Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 2.
Published in final edited form as: Clin Exp Pharmacol Physiol. 2015 Jan;42(1):1–13. doi: 10.1111/1440-1681.12320

Pharmacology of manipulating lean body mass

Patricio V Sepulveda *, Ernest D Bush , Keith Baar
PMCID: PMC4383600  NIHMSID: NIHMS671877  PMID: 25311629

Summary

  1. Dysfunction and wasting of skeletal muscle as a consequence of illness decreases the length and quality of life. Currently, there are few, if any, effective treatments available to address these conditions. Hence, the existence of this unmet medical need has fuelled large scientific efforts.

  2. Fortunately, these efforts have shown many of the underlying mechanisms adversely affecting skeletal muscle health.

  3. With increased understanding have come breakthrough disease-specific and broad spectrum interventions, some progressing through clinical development.

  4. The present review focuses its attention on the role of the antagonistic process regulating skeletal muscle mass before branching into prospective promising therapeutic targets and interventions. Special attention is given to therapies in development against cancer cachexia and Duchenne muscular dystrophy before closing remarks on design and conceptualization of future therapies are presented to the reader.

Keywords: atrophy, cachexia, Duchenne muscular dystrophy, exercise, hypertrophy, regeneration, skeletal muscle

Introduction

Skeletal muscle accounts for nearly half of the mass of the human body. It is not surprising then that this tissue plays a fundamental role in human health and well-being. Skeletal muscle is not only required for locomotion, but it plays an important role in many metabolic pathways and directly impacts our overall quality of life.1,2 In adults, interventions promoting an increase in muscle mass (hypertrophy) and strength are generally associated with positive health consequences, such as delaying the onset and reducing the severity of disease.3 For these reasons, an increase in muscle size and strength is associated with an increase in longevity.4 By contrast, skeletal muscle loss (atrophy) is often a reflection of the seriousness of a disease, and has a negative impact on quality of life and the ability to respond to and/or recover from disease.58

Diseases affecting the maintenance of lean body mass most often occur as muscle loss or wasting conditions. The medical severity of this muscle atrophy will often depend on health status, sex and the type of muscle affected.9,10 Although most forms of muscle wasting share similar gross phenotypic similarities, the molecular events that lead to atrophy can vary, and these differences are important to consider when designing new therapeutic interventions.11,12 Outlined here are some of the major muscle disorders where pharmacological interventions are common or desired. The objective of the present review was to show the overlapping threads that tie these interventions together, and to illustrate where synergistic pharmacologies might lead to new treatment paradigms or where new interventions might add value.

Maintenance of Skeletal Muscle: Negative Factors Effecting the Balance

Muscle loss occurs when the equilibrium between myofibrillar protein synthesis and degradation is perturbed (Fig. 1). This occurs in conditions where the protein synthesis machinery fails, either by failing to keep up with the rate at which proteins are being degraded or by making unstable/malformed proteins. A negative balance exists, tilting this balance towards degradation, in genetic neuromuscular disorders, sepsis, fasting, disuse, paralysis and ageing.1321 Excessive breakdown of proteins can also occur as an unwanted consequence of therapeutic treatments, such as corticosteroid administration in intensive care, in burn victims and after treatment with lipid-lowering drugs.2226 The general consensus is that the increase in degradation involves one or more of the proteolytic pathways: the ubiquitin proteasome pathway, autophagy, the lysosomal pathway, calpains and/or the caspase (or apoptotic) protease pathway.14,2729 It is currently unclear whether every proteolytic pathway is involved in the breakdown of muscular proteins. However, it is clear that multiple factors contribute to accelerated muscle loss. More in depth discussion of these disorders is presented here.

Fig. 1.

Fig. 1

The balance between muscle growth and wasting in a variety of conditions. Muscle growth conditions largely signal through mammalian target of rapamycin (mTOR), whereas wasting has multiple triggers and molecular targets including nuclear factor kappa B (NF-κB), growth arrest and DNA damage-inducible 45 (Gadd45) and small mother of decapentaplegic (Smad)2/3. ATF4, activating transcription factor 4; DMD, Duchenne muscular dystrophy; Facioscapulohumeral Muscular Dystrophy (FSH); histone deacetylase (HDAC); Limb-girdle muscular dystrophy (LGD); SARM, selective androgen receptor modulators; TWEAK, tumour necrosis factor superfamily 12.

Genetic disorders

Inherited myopathies are a large group of deleterious conditions leading to progressive muscle loss, weakness and a shorter lifespan.30 The most common myopathy, Duchenne muscular dystrophy (DMD), occurs as a result of aberrant expression (or lack) of dystrophin.31 In the absence of dystrophin, the muscle becomes more prone to damage, and as a result shows a large increase in the rate of muscle protein degradation that cannot be compensated for by simply increasing de novo protein synthesis. Potential therapeutic developments against DMD have focused on rectifying the gene mutation, enhancing muscle function and supressing the resulting inflammatory response that leads to necrosis and fibrosis.

Cachexia

Cachexia is the loss of muscle mass occurring from increased circulation of muscle-wasting factors as a result of disease. Cachexia occurs in some types of cancer, uraemia and stroptozotocin-induced diabetes.32 Cachexia also results from chronic heart failure, COPD or after the administration of corticosteroids or blood pressure-lowering medications.26,3337 Although there is some evidence that muscle protein synthesis is depressed in patients with cachexia, increases in the ubiquitinatin proteasome pathway and autophagy resulting in an increase in the rate of protein degradation also play an important role in this form of muscle loss.38,39 A hallmark of cachexia is that the loss of lean body mass cannot be prevented or reversed simply by increasing nutritional intake.40

Sarcopenia

The decline in muscle mass and decrease in muscle function as a consequence of age in the absence of disease is known as sarcopenia.4143 The rate of protein breakdown in muscles of individuals with sarcopenia is the same as that of young controls, suggesting that a deficit in protein synthesis is the basis for age-associated muscle loss.44 In support of this hypothesis, sarcopenia is characterized by decreased myogenic (anabolic) markers at rest, and on load, increased production of oxidative species, decreased anti-oxidant levels and reduced capacity to activate satellite cells.43, 45,46 This loss in skeletal muscle plasticity occurs concomitantly with an impaired ability to increase protein synthesis in response to exercise or nutrition.4749

Non-degenerative conditions of muscle wasting

There are several non-degenerative inducers of muscle wasting. These include, malnutrition, inactivity, injury or trauma to nerves (denervation) and/or tendons.50 Loss of innervation leading to paralysis can occur as a result of damage or death of the motor unit, sectioning of the nerve, or disruption between acetylcholine and its receptor (such as a result of post-synaptic block or neuromuscular blockade while in intensive care unit).18,51 Experimentally, loss of innervation is often used to model the progression of neuromuscular diseases such as amyotrophic lateral sclerosis.5254 In denervated muscles, early atrophy results from an increase in degradation in the absence of changes in protein synthesis.55 However, after prolonged denervation, there is an increase in protein synthesis that partially preserves muscle mass, suggesting that in some cases compensatory mechanisms exist to maintain muscle in the absence of proper signals (see below).55,56 In contrast to denervation, tendon trauma or forced inactivity (bedrest) leads to the mechanical unloading of a muscle while still retaining the intact nerve that supplies both electrical activity and trophic factors.50 As a result, the muscle atrophy is less severe in humans, and is largely the result of decreases in the rate of muscle protein synthesis.57

Signalling Underlying the Negative Control of Muscle Mass

Growth arrest and DNA damage-inducible 45 signalling

As aforementioned, both fasting and denervation lead to rapid muscle atrophy. Even though these are very different physiological insults, they share common molecular responses. Both denervation and fasting induce growth arrest and DNA damage-inducible 45 (Gadd45), a small nuclear protein that alters the transcriptional profile of skeletal muscle towards protein degradation and away from protein synthesis.11,58,19 Gadd45 increases and is required for normal muscle atrophy after fasting, immobilization and denervation, suggesting that Gadd45 is central to acute in vivo skeletal muscle atrophy.59 In the case of fasting, the increase in Gadd45 is driven by activating transcription factor (ATF) 4.58 Mice that lack a functional ATF4 do not increase Gadd45, and suffer much less atrophy than control mice when fasted.58 However, ATF4 knockout mice still activate Gadd45 and atrophy normally after denervation.11 In denervation, the increase in Gadd45 is therefore not the result of an increase in ATF4, but rather results from the upregulation of the class II histone deacetylases, specifically HDAC4.11 At present, how immobilization induces Gadd45 and whether Gadd45 is involved in sarcopenia or cachexia has yet to be shown. However, the importance of Gadd45 in skeletal muscle atrophy suggests that Gadd45 is a potential pharmaceutical target for preventing muscle loss.

