a selective androgen receptor modulator, in a mouse model of hindlimb immobilization, BMC Musculoskelet. Disord. 15 (2014) 291. [140] A. Cozzoli, R.F. Capogrosso, V.T. Sblendorio, M.M. Dinardo, C. Jagerschmidt, F. Namour, et al., GLPG0492, a novel selective androgen receptor modulator, improves muscle performance in the exercised-mdx mouse model of muscular dystrophy, Pharmacol. Res. 72 (2013) 9–24.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317

G Model YPHRS 2837 1–16 16 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343

ARTICLE IN PRESS V. Dutt et al. / Pharmacological Research xxx (2015) xxx–xxx

[141] M. Chasen, S.Z. Hirschman, R. Bhargava, Phase II study of the novel peptidenucleic acid OHR118 in the management of cancer-related anorexia/cachexia, J. Am. Med. Dir. Assoc. 12 (2011) 62–67. [142] ClinicalTrials.gov. Open label study with OHR/AVR118 in advanced cancer patients with anorexia–cachexia. http://clinicaltrials.gov/ct2/show/ NCT01206335. [143] G. Mantovani, C. Madeddu, A. Macciò, Drugs in development for treatment of patients with cancer-related anorexia and cachexia syndrome, Drug Des. Dev. Ther. 7 (2013) 645–656 (article has retracted). [144] ClinicalTrials.gov. Pilot Study of APD209 in Cancer Cachexia http:// clinicaltrials.gov/show/NCT00895726. [145] X. Zhou, J.L. Wang, J. Lu, Y. Song, K.S. Kwak, Q. Jiao, et al., Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival, Cell 142 (2010) 531–543. [146] T.A. Zimmers, M.V. Davies, L.G. Koniaris, P. Haynes, A.F. Esquela, K.N. Tomkinson, et al., Induction of cachexia in mice by systemically administered myostatin, Science 296 (2002) 1486–1488. [147] K. Patel, H. Amthor, The function of myostatin and strategies of myostatin blockade-new hope for therapies aimed at promoting growth of skeletal muscle, Neuromuscul. Disord. 15 (2005) 117–126. [148] S. Roth, S. Walsh, Myostatin: a therapeutic target for skeletal muscle wasting, Curr. Opin. Clin. Nutr. Metab. Care 7 (2004) 259–263. [149] S. Bogdanovich, T.O. Krag, E.R. Barton, L.D. Morris, L.A. Whittemore, R.S. Ahima, et al., Functional improvement of dystrophic muscle by myostatin blockade, Nature 420 (2002) 418–421. [150] https://clinicaltrials.gov/ct2/show/NCT02250443. [151] https://clinicaltrials.gov/ct2/show/NCT01963598.

[152] C.L. Donohoe, A.M. Ryan, J.V. Reynolds, Cancer cachexia: mechanisms and clinical implications, Gastroenterol. Res. Pract. 2011 (2011) 601434. [153] O. Kanat, E. Cubukcu, N. Avci, F. Budak, I. Ercan, M. Canhoroz, et al., Comparison of three different treatment modalities in the management of cancer cachexia, Tumori 99 (2013) 229–233. [154] A. Pascual López, M. Roqué i Figuls, G. Urrútia Cuchi, E.G. Berenstein, B. Almenar Pasies, M. Balcells Alegre, et al., Systematic review of megestrol acetate in the treatment of anorexia–cachexia syndrome, J. Pain Symptom Manage 27 (2004) 360–369. [155] A. Laviano, A. Inui, D.L. Marks, M.M. Meguid, C. Pichard, F. Rossi Fanelli, et al., Neural control of the anorexia–cachexia syndrome, Am. J. Physiol. Endocrinol. Metab. 295 (2008), E1000-1008. [156] T. Martins, R. Vitorino, D. Moreira-Gonc¸alves, F. Amado, J.A. Duarte, R. Ferreira, Recent insights on the molecular mechanisms and therapeutic approaches for cardiac cachexia, Clin. Biochem. 47 (2014) 8–15. [157] H.D. McCarthy, R.E. Crowder, S. Dryden, G. Williams, Megestrol acetate stimulates food and water intake in the rat: effects on regional hypothalamic neuropeptide Y concentrations, Eur. J. Pharmacol. 265 (1994) 99–102. [158] G. Mantovani, A. Macciò, E. Massa, C. Madeddu, Managing cancer-related anorexia/cachexia, Drugs 61 (2001) 499–514. [159] S. Busquets, R. Serpe, S. Sirisi, M. Toledo, J. Coutinho, R. Martínez, et al., Megestrol acetate: its impact on muscle protein metabolism supports its use in cancer cachexia, Clin. Nutr. 29 (2010) 733–737. [160] S.W. Jones, R.J. Hill, P.A. Krasney, B. O’Conner, N. Peirce, P.L. Greenhaff, Disuse atrophy and exercise rehabilitation in humans profoundly affects the expression of genes associated with the regulation of skeletal muscle mass, FASEB J. 18 (2004) 1025–1027.

Please cite this article in press as: V. Dutt, et al., Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action, Pharmacol Res (2015), http://dx.doi.org/10.1016/j.phrs.2015.05.010

1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370

Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action.

Over the last two decades, new insights into the etiology of skeletal muscle wasting/atrophy under diverse clinical settings including denervation, AI...
2MB Sizes 1 Downloads 13 Views