Tumour necrosis factor superfamily 12 signalling

Cytokines, and in particular tumour necrosis factor (TNF) superfamily 12 (TWEAK), have been shown to play a key role in accelerating the breakdown of skeletal muscle proteins during inflammatory conditions, such as cancer cachexia and chronic heart disease.60,61 TWEAK signals through nuclear factor kappa B (NF-κB) – a transcription factor involved in immune, inflammatory and cell survival responses that is heavily associated with protein degradation.6265 Absence of TWEAK is linked to a slight decrease in muscle cross-sectional area and a decrease in proteasome activity, improved skeletal muscle regeneration, and protection against denervation-mediated muscle wasting in mice.66,67 Accordingly, deletion of NF-κB has shown to increase muscle mass, force (in fast oxidative muscle fibres), protect against atrophy and enhance muscle regeneration.68 It is possible that the benefits observed in muscle through inhibition of this pathway are a result of activation of mammalian target of rapamycin (mTOR) through the Akt/growth factor pathway and/or decreased levels of ubiquitin ligases targeting muscle proteins.69,70 The recent discovery that NF-κB controls the transcription of the muscle-specific E3 ligase, MuRF1, suggests that TWEAK likely drives atrophy through the activation of degradation downstream of NF-κB.71

Maintenance of Skeletal Muscle: Positive Factors Affecting the Balance

Muscle mass gains occur during developmental growth, in response to growth factors, diet and exercise.7274 As with muscle atrophy, muscle hypertrophy is the result of a change in the net balance between protein synthesis (anabolism) and degradation (catabolism).63,7581 However, it has become clear over the past few years that muscle hypertrophy and atrophy are not identical processes in reverse. Even so, improved understanding of the biology of growth has led to diverse approaches for the positive regulation of muscle mass.82,83 These interventions have been designed to mimic, amplify or block a subset of signalling pathways implicated in muscle growth/wasting, and could in turn impact on hundreds, if not thousands, of muscle remodelling genes/gene regulators.18,32,40,63,8490

Signalling underlying the positive control of muscle mass

Over the last twenty years we have begun to understand the molecular mechanisms underlying the control of skeletal muscle mass development. Some of these are generalized pathways, molecular events that are required for any cell to grow, whereas others appear to be specific for controlling the size of skeletal muscle, independent of other tissues in the body. Therapeutically, it is the muscle-specific events that are the most attractive as a way to decrease side effects of any treatment. However, if more generalized growth pathways can be targeted to muscle this could make them valuable tools in treating muscle diseases. Below, we will briefly describe some of the known pathways that control muscle size in the adult.

Mammalian target of rapamycin pathway

Activation of mTOR is one of the key events involved in muscle growth. mTOR can be activated by: (i) growth factors, through the PI-3kinase/Akt pathway;91 (ii) mechanical loading, through the removal of the inhibitor TSC2 from the mTOR/Rheb complex;92 and (iii) feeding, through the GATOR/Rag/Ragulator pathway.93,94 In this way, mTOR can directly control muscle growth by integrating hormonal, nutritional and loading cues. mTOR activation after exercise correlates with muscle hypertrophy in both rodents and humans.94,96 Furthermore, when mTOR is specifically blocked by the bacterial macrolide, rapamycin, there is no acute rise in muscle protein synthesis after exercise or feeding, and muscle hypertrophy is prevented after overload.9799 Taken together, these data suggest that mTOR is required for the acute response of muscle to feeding and exercise, and is important in the regulation of muscle protein synthesis.

The activation of mTOR directly increases the rate of protein synthesis, and stimulates muscle hypertrophy through phosphorylation of protein 70 S6 kinase (p70S6K) and eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1) also known as PHAS-1.63,78 Additionally, phosphorylated 4E-BP1 can exert a positive feedback loop by binding to the regulatory associated protein of mTOR (raptor) and further activate mTOR.100 Phosphorylation of both p70S6K and 4E-BP1 are common readouts of anabolic processes.101 Activation of this pathway as a means to enhance muscle function has been the focus of intense efforts. For example, chemical activation of Akt has shown to protect against sepsis in mice.102 Also, exogenous delivery of recombinant human insulin-like growth factor-1 (IGF-1; an upstream activator of Akt) improves muscle function in dystrophic rodents and enhances the benefits of gene therapy.103,104 IGF-1 treatment seems safe (without side effects humans), and has the potential to improve body composition and metabolism in dystrophic patients.105107 Unfortunately, there are reasons to believe that stimulation of Akt through IGF-1 will not produce substantial functional improvements.73

Transforming growth factor-β signalling pathway

The transforming growth factor-β (TGF-β) signalling pathway is pivotal in controlling developmental muscle growth. The TGF-β family of ligands that activate this pathway, such as myostatin, TGF-β, bone morphogenic proteins (BMP) or Activin, act through a series of receptors and effector small mother of decapentaplegic (Smad) proteins to regulate hundreds of gene targets, many of which are involved in muscle growth and wasting.108110 The TGF-β family ligands bind to type II and type I transmembrane serine/threonine kinase receptors in the membrane of the muscle cell to exert biological action, predominantly through the Smad2 and Smad3 (Smad2/3) or Smad1, Smad5 or Smad8 (Smad1/5/8) axis (Fig. 2). 111

Fig. 2.

Fig. 2

The interplay between multiple factors in the transforming growth factor-β (TGF-β) signalling pathway and the control of muscle mass. Myostatin and similar members of the TGF-β superfamily can activate small mother of decapentaplegic (Smad)2/3 and drive transcriptional events that result in the transcription of genes that limit muscle size. This can be prevented at a variety of drug/protein inhibitors (indicated in the red text and red bars). Smad2/3 signalling can also be blocked by growth and differentiation factor 5 (GDF5) and other members of the TGF-β superfamily that activate Smad1/5/8, and this competes for Smad4 binding and shifts transcription. Resistance exercise can also limit Smad2/3 signalling by activating Notch and blocking Smad2/3 transcription. bone morphogenetic protein receptor 1 (BMPR1); dominant negative (DN); and myostatin (MSTN).

The most cited example of Smad2/3 modulation of muscle mass is myostatin, or its absence, which was first found as being responsible of the hypermuscular phenotype of the Belgium Blue cattle.112 Myostatin is an important factor in cellular differentiation and cell growth inhibition during wasting conditions.113115 The inhibition of the TGF-β pathway and positive regulation of muscle growth can occur extracellularly by limiting ligand activity/availability through antibodies, ligand antagonists such as Gasp-1, Myostatin pro-peptides, antisense nucleotides, dominant negative receptor competitors, soluble receptor inhibitors, the BMP Activin membrane-bound inhibitor (Bambi) and Follistatin (Fst).116126 Inhibition of the TGF-β pathway can also occur intracellularly by limiting ligand production through molecular interruption of protein production, inhibition of Smad signalling by SKI, release of Notch from the plasma membrane or competitive activation of Smad1/5/8.121,127131 In accordance, the absence or direct or indirect inhibition of myostatin produces muscle growth, and improves (in rodents) muscle size and strength in diseases such as Duchenne Muscular Dystrophy, amyotrophic lateral sclerosis and cachexia.132138 It is also believed that inhibition of myostatin could increase the success of myoblast transplantation.134,139

PGC-1α signalling

Recently, a potential role for the mitochondrial transcriptional cofactor the peroxisome proliferating activated receptor co-activator (PGC-1α) has been suggested.140 In these experiments, high load caused PGC-1α to be expressed from a different promoter resulting in the production of a novel mRNA that has been termed PGC-1α4. However, almost as quickly as the discovery of PGC-1α4 and its potential role in muscle growth came a report that showed that muscle growth occurred completely normally in the PGC-1α knockout mice who are unable to express PGC-1α4.141 It is still possible that PGC-1α4 plays a role in the regulation of developmental, but not load-induced, muscle growth through the regulation of either myostatin signaling or a G-protein coupled receptor. However, this remains to be conclusively shown.

G-protein signalling

The hypertrophic potential of β2 agonists has been known for decades.142 However, the potential of multiple different G-protein activators to produce skeletal muscle hypertrophy suggests that this is a more generalized phenomenon. G-protein activation by Wnt, ghrelin, lysophosphatidic acid, as well as clenbuterol, fenoterol and formoterol, can increase muscle mass.142146 Interestingly, this effect is dependent on mTOR, as inactivation of mTOR by rapamycin prevents G-protein coupled muscle hypertrophy, suggesting that there are other therapeutically relevant ways to activate mTOR within muscle.143,147 One of the clinical drawbacks to this family of therapeutics has been that the current β2 agonists produce cardiac hypertrophy as well as skeletal muscle hypertrophy.148 However, the discovery of a skeletal muscle specific G-protein coupled receptor (possibly the receptor of Wnt7a) has the potential to revolutionize muscle therapeutics.

Potential Interventions Against Muscle Wasting Conditions

There is a large unmet medical need for an intervention that maintains or re-establishes skeletal muscle mass and function. Current treatments to reduce the loss of muscle mass, such as nutritional supplementation (providing increased calorie intake and special amino acid compositions), hormonal treatment (growth hormone, testosterone, IGF-1 and insulin) and anti-inflammatories, have been largely ineffective or are associated with severe side-effects.5 Although it would be more cost-effective to develop a ‘one for all’ treatment than targeting each disease independently, it is unlikely that a single intervention will have the desired therapeutic effect across the variety of conditions associated with muscle atrophy. For these reasons, the following section highlights a selection of targets that in the opinion of the authors are likely to advance through clinical development.

Tgf-β Signalling Pathway

There has been an immense amount of attention devoted to developing strategies to inhibit the TGF-β family ligands, as it largely targets skeletal muscle and holds immense promise as a therapeutic target. As mentioned earlier, these interventions vary from a compound approach, to other approaches, such as the delivery of antibodies against myostatin in cancer patients (phase II trial by Eli Lilly, clinicaltrials.gov identifier: NCT01505530), delivery of pro-peptides that can bind to and block myostatin in solution or by transient DNA expression that elicits muscle growth and improves muscle function.116,149151 Encouraging results have also been produced from delivering short interfering hairpin RNA against myostatin, showing increased muscle mass in rodents. Other alternatives to manipulate this pathway are the inhibition of Activin receptor type IIB (ActRIIB) through rAAV delivery of a dominant negative receptor or antibody-targeted receptor inhibitors.139,152,153 It is well known that Fst and Fst-related proteins can promote hypertrophy in the skeletal muscle of rodents and non-human primates, so it is not surprising that a clinical trial is currently underway to test the potential of Fst in patients with Becker muscular dystrophy (BMD) and sporadic inclusive myositis (NCT01519349).134,154157 In addition, Acceleron Pharma/Shire developed ACE-031, a ActRIIB receptor chimera with strong capacity to decrease body fat, increase muscle mass and improve insulin sensitivity.136,158,159 However, ACE-031 has been discontinued given that a phase II clinical trial raised safety concerns (NCT01099761). Although the safety concerns about ACE-031 have not been published, it is possible that this molecule had off-target effects, given its lack of target tissue specificity.

Another strategy for TGF-β signalling pathway inhibition is the treatment with halofuginone hydrobromide, which in animals and humans, acts as a potent inhibitor of the synthesis of collagen type I at the transcriptional level by inhibiting Smad3 phosphorylation, resulting in ubiquituous reduction of fibrosis and collagen synthesis.160,160 Importantly, when administered at intended doses, halofuginone hydrobromide has a minimal effect on collagen content in non-fibrotic tissues.162165 Halofuginone hydrobromide also produces anti-inflammatory and antifibrotic effects by inhibiting activated peripheral blood T-cell function and pro-inflammatory cytokine production, including interferon-γ and TNF-α, through inhibition of NF-κB activation and p38 mitogen-activated protein kinase (MAPK) in a dose-dependent manner.160 Finally, halofuginone hydrobromide has recently been shown to enhance myotube fusion by stimulating serine/threonine protein kinase Akt, MAPK/extracellular- signal-related kinase and MAPK phosphorylation, and inhibiting Smad3 phosphorylation in myotubes, all of which are crucial for myotube formation.166

Recently, the protagonist role of the Smad2/3 axis has been challenged by two reports showing that the Smad1/5/8 axis can modulate Smad2/3 activity, and might even have a dominant role over Smad2/3. Almost simultaneously, Sartori et al. and Winbanks et al. reported that the Smad1/5/8 axis of the TGF-β pathway can promote muscle growth and override Smad2/3 signalling in mice (Fig. 2).56,167 Using transgenic mice models, Sartori et al. showed that phosphorylated Smad2/3 forms a complex with Smad4 to accelerate muscle mass loss, whereas the opposite occurred on its release from the Smad2/3 complex and association with Smad1/5/8 (Fig. 2). Consequently, inhibition of Smad1/5/8 increased denervation-mediated muscle wasting, and overactivation of Smad1/5/8 inhibited myostatin-mediated muscle growth independent of IGF-1 signalling. Winbanks et al. used adeno-associated viral vectors (AAV) to activate this pathway, and promote growth in adult mice. AAV-mediated overexpression of the ligand, BMP7, or introduction of a constitutively active ALK3 (a subunit of the BMP receptor) was sufficient to promote muscle growth. Additionally, AAV-mediated upregulation of the naturally occurring Smad1/5/8 inhibitor, Smad6, blocked Fst-mediated muscle growth. Together, these findings highlight the importance of testing the potential of Smad1/5/8-activating interventions for reversal of atrophy and frailty.

Selective Androgen Receptor Modulators

Selective androgen receptor modulators (SARM) are a class of pharmaceuticals intended for oral delivery that offer to stimulate the normal androgen response, but do so with tissue selectivity; that is, to stimulate a normal testosterone-like response in those tissues in need of the targeted effect, but avoiding those tissues where the testosterone effect would be considered adverse. The recent examples in this field are targeting skeletal muscle (i.e. anabolic effects), but avoiding prostate gland (i.e. the androgenic effects). The ratios of activity in these organs that are used in the development SARM therapies are 3:1 to >10:1, indicating the drug has a three- or 10-fold greater effect on skeletal muscle than the prostate.168,169

There are many mechanisms by which testosterone and other androgens are believed to provide their effects, but increasingly for muscle mass increases the SARM are believed to act through ‘anticatabolic’ pathways (Fig. 1).170,171 That is they operate not so much as stimulators of new protein synthesis, but rather through inhibiting the normal degradation rate of muscle protein.168,169

Personalized Therapies

The increased understanding on the underlying causes of genetic musculopathies have led to the development of targeted therapies that allow correction or replacement of the deficient gene. A good example is the dystrophin gene, which is mutated in DMD. The dystrophin gene comprises 79 exons, which are interspersed with non-coding introns. Deletions of one or more exons disrupts the synthesis of the dystrophin protein, which plays an essential role in muscle fibre function. Dystrophinopathies result in progressive muscle fibre degeneration through normal degenerative and inflammatory processes leading to necrosis and fibrosis. The extent and nature of the mutations vary across different subpopulations of DMD patients, but the key factor of the disease is a disruption in the coding of mRNA for dystrophin. If the defective mRNA is ‘out of frame’, this results in a premature termination of the protein, leading to a truncated and non-functional dystrophin protein within their muscles.

Patients with BMD show intermediate to milder phenotypes with mostly longer to normal life expectancies when compared with DMD patients. Here, the mutations in the dystrophin gene maintain the open reading frame and result in an internally truncated, but semi-functional, protein. Therefore, the specific skipping of one or more exons flanking the deletion in a DMD patient might allow restoration of the mutated open reading frame, introduction of novel, BMD-like dystrophin and conversion of a severe DMD into a typically milder BMD phenotype. As such, the aim of exon-skipping therapies is restoration of coding reading frame so that a shorter, but still functional, dystrophin protein can be produced.

Although exon skipping is a mutation-specific treatment, representing personalized medicine, an important intrinsic advantage compared with conventional gene therapy is that it simultaneously corrects all dystrophin isoforms. It also maintains the original tissue-specific gene regulation. Furthermore, the antisense compounds that induce exon skipping are small, can be engineered synthetically, and are highly sequence-specific.172,173

Although the exon-skipping approach has shown promise and several groups have published positive results, the need for many different gene-skipping therapies to cover a wide array of patient mutations is very challenging and indeed some mutations will never be addressed by exon skipping. Analogously, any viral vector or DMD gene therapy will require adequate levels of long-term expression across multiple muscle types in order to be effective. The applicability of AAV has improved the development of this technology, and with more recent CRISPR technology this type of therapy is on the cusp of clinical trials to prove its utility.174,175 For a recent review on this technology, see Jarmin et al. who made a comparison of the two approaches to gene therapy.173

Examples of New Therapy Development

Given the complexities of muscle homeostasis shown in Fig. 1, it would seem not only reasonable, but in fact required, that the optimal treatment of muscle loss conditions will involve multiple simultaneous therapeutic interventions. This is especially true for those conditions that arise from diseases originating from loss of protein as a result of genetic defects. As such, new drug developers should consider treatment paradigms that would include simultaneous interventions across multiple pathways as the ultimate end-points. As examples, two disease areas are summarized below.

DMD

Perhaps the best case in point is the treatment of DMD by prednisone. As aforementioned, DMD results from a genetic defect in the dystrophin gene, and is a serious condition leading to patient deaths in the late teens to 20s. The only known effective treatment of this disease is prednisone, a steroidal anti-inflammatory that has been show to act through multiple pathways including:

  1. Inhibition of transcription of COX-2, cytokines, cell adhesion molecules and inducible nitric oxide synthase.

  2. Blockage of vitamin D3-mediated induction of osteocalcin gene in osteoblasts.

  3. Modification of collagenase gene transcription.

  4. Increased synthesis of annexin-1, an important protein for negative feedback to the hypothalamus and anterior pituitary gland.

Interestingly, more focused anti-inflammatories, such as NSAIDs, have not been very helpful in treating the disease and it is clear the multipharmacology nature of prednisone is responsible for its effectiveness in DMD. As such, although exon skipping, or other gene replacement therapies, might be part of the answer, it is clear that combinatorial therapies will also be required, even if it is only because of the fact that not all genetic mutations in DMD are treatable by exon skipping. Other promising treatments include the restoration of dystrophin through exogenous expression of the protein or parts of it, or upregulation or delivery of utrophin, an alternative gene that shares functions with dystrophin.104,176178 Complementary approaches include development of new anti-fibrotics, as well as a number of new, multifunction anti-inflammatories.164 For an overview of the major programs in therapy development for DMD, see the website: http://tinyurl.com/ltkf2do. What should be clear from this overview is the wide diversity of therapies in development.

Cachexia

In addition to calorie supplementation, current therapeutic strategies for cachexia have been based on either blocking cytokine synthesis or action. Thalidomide has been shown to suppress TNF-α production in monocytes in vitro and to normalize elevated TNF-alpha levels in vivo.179 A recent randomized, placebo-controlled trial in patients with cancer cachexia showed the drug was well tolerated and effective at attenuating loss of weight and lean body mass in patients with advanced pancreatic cancer.180 An improvement in lean body mass and improved quality of life were also observed in a randomized, double-blind trial using a protein and energy-dense, omega-3 fatty acid-enriched oral supplement, provided that the concentration of eicosapentaenoic acid was equal to or greater than 2.2 g/day.181 This was also thought to work through decreasing TNF-α production. However, recent data arising from a large, multicenter, double-blind, placebo-controlled trial showed eicosapentaenoic acid administration alone is not successful in the treatment of weight loss in patients with advanced gastrointestinal or lung cancer.182 Peripheral muscle proteolysis, as it occurs in cancer cachexia, serves to mobilize amino acids required for the synthesis of liver and tumour proteins. Therefore, the administration of exogenous amino acids might theoretically serve as a muscle-sparing intervention by providing substrates for both muscle metabolism and gluconeogenesis. Recent studies have shown dietary supplementation with a specific combination of high protein, leucine and fish oil improves muscle function, daily activity and the immune response in cachectic tumour-bearing mice.183 In addition, when β-hydroxy-β-methylbutyrate, a metabolite derived from leucine catabolism, was used as a supplement in tumour-bearing rats, cachexia was prevented in part by modifying NF-κB expression.184 Also, a recent phase II study involving the administration of anti-oxidants, pharmaconutritional support, progestogen (megestrol acetate and medroxyprogesterone acetate) and anticyclooxygenase-2 drugs, showed efficacy and safety in the treatment of patients suffering from cachexia as a result of advanced cancer. These data reinforce the use of the multitargeted therapies (nutritional supplementation, appetite stimulants and physical activity regimen) in the treatment of cancer cachexia.184

Not surprisingly, myostatin inhibition has also been shown to protect against cachexia-mediated atrophy and increased survival.137,138,185 Leading this space is Novartis's Activin receptor A/B antibody in phase III clinical trials. In the same therapeutic space, another phase III clinical trial using the SARM category and the Enobosarm from GTX are most advanced.186

Opinion on Design and Conceptualization of Future Muscle Therapies Strategic considerations

The ramifications of the polypharmaceutical nature of treating muscle wasting diseases have the following implications for drug developers:

  1. Whenever two or more drugs are taken simultaneously, the potential exists for drug–drug interactions. As such, new drug development in this area should be especially focused on those compounds with reduced or absent ability to negatively interact with other drugs.

  2. Combining multiple therapies into one pill has repeatedly proven to enhance patient compliance with a regimen and therefore improved efficacy measures. Unfortunately, for this to be effective, the medicines within a single tablet or capsule need to have similar pharmacokinetic properties that support concomitant administration (e.g. once a day dosing).

  3. Drug discovery and development should explore combination therapies very early on in the program, and even co-develop therapies in order to hasten the development and increase the odds of success. Currently, the process, both from a pharmacological and a regulatory perspective, is focused on the preclinical and clinical development of one therapy at a time as single agents. This needs to change if we are going to expeditiously develop polypharmaceutical approaches to complex diseases.

Pharmacological aspect

From a pharmaceutical development perspective, drugs to treat diseases that alter muscle balance can be divided into three categories:

  1. Therapies that treat the primary cause of the disease; that is, a cure. For some diseases, such as DMD, this means replacing or fixing the defective gene or gene product; for other diseases, such as cachexia, it means curing the primary cause, which is most often cancer or heart failure.

  2. Therapies that do not cure the primary cause of the disease, but work to maintain or restore a normal balance of muscle protein synthesis and degradation, and as such maintain normal muscle function for longer periods of time.

  3. Therapies that treat the symptoms of the disease, such as antifibrotics and anti-inflammatories, that reduce the progression of the muscle damage caused by the primary disease processes and thereby indirectly maintain normal muscle function for longer periods of time.

Because muscle cells are constantly renewing, full cures of muscle diseases are difficult to obtain (e.g. so far gene therapy approaches have shown improvements, but not really full reversal of the disease). Therefore, a reasonable strategy for developing therapies in the muscle disease therapeutic area would be to span all three categories.

Conclusions

Future therapies for non-genetic muscle wasting conditions should first identify how muscle balance is being disturbed: decreased synthesis or increased degradation (Fig. 1). Next, the intrinsic (loss of signalling within muscle) or extrinsic (increased inflammatory cytokines) cause of the imbalance needs to be identified. With this information, any muscle wasting disease can be attacked at multiple points: blocking the primary cause and promoting an anabolic response. For example, muscle wasting as a result of generalized inflammation (for example, after burn) would be treated with an anti-inflammatory together with an agent to decrease protein degradation within muscle, such as a SARM. Simultaneous treatment with agents to stimulate anabolic processes, from at simplest a leucine-rich protein diet to more complex products designed to target specific growth pathways, would maximize the recovery or maintenance of muscle mass and function.

As we move forward in the development of drugs that promote muscle anabolism, it is clear from the G-protein, mTOR and TGF-β sections in the present article, that these growth pathways have multiple points of intersection. It is therefore conceivable that novel drugs can be developed that specifically target these intersection points within skeletal muscle and activate multiple growth pathways simultaneously. The development of drugs that have this capacity is the ultimate goal in the battle to increase muscle protein synthesis. However, as should be clear from the discussion here, such a drug alone would have limited use in a disease whose primary feature is an increase in degradation.

In conclusion, there are many opportunities and targets for treating conditions that disrupt the normal balance of muscle mass. As described here, each disease is the result of its ability to alter both synthesis and degradation. Therefore, we need to develop combination or monotherapies that attack each condition with its own unique amalgam of anabolic and anticatabolic agents. Tailoring treatments in this manner will offer enhanced responses and greater efficacy.

Acknowledgments

KB was supported by a project grant from the National Institute on Ageing of the National Institutes of Health under award number R01AG045375. PS is a shareholder in Myostin Therapeutics. KB is a stakeholder in Advanced Muscle Technologies. EB is CSO and shareholder in Akashi Therapeutics.

Footnotes

All authors contributed equally to the writing of this review.

References

  • 1.DeFronzo RA, Ferrannini E, Sato Y, Felig P, Wahren J. Synergistic interaction between exercise and insulin on peripheral glucose uptake. J Clin Invest. 1981;68(6):1468–74. doi: 10.1172/JCI110399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, Raptis SA. Insulin effects in muscle and adipose tissue. Diabetes research and clinical practice. 2011;93:S52–S9. doi: 10.1016/S0168-8227(11)70014-6. [DOI] [PubMed] [Google Scholar]
  • 3.Payne C, Wiffen Philip J, Martin S. Interventions for fatigue and weight loss in adults with advanced progressive illness. Cochrane Database of Systematic Reviews. 2012;(1) doi: 10.1002/14651858.CD008427.pub2. Internet. Available from: http://www.mrw.interscience.wiley.com/cochrane/clsysrev/articles/CD008427/frame.html. [DOI] [PubMed]
  • 4.Jonatan RR, Xuemei S, Felipe L, Morrow James R, Jr, Allen WJ, Michael S, et al. Association between muscular strength and mortality in men: prospective cohort study. BMJ. 2008;337 doi: 10.1136/bmj.a439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lynch GS, Schertzer JD, Ryall JG. Therapeutic approaches for muscle wasting disorders. Pharmacol Ther. 2007 Mar;113(3):461–87. doi: 10.1016/j.pharmthera.2006.11.004. [DOI] [PubMed] [Google Scholar]
  • 6.Janssen I, Shepard DS, Katzmarzyk PT, Roubenoff R. The healthcare costs of sarcopenia in the United States. Journal of the American Geriatric Society. 2004 Jan;52(1):80–5. doi: 10.1111/j.1532-5415.2004.52014.x. [DOI] [PubMed] [Google Scholar]
  • 7.Australian Institute of Health and Welfare Canberra. Health expenditure on national health priority areas. 2006 [Google Scholar]
  • 8.Kokkinos P, Myers J, Faselis C, Panagiotakos DB, Doumas M, Pittaras A, et al. Exercise capacity and mortality in older men. Circulation. 2010;122(8):790–7. doi: 10.1161/CIRCULATIONAHA.110.938852. [DOI] [PubMed] [Google Scholar]
  • 9.Martel GF, Roth SM, Ivey FM, Lemmer JT, Tracy BL, Hurlbut DE, et al. Age and sex affect human muscle fibre adaptations to heavy-resistance strength training. Experimental Physiology. 2006;91(2):457–64. doi: 10.1113/expphysiol.2005.032771. [DOI] [PubMed] [Google Scholar]
  • 10.Sandri M, Lin J, Handschin C, Yang W, Arany ZP, Lecker SH, et al. PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proceedings of the National Academy of Sciences. 2006;103(44):16260–5. doi: 10.1073/pnas.0607795103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bongers KS, Fox DK, Ebert SM, Kunkel SD, Dyle MC, Bullard SA, et al. Skeletal muscle denervation causes skeletal muscle atrophy through a pathway that involves both Gadd45a and HDAC4. American journal of physiology Endocrinology and metabolism. 2013 Oct 1;305(7):E907–15. doi: 10.1152/ajpendo.00380.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bialek P, Morris C, Parkington J, Andre M, St, Owens J, Yaworsky P, et al. Distinct protein degradation profiles are induced by different disuse models of skeletal muscle atrophy. Physiological Genomics. 2011;43(19):1075–86. doi: 10.1152/physiolgenomics.00247.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pearen MA, Myers SA, Raichur S, Ryall JG, Lynch GS, Muscat GEO. The Orphan Nuclear Receptor, NOR-1, a target of {beta}-adrenergic signaling, regulates gene expression that controls oxidative metabolism in skeletal muscle. Endocrinology. 2008;149(6):2853–65. doi: 10.1210/en.2007-1202. [DOI] [PubMed] [Google Scholar]
  • 14.Deval C, Mordier S, Obled C, Bechet D, Combaret L, Attaix D, et al. Identification of cathepsin L as a differentially expressed message associated with skeletal muscle wasting. The Biochemical Journal. 2001;360(Pt 1):143–50. doi: 10.1042/0264-6021:3600143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jagoe RT, Lecker SH, Gomes M, Goldberg AL. Patterns of gene expression in atrophying skeletal muscles: response to food deprivation. The FASEB Journal. 2002;16(13):1697–712. doi: 10.1096/fj.02-0312com. [DOI] [PubMed] [Google Scholar]
  • 16.Herbison GJ, Jaweed MM, Ditunno JF. Muscle atrophy in rats following denervation, casting, inflammation, and tenotomy. Archives Of Physical Medicine And Rehabilitation. 1979;60(9):401–4. [PubMed] [Google Scholar]
  • 17.Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nature Cell Biology. 2001;3(11):1014–9. doi: 10.1038/ncb1101-1014. [DOI] [PubMed] [Google Scholar]
  • 18.Sacheck JM, Hyatt JPK, Raffaello A, Jagoe RT, Roy RR, Edgerton VR, et al. Rapid disuse and denervation atrophy involve transcriptional changes similar to those of muscle wasting during systemic diseases. The FASEB Journal. 2007;21(1):140–55. doi: 10.1096/fj.06-6604com. [DOI] [PubMed] [Google Scholar]
  • 19.Clark BC, Manini TM, Hoffman RL, Russ DW. Restoration of voluntary muscle strength after 3 weeks of cast immobilization is suppressed in women compared with men. Archives of Physical Medicine and Rehabilitation. 2009;90(1):178–80. doi: 10.1016/j.apmr.2008.06.032. [DOI] [PubMed] [Google Scholar]
  • 20.Pasino E, Buffelli M, Busetto G, Cangiano A. Use of dexamethasone with TTX block of nerve conduction shows that muscle membrane properties are fully controlled by evoked activity. Brain Research. 1997;770(1-2):242–7. doi: 10.1016/s0006-8993(97)00881-0. [DOI] [PubMed] [Google Scholar]
  • 21.Adamo ML, Farrar RP. Resistance training, and IGF involvement in the maintenance of muscle mass during the aging process. Ageing Research Reviews. 2006;5(3):310–31. doi: 10.1016/j.arr.2006.05.001. [DOI] [PubMed] [Google Scholar]
  • 22.Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell. 2004 Apr 30;117(3):399–412. doi: 10.1016/s0092-8674(04)00400-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Clarke BA, Drujan D, Willis MS, Murphy LO, Corpina RA, Burova E, et al. The E3 Ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-treated skeletal muscle. Cell Metabolism. 2007;6(5):376–85. doi: 10.1016/j.cmet.2007.09.009. [DOI] [PubMed] [Google Scholar]
  • 24.Waddell DS, Baehr LM, van den Brandt J, Johnsen SA, Reichardt HM, Furlow JD, et al. The glucocorticoid receptor and Foxo1 synergistically activate the skeletal muscle atrophy associated MuRF1 gene. American Journal of Physiology - Endocrinology And Metabolism. 2008:646. doi: 10.1152/ajpendo.00646.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tuvdendorj D, Chinkes DL, Zhang XJ, Suman OE, Aarsland A, Ferrando A, et al. Long-term oxandrolone treatment increases muscle protein net deposition via improving amino acid utilization in pediatric patients 6 months after burn injury. Surgery. 2011;149(5):645–53. doi: 10.1016/j.surg.2010.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Phillips PS, Haas RH. Statin myopathy as a metabolic muscle disease. Expert Review Of Cardiovascular Therapy. 2008;6(7):971–8. doi: 10.1586/14779072.6.7.971. [DOI] [PubMed] [Google Scholar]
  • 27.Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL. Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proceedings of the National Academy of Sciences. 2001;98(25):14440–5. doi: 10.1073/pnas.251541198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Richard I, Broux O, Allamand V, Fougerousse F, Chiannilkulchai N. Mutations in the proteolytic enzyme calpain 3 cause limb-girdle muscular dystrophy type 2A. Cell. 1995;81:27. doi: 10.1016/0092-8674(95)90368-2. [DOI] [PubMed] [Google Scholar]
  • 29.Kroemer G, Dallaporta B, Resche-Rigon ML. The mitochondrial death/life regulator in apoptosis and necrosis. Annual Review of Physiology. 1998;60(1):619–42. doi: 10.1146/annurev.physiol.60.1.619. [DOI] [PubMed] [Google Scholar]
  • 30.Emery AEH. The muscular dystrophies. The Lancet. 2002;359(9307):687–95. doi: 10.1016/S0140-6736(02)07815-7. [DOI] [PubMed] [Google Scholar]
  • 31.Reed UC. Congenital muscular dystrophy. Part I: a review of phenotypical and diagnostic aspects. Arquivos de Neuro-Psiquiatria. 2009;67:144–68. doi: 10.1590/s0004-282x2009000100038. [DOI] [PubMed] [Google Scholar]
  • 32.Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, et al. Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. The FASEB Journal. 2004;18(1):39–51. doi: 10.1096/fj.03-0610com. [DOI] [PubMed] [Google Scholar]
  • 33.Hambrecht R, Schulze PC, Gielen S, Linke A, Mobius-Winkler S, Yu J, et al. Reduction of insulin-like growth factor-I expression in the skeletal muscle of noncachectic patients with chronic heart failure. Journal of the American College of Cardiology. 2002;39(7):1175–81. doi: 10.1016/s0735-1097(02)01736-9. [DOI] [PubMed] [Google Scholar]
  • 34.Carvalho R, Castan E, Coelho C, Lopes F, Almeida F, Michelin A, et al. Heart failure increases atrogin-1 and MuRF1 gene expression in skeletal muscle with fiber type-specific atrophy. Journal of Molecular Histology. 2010 doi: 10.1007/s10735-010-9262-x. [DOI] [PubMed] [Google Scholar]
  • 35.Doucet M, Russell AP, Leger B, Debigare R, Joanisse DR, Caron MA, et al. Muscle atrophy and hypertrophy signaling in patients with chronic obstructive pulmonary disease. American Journal of Respiratory Critical Care Medicine. 2007;176(3):261–9. doi: 10.1164/rccm.200605-704OC. [DOI] [PubMed] [Google Scholar]
  • 36.Plant PJ, Brooks D, Faughnan M, Bayley T, Bain J, Singer L, et al. Cellular markers of muscle atrophy in chronic obstructive pulmonary disease (COPD) American Journal of Respiratory Cellular Molecular Biology. 2009 Jun 11; doi: 10.1165/rcmb.2008-0382OC. 2009:2008-0382OC. [DOI] [PubMed] [Google Scholar]
  • 37.Hasselgren PO. Glucocorticoids and muscle catabolism. Current Opinion In Clinical Nutrition And Metabolic Care. 1999;2(3):201–5. doi: 10.1097/00075197-199905000-00002. [DOI] [PubMed] [Google Scholar]
  • 38.Emery PW, Edwards RHT, Rennie MJ. Protein synthesis in muscle measured in vivo in cachectic patients with cancer. British Medical Journal. 1984;289(6445):584–6. doi: 10.1136/bmj.289.6445.584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rennie MJ, Edwards RHT, Emery PW, Halliday D, Lundholm K, Millward DJ. Depressed protein synthesis is the dominant characteristic of muscle wasting and cachexia. Clinical Physiology. 1983;3(5):387–98. doi: 10.1111/j.1475-097x.1983.tb00847.x. [DOI] [PubMed] [Google Scholar]
  • 40.Lee SJ, Glass D. Treating cancer cachexia to treat cancer. Skeletal Muscle. 2011;1(1):2. doi: 10.1186/2044-5040-1-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rosenberg IH. Sarcopenia: origins and clinical relevance. Journal of Nutrition. 1997;127(5):990S. doi: 10.1093/jn/127.5.990S. [DOI] [PubMed] [Google Scholar]
  • 42.Tzankoff SP, Norris AH. Effect of muscle mass decrease on age-related BMR changes. Journal of Applied Physiology. 1977;43(6):1001–6. doi: 10.1152/jappl.1977.43.6.1001. [DOI] [PubMed] [Google Scholar]
  • 43.Marcell TJ. Review article: sarcopenia: causes, consequences, and preventions. Journal of Gerontology. 2003;58(10):M911–6. doi: 10.1093/gerona/58.10.m911. [DOI] [PubMed] [Google Scholar]
  • 44.Fry CS, Drummond MJ, Glynn EL, Dickinson JM, Gundermann DM, Timmerman KL, et al. Skeletal Muscle Autophagy and Protein Breakdown Following Resistance Exercise are Similar in Younger and Older Adults. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences. 2013;68(5):599–607. doi: 10.1093/gerona/gls209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kim JS, Kosek DJ, Petrella JK, Cross JM, Bamman MM. Resting and load-induced levels of myogenic gene transcripts differ between older adults with demonstrable sarcopenia and young men and women. Journal of Applied Physiology 2. 2005;99(6):2149–58. doi: 10.1152/japplphysiol.00513.2005. [DOI] [PubMed] [Google Scholar]
  • 46.Kosek DJ, Bamman MM. Modulation of the dystrophin-associated protein complex in response to resistance training in young and older men. Journal of Applied Physiology. 2008;104(5):1476–84. doi: 10.1152/japplphysiol.00708.2007. [DOI] [PubMed] [Google Scholar]
  • 47.Slivka D, Raue U, Hollon C, Minchev K, Trappe S. Single muscle fiber adaptations to resistance training in old (>80 yr) men: evidence for limited skeletal muscle plasticity. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2008;295(1):R273–80. doi: 10.1152/ajpregu.00093.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kumar V, Selby A, Rankin D, Patel R, Atherton P, Hildebrandt W, et al. Age-related differences in the dose–response relationship of muscle protein synthesis to resistance exercise in young and old men. The Journal of Physiology. 2009;587(1):211–7. doi: 10.1113/jphysiol.2008.164483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yang Y, Breen L, Burd NA, Hector AJ, Churchward-Venne TA, Josse AR, et al. Resistance exercise enhances myofibrillar protein synthesis with graded intakes of whey protein in older men. British Journal of Nutrition. 2012;108(10):1780–8. doi: 10.1017/S0007114511007422. [DOI] [PubMed] [Google Scholar]
  • 50.Belsh JM, Michael JA, Robert BD. Denervation Encyclopedy of the Neurological Sciences. New York: Academic Press; 2003. pp. 851–3. [Google Scholar]
  • 51.Martyn J, Fagerlungn JL, Eriksson I. Basic principles of neuromuscular transmission. Anaesthesia. 2009;64(s1):1–9. doi: 10.1111/j.1365-2044.2008.05865.x. [DOI] [PubMed] [Google Scholar]
  • 52.Schoser BGH, Wehling S, Blottner D. Cell death and apoptosis-related proteins in muscle biopsies of sporadic amyotrophic lateral sclerosis and polyneuropathy. Muscle & Nerve. 2001;24(8):1083–9. doi: 10.1002/mus.1114. [DOI] [PubMed] [Google Scholar]
  • 53.Allen JA, Stein R, Baker RA, Royden Jones H. Muscle atrophy associated with multiple sclerosis: A benign condition or the onset of amyotrophic lateral sclerosis? Journal of Clinical Neuroscience. 2008;15(6):706–8. doi: 10.1016/j.jocn.2007.04.024. [DOI] [PubMed] [Google Scholar]
  • 54.Zhou H, Huang C, Chen H, Wang D, Landel CP, Xia PY, et al. Transgenic rat model of neurodegeneration caused by mutation in the TDP gene. PLoS Genetics. 2010;6(3):e1000887. doi: 10.1371/journal.pgen.1000887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Gomes AV, Waddell DS, Siu R, Stein M, Dewey S, Furlow JD, et al. Upregulation of proteasome activity in muscle RING finger 1-null mice following denervation. The FASEB Journal. 2012 Jul 1;26(7):2986–99. doi: 10.1096/fj.12-204495. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sartori R, Schirwis E, Blaauw B, Bortolanza S, Zhao J, Enzo E, et al. BMP signaling controls muscle mass. Nat Genet. 2013;45(11):1309–18. doi: 10.1038/ng.2772. [DOI] [PubMed] [Google Scholar]
  • 57.Furlow JD, Watson ML, Waddell DS, Neff ES, Baehr LM, Ross AP, et al. Altered gene expression patterns in muscle ring finger 1 null mice during denervation- and dexamethasone-induced muscle atrophy. Physiological Genomics. 2013;45(23):1168–85. doi: 10.1152/physiolgenomics.00022.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ebert SM, Monteys AM, Fox DK, Bongers KS, Shields BE, Malmberg SE, et al. The transcription factor ATF4 promotes skeletal myofiber atrophy during fasting. Molecular endocrinology. 2010;24(4):790–9. doi: 10.1210/me.2009-0345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ebert SM, Dyle MC, Kunkel SD, Bullard SA, Bongers KS, Fox DK, et al. Stress-induced skeletal muscle Gadd45a expression reprograms myonuclei and causes muscle atrophy. The Journal of biological chemistry. 2012;287(33):27290–301. doi: 10.1074/jbc.M112.374777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Culp PA, Choi D, Zhang Y, Yin J, Seto P, Ybarra SE, et al. Antibodies to TWEAK Receptor Inhibit Human Tumor Growth through Dual Mechanisms. Clinical Cancer Research. 2010;16(2):497–508. doi: 10.1158/1078-0432.CCR-09-1929. [DOI] [PubMed] [Google Scholar]
  • 61.Anker SD, Coats AJS. Cardiac cachexia: A syndrome with impaired survival and immune and neuroendocrine activation. CHEST Journal. 1999;115(3):836–47. doi: 10.1378/chest.115.3.836. [DOI] [PubMed] [Google Scholar]
  • 62.Whitehouse AS, Tisdale MJ. Increased expression of the ubiquitin-proteasome pathway in murine myotubes by proteolysis-inducing factor (PIF) is associated with activation of the transcription factor NF-kappaB. British Journal Of Cancer. 2003;89(6):1116–22. doi: 10.1038/sj.bjc.6601132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Glass DJ. Skeletal muscle hypertrophy and atrophy signaling pathways. The International Journal Of Biochemistry & Cell Biology. 2005;37(10):1974–84. doi: 10.1016/j.biocel.2005.04.018. [DOI] [PubMed] [Google Scholar]
  • 64.Li H, Malhotra S, Kumar A. Nuclear factor-kappa B signaling in skeletal muscle atrophy. Journal of Molecular Medicine. 2008;86(10):1113–26. doi: 10.1007/s00109-008-0373-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mourkioti F, Rosenthal N. NF-kappaB signaling in skeletal muscle: prospects for intervention in muscle diseases. Journal Of Molecular Medicine. 2008;86(7):747–59. doi: 10.1007/s00109-008-0308-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Girgenrath M, Weng S, Kostek CA, Browning B, Wang M, Brown SAN, et al. TWEAK, via its receptor Fn14, is a novel regulator of mesenchymal progenitor cells and skeletal muscle regeneration. EMBO J. 2006;25(24):5826–39. doi: 10.1038/sj.emboj.7601441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mittal A, Bhatnagar S, Kumar A, Lach-Trifilieff E, Wauters S, Li H, et al. The TWEAK-Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice. Journal of Cell Biology. 2010;188(6):833–49. doi: 10.1083/jcb.200909117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mourkioti F, Kratsios P, Luedde T, Song YH, Delafontaine P, Adami R, et al. Targeted ablation of IKK2 improves skeletal muscle strength, maintains mass, and promotes regeneration. J Clin Invest. 2006;116(11):2945–54. doi: 10.1172/JCI28721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dogra C, Hall SL, Wedhas N, Linkhart TA, Kumar A. Fibroblast Growth Factor Inducible 14 (Fn14) Is Required for the Expression of Myogenic Regulatory Factors and Differentiation of Myoblasts into Myotubes: Evidence for TWEAK-independent functions of Fn14 during myogenesis. Journal of Biological Chemistry. 2007;282(20):15000–10. doi: 10.1074/jbc.M608668200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cai D, Frantz JD, Tawa NE, Melendez PA, Oh BC, Lidov HGW, et al. IKKbeta/NF-kappaB Activation Causes Severe Muscle Wasting in Mice. Cell. 2004;119(2):285–98. doi: 10.1016/j.cell.2004.09.027. [DOI] [PubMed] [Google Scholar]
  • 71.Wu CL, Jackman RW, Kandarian SC. NF-kappaB but not FoxO sites in the MuRF1 promoter are required for transcriptional activation in disuse muscle atrophy. American Journal of Physiology - Cell Physiology. 2014 doi: 10.1152/ajpcell.00361.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zhao W, Pan J, Wang X, Wu Y, Bauman WA, Cardozo CP. Expression of the muscle atrophy factor muscle atrophy F-Box is suppressed by testosterone. Endocrinology. 2008;149(11):5449–60. doi: 10.1210/en.2008-0664. [DOI] [PubMed] [Google Scholar]
  • 73.Heatwole CR, Eichinger KJ, Friedman DI, Hilbert JE, Jackson CE, Logigian EL, et al. Open-label trial of recombinant human insulin-like growth factor 1/recombinant human Insulin-like growth factor binding protein 3 in Myotonic Dystrophy Type 1. Archives of Neurology. 2011;68(1):37–44. doi: 10.1001/archneurol.2010.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hawley JA, Burke LM, Phillips SM, Spriet LL. Nutritional modulation of training-induced skeletal muscle adaptations. Journal of Applied Physiology. 2011 Mar 1;110(3):834–45. doi: 10.1152/japplphysiol.00949.2010. 2011. [DOI] [PubMed] [Google Scholar]
  • 75.Kubica N, Bolster DR, Farrell PA, Kimball SR, Jefferson LS. Resistance exercise increases muscle protein synthesis and translation of eukaryotic initiation factor 2Bepsilon mRNA in a mammalian target of rapamycin-dependent manner. Journal of Biological Chemistry. 2005;280(9):7570–80. doi: 10.1074/jbc.M413732200. [DOI] [PubMed] [Google Scholar]
  • 76.Guttridge DC. Signaling pathways weigh in on decisions to make or break skeletal muscle. Current Opinion In Clinical Nutrition And Metabolic Care. 2004;7(4):443–50. doi: 10.1097/01.mco.0000134364.61406.26. [DOI] [PubMed] [Google Scholar]
  • 77.Jackman RW, Kandarian SC. The molecular basis of skeletal muscle atrophy. American Journal of Physiology - Cell Physiology. 2004;287(4):C834–43. doi: 10.1152/ajpcell.00579.2003. [DOI] [PubMed] [Google Scholar]
  • 78.Kandarian SC, Jackman RW. Intracellular signaling during skeletal muscle atrophy. Muscle & Nerve. 2006;33(2):155–65. doi: 10.1002/mus.20442. [DOI] [PubMed] [Google Scholar]
  • 79.Bassel-Duby R, Olson EN. Signaling pathways in skeletal muscle remodeling. Annual Review Of Biochemistry. 2006;75:19–37. doi: 10.1146/annurev.biochem.75.103004.142622. [DOI] [PubMed] [Google Scholar]
  • 80.Vernace VA, Schmidt-Glenewinkel T, Figueiredo-Pereira ME. Aging and regulated protein degradation: who has the UPPer hand? Aging Cell. 2007;6(5):599–606. doi: 10.1111/j.1474-9726.2007.00329.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sandri M. Signaling in muscle atrophy and hypertrophy. Physiology. 2008;23:160–70. doi: 10.1152/physiol.00041.2007. [DOI] [PubMed] [Google Scholar]
  • 82.Gehrig SM, Lynch GS. Emerging drugs for treating skeletal muscle injury and promoting muscle repair. Expert Opinion on Emerging Drugs. 2011;16(1):163–82. doi: 10.1517/14728214.2010.524743. [DOI] [PubMed] [Google Scholar]
  • 83.Murphy KT, Lynch GS. Editorial update on emerging drugs for cancer cachexia. Expert Opinion on Emerging Drugs. 2012;0(0):1–5. doi: 10.1517/14728214.2012.652946. [DOI] [PubMed] [Google Scholar]
  • 84.Hood DA. Invited review: contractile activity-induced mitochondrial biogenesis in skeletal muscle. Journal of Applied Physiology. 2001;90(3):1137–57. doi: 10.1152/jappl.2001.90.3.1137. [DOI] [PubMed] [Google Scholar]
  • 85.Flück M, Hoppeler H. Molecular basis of skeletal muscle plasticity-from gene to form and function. Reviews of Physiology, Biochemistry and Pharmacology. 2003:159–216. doi: 10.1007/s10254-002-0004-7. [DOI] [PubMed] [Google Scholar]
  • 86.Raffaello A, Laveder P, Romualdi C, Bean C, Toniolo L, Germinario E, et al. Denervation in murine fast-twitch muscle: short-term physiological changes and temporal expression profiling. Physiological Genomics. 2006 Mar 13;25(1):60–74. doi: 10.1152/physiolgenomics.00051.2005. 2006. [DOI] [PubMed] [Google Scholar]
  • 87.Dupont-Versteegden EE, Nagarajan R, Beggs ML, Bearden ED, Simpson PM, Peterson CA. Identification of cold-shock protein RBM3 as a possible regulator of skeletal muscle size through expression profiling. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2008 Aug 27; doi: 10.1152/ajpregu.90455.2008. 2008:90455.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mahoney DJ, Safdar A, Parise G, Melov S, Fu M, MacNeil L, et al. Gene expression profiling in human skeletal muscle during recovery from eccentric exercise. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2008;294(6):R1901–10. doi: 10.1152/ajpregu.00847.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Oita RC, Mazzatti DJ, Lim FL, Powell JR, Merry BJ. Whole-genome microarray analysis identifies up-regulation of Nr4a nuclear receptors in muscle and liver from diet-restricted rats. Mechanisms of Ageing and Development. 2009;130(4):240–7. doi: 10.1016/j.mad.2008.12.004. [DOI] [PubMed] [Google Scholar]
  • 90.Welle S, Cardillo A, Zanche M, Tawil R. Skeletal muscle gene expression after myostatin knockout in mature mice. Physiological Genomics. 2009:54. doi: 10.1152/physiolgenomics.00054.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Navé BT, Ouwens M, Withers DJ, Alessi DR, Shepherd PR. Mammalian target of rapamycin is a direct target for protein kinase B: identification of a convergence point for opposing effects of insulin and amino-acid deficiency on protein translation. Biochem J. 1999;344(2):427–31. [PMC free article] [PubMed] [Google Scholar]
  • 92.Jacobs BL, You JS, Frey JW, Goodman CA, Gundermann DM, Hornberger TA. Eccentric Contractions Increase TSC2 Phosphorylation and Alter the Targeting of TSC2 and mTOR to the Lysosome. The Journal of physiology. 2013 doi: 10.1113/jphysiol.2013.256339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM. Ragulator-Rag Complex Targets mTORC1 to the Lysosomal Surface and Is Necessary for Its Activation by Amino Acids. Cell. 2010;141(2):290–303. doi: 10.1016/j.cell.2010.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Bar-Peled L, Schweitzer Lawrence D, Zoncu R, Sabatini David M. Ragulator Is a GEF for the Rag GTPases that Signal Amino Acid Levels to mTORC1. Cell. 2012;150(6):1196–208. doi: 10.1016/j.cell.2012.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Baar K, Esser K. Phosphorylation of p70S6kcorrelates with increased skeletal muscle mass following resistance exercise. American Journal of Physiology - Cell Physiology. 1999;276(1):C120–C7. doi: 10.1152/ajpcell.1999.276.1.C120. [DOI] [PubMed] [Google Scholar]
  • 96.Terzis G, Georgiadis G, Stratakos G, Vogiatzis I, Kavouras S, Manta P, et al. Resistance exercise-induced increase in muscle mass correlates with p70S6 kinase phosphorylation in human subjects. Eur J Appl Physiol. 2008 Jan;102(2):145–52. doi: 10.1007/s00421-007-0564-y. [DOI] [PubMed] [Google Scholar]
  • 97.Drummond MJ, Fry CS, Glynn EL, Dreyer HC, Dhanani S, Timmerman KL, et al. Rapamycin administration in humans blocks the contraction-induced increase in skeletal muscle protein synthesis. J Physiol. 2009;587(Pt 7):1535–46. doi: 10.1113/jphysiol.2008.163816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Dickinson JM, Fry CS, Drummond MJ, Gundermann DM, Walker DK, Glynn EL, et al. Mammalian target of rapamycin complex 1 activation is required for the stimulation of human skeletal muscle protein synthesis by essential amino acids. The Journal of nutrition. 2011 May;141(5):856–62. doi: 10.3945/jn.111.139485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Goodman CA, Frey JW, Mabrey DM, Jacobs BL, Lincoln HC, You JS, et al. The role of skeletal muscle mTOR in the regulation of mechanical load-induced growth. The Journal of physiology. 2011 Nov 15;589(Pt 22):5485–501. doi: 10.1113/jphysiol.2011.218255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Choi KM, McMahon LP, Lawrence JC., Jr Two motifs in the translational repressor PHAS-I required for efficient phosphorylation by mammalian target of rapamycin and for recognition by raptor. Journal of Biological Chemistry. 2003;278(22):19667–73. doi: 10.1074/jbc.M301142200. [DOI] [PubMed] [Google Scholar]
  • 101.Leger B, Cartoni R, Praz M, Lamon S, Doriaz O, Crettenand A, et al. Akt signalling through GSK-3beta, mTOR and Foxo1 is involved in human skeletal muscle hypertrophy and atrophy. The Journal of Physiology. 2006;576(Pt 3):923–33. doi: 10.1113/jphysiol.2006.116715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.McDunn J, Davis C, Muenzer J, Drewry A, Hotchkiss R. A peptide agonist of Akt-signaling abrogates sepsis-induced lymphocyte apoptosis. Shock. 2006;25(6):18. [Google Scholar]
  • 103.Gregorevic P, Plant DR, Lynch GS. Administration of insulin-like growth factor-I improves fatigue resistance of skeletal muscles from dystrophic ‘mdx’ mice. Muscle & Nerve. 2004;30(3):295–304. doi: 10.1002/mus.20082. [DOI] [PubMed] [Google Scholar]
  • 104.Abmayr S, Gregorevic P, Allen JM, Chamberlain JS. Phenotypic improvement of dystrophic muscles by rAAV/microdystrophin vectors is augmented by Igf1 codelivery. Molecular Therapy. 2005;12(3):441–50. doi: 10.1016/j.ymthe.2005.04.001. [DOI] [PubMed] [Google Scholar]
  • 105.Misra M, McGrane J, Miller KK, Goldstein MA, Ebrahimi S, Weigel T, et al. Effects of rhIGF-1 administration on surrogate markers of bone turnover in adolescents with anorexia nervosa. Bone. 2009;45(3):493–8. doi: 10.1016/j.bone.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Raoul C, Aebischer P. ALS, IGF-1 and gene therapy: It's never too late to mend. Gene Therapy. 2004;11(5):429–30. [Google Scholar]
  • 107.Franz CK, Federici T, Yang J, Backus C, Oh SS, Teng Q, et al. Intraspinal cord delivery of IGF-I mediated by adeno-associated virus 2 is neuroprotective in a rat model of familial ALS. Neurobiology of Disease. 2009;33(3):473–81. doi: 10.1016/j.nbd.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 108.Schmierer B, Hill CS. Kinetic analysis of Smad nucleocytoplasmic shuttling reveals a mechanism for transforming Growth Factor B-dependent nuclear accumulation of Smads. Molecular and Cellular Biology. 2005;25(22):9845–58. doi: 10.1128/MCB.25.22.9845-9858.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003 Jun 13;113(6):685–700. doi: 10.1016/s0092-8674(03)00432-x. [DOI] [PubMed] [Google Scholar]
  • 110.McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387(6628):83–90. doi: 10.1038/387083a0. [DOI] [PubMed] [Google Scholar]
  • 111.Massague J, Seoane J, Wotton D. Smad transcription factors. Genes & Development. 2005 Dec 1;19(23):2783–810. doi: 10.1101/gad.1350705. [DOI] [PubMed] [Google Scholar]
  • 112.McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proceedings of the National Academy of Sciences. 1997;94(23):12457–61. doi: 10.1073/pnas.94.23.12457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Jespersen J, Kjaer M, Schjerling P. The possible role of myostatin in skeletal muscle atrophy and cachexia. Scandinavan Journal of Medical Sciences and Sports. 2006 Apr;16(2):74–82. doi: 10.1111/j.1600-0838.2005.00498.x. [DOI] [PubMed] [Google Scholar]
  • 114.Wang M, Yu H, Kim YS, Bidwell CA, Kuang S. Myostatin facilitates slow and inhibits fast myosin heavy chain expression during myogenic differentiation. Biochemical and Biophysical Research Communications. 2012;426(1):83–8. doi: 10.1016/j.bbrc.2012.08.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Costelli P, Muscaritoli M, Bonetto A, Penna F, Reffo P, Bossola M, et al. Muscle myostatin signalling is enhanced in experimental cancer cachexia. European Journal Of Clinical Investigation. 2008;38(7):531–8. doi: 10.1111/j.1365-2362.2008.01970.x. [DOI] [PubMed] [Google Scholar]
  • 116.Wolfman NM, McPherron AC, Pappano WN, Davies MV, Song K, Tomkinson KN, et al. Activation of latent myostatin by the BMP-1/tolloid family of metalloproteinases. Proceedings of the National Academy of Sciences. 2003;100(26):15842–6. doi: 10.1073/pnas.2534946100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Bogdanovich S, Krag TOB, Barton ER, Morris LD, Whittemore LA, Ahima RS, et al. Functional improvement of dystrophic muscle by myostatin blockade. Nature. 2002;420(6914):418–21. doi: 10.1038/nature01154. [DOI] [PubMed] [Google Scholar]
  • 118.Bogdanovich S, McNally EM, Khurana TS. Myostatin blockade improves function but not histopathology in a murine model of limb-girdle muscular dystrophy 2C. Muscle & Nerve. 2008;37(3):308–16. doi: 10.1002/mus.20920. [DOI] [PubMed] [Google Scholar]
  • 119.Hill JJ, Qiu Y, Hewick RM, Wolfman NM. Regulation of Myostatin in vivo by growth and differentiation factor-associated Serum Protein-1: a novel protein with protease inhibitor and Follistatin domains. Molecular Endocrinology. 2003;17(6):1144–54. doi: 10.1210/me.2002-0366. [DOI] [PubMed] [Google Scholar]
  • 120.Hill JJ, Davies MV, Pearson AA, Wang JH, Hewick RM, Wolfman NM, et al. The Myostatin propeptide and the Follistatin-related gene are inhibitory binding proteins of Myostatin in normal serum. Journal of Biological Chemistry. 2002;277(43):40735–41. doi: 10.1074/jbc.M206379200. [DOI] [PubMed] [Google Scholar]
  • 121.Magee TR, Artaza JN, Ferrini MG, Vernet D, Zuniga FI, Cantini L, et al. Myostatin short interfering hairpin RNA gene transfer increases skeletal muscle mass. The Journal of Gene Medicine. 2006;8(9):1171–81. doi: 10.1002/jgm.946. [DOI] [PubMed] [Google Scholar]
  • 122.Massague J, Chen YG. Controlling TGF-B signaling. Genes & Development. 2000 Mar 15;14(6):627–44. 2000. [PubMed] [Google Scholar]
  • 123.Morine KJ, Bish LT, Selsby JT, Gazzara JA, Pendrak K, Sleeper MM, et al. Activin IIB receptor blockade attenuates dystrophic pathology in a mouse model of duchenne muscular dystrophy. Muscle & Nerve. 2010;42(5):722–30. doi: 10.1002/mus.21743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lin Z, Gao C, Ning Y, He X, Wu W, Chen YG. The pseudoreceptor BMP and Activin membrane-bound inhibitor positively modulates Wnt/B-Catenin signaling. Journal of Biological Chemistry. 2008;283(48):33053–8. doi: 10.1074/jbc.M804039200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Yan X, Lin Z, Chen F, Zhao X, Chen H, Ning Y, et al. Human BAMBI cooperates with Smad7 to inhibit transforming growth factor-B signaling. Journal of Biological Chemistry. 2009;284(44):30097–104. doi: 10.1074/jbc.M109.049304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hashimoto O, Kawasaki N, Tsuchida K, Shimasaki S, Hayakawa T, Sugino H. Difference between follistatin isoforms in the inhibition of activin signalling: Activin neutralizing activity of follistatin isoforms is dependent on their affinity for activin. Cellular Signalling. 2000;12(8):565–71. doi: 10.1016/s0898-6568(00)00099-1. [DOI] [PubMed] [Google Scholar]
  • 127.Milazzotto MP, Goissis MD, Feitosa WB, Martins LF, Strauss BE, Bajgelman MC, et al. Myostatin gene knockdown through lentiviral-mediated delivery of shRNA for in vitro production of transgenic bovine embryos. Zygote. 2010;18(04):339–44. doi: 10.1017/S0967199410000031. [DOI] [PubMed] [Google Scholar]
  • 128.Kang JK, Malerba A, Popplewell L, Foster K, Dickson G. Antisense-induced Myostatin Exon Skipping Leads to Muscle Hypertrophy in Mice Following Octa guanidine Morpholino Oligomer Treatment. Mol Ther. 2011 doi: 10.1038/mt.2010.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sutrave P, Kelly AM, Hughes SH. Ski Can Cause Selective Growth of Skeletal Muscle in Transgenic Mice. Genes & development. 1990 Sep;4(9):1462–72. doi: 10.1101/gad.4.9.1462. [DOI] [PubMed] [Google Scholar]
  • 130.MacKenzie MG, Hamilton DL, Pepin M, Patton A, Baar K. Inhibition of Myostatin Signaling through Notch Activation following Acute Resistance Exercise. PLoS ONE. 2013;8(7):e68743. doi: 10.1371/journal.pone.0068743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sartori R, Schirwis E, Blaauw B, Bortolanza S, Zhao J, Enzo E, et al. BMP signaling controls muscle mass. Nature genetics. 2013;45(11):1309–18. doi: 10.1038/ng.2772. [DOI] [PubMed] [Google Scholar]
  • 132.Welle S, Burgess K, Mehta S. Stimulation of skeletal muscle myofibrillar protein synthesis, p70 S6 kinase phosphorylation, and ribosomal protein S6 phosphorylation by inhibition of myostatin in mature mice. American Journal of Physiology - Endocrinology And Metabolism. 2009 Mar 1;296(3):E567–72. doi: 10.1152/ajpendo.90862.2008. 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Bogdanovich S, Perkins KJ, Krag TOB, Whittemore LA, Khurana TS. Myostatin propeptide-mediated amelioration of dystrophic pathophysiology. The FASEB Journal. 2005 Apr 1;19(6):543–9. doi: 10.1096/fj.04-2796com. 2005. [DOI] [PubMed] [Google Scholar]
  • 134.Benabdallah BF, Bouchentouf M, Rousseau J, Bigey P, Michaud A, Chapdelaine P, et al. Inhibiting myostatin with follistatin improves the success of myoblast transplantation in dystrophic mice. Cell Transplantation. 2008;17(3):337–50. doi: 10.3727/096368908784153913. [DOI] [PubMed] [Google Scholar]
  • 135.Morine KJ, Bish LT, Pendrak K, Sleeper MM, Barton ER, Sweeney HL. Systemic Myostatin inhibition via liver-targeted gene transfer in normal and dystrophic mice. PLoS ONE. 2010;5(2):E9176–E. doi: 10.1371/journal.pone.0009176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Morrison BM, Lachey JL, Warsing LC, Ting BL, Pullen AE, Underwood KW, et al. A soluble activin type IIB receptor improves function in a mouse model of amyotrophic lateral sclerosis. Experimental Neurology. 2009;217(2):258–68. doi: 10.1016/j.expneurol.2009.02.017. [DOI] [PubMed] [Google Scholar]
  • 137.Benny Klimek ME, Aydogdu T, Link MJ, Pons M, Koniaris LG, Zimmers TA. Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia. Biochemical & Biophysical Research Communications. 2010;391(3):1548–54. doi: 10.1016/j.bbrc.2009.12.123. [DOI] [PubMed] [Google Scholar]
  • 138.Murphy KT, Chee A, Gleeson BG, Naim T, Swiderski K, Koopman R, et al. Antibody-directed myostatin inhibition enhances muscle mass and function in tumor-bearing mice. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2011;301(3):R716–R26. doi: 10.1152/ajpregu.00121.2011. [DOI] [PubMed] [Google Scholar]
  • 139.Fakhfakh R, Michaud A, Tremblay JP. Blocking the myostatin signal with a dominant negative receptor improves the success of human myoblast transplantation in dystrophic mice. Molecular Therapy. 2011;19(1):204–10. doi: 10.1038/mt.2010.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ruas JL, White JP, Rao RR, Kleiner S, Brannan KT, Harrison BC, et al. A PGC-1alpha isoform induced by resistance training regulates skeletal muscle hypertrophy. Cell. 2012;151(6):1319–31. doi: 10.1016/j.cell.2012.10.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Perez-Schindler J, Summermatter S, Santos G, Zorzato F, Handschin C. The transcriptional coactivator PGC-1alpha is dispensable for chronic overload-induced skeletal muscle hypertrophy and metabolic remodeling. Proc Natl Acad Sci U S A. 2013;110(50):20314–9. doi: 10.1073/pnas.1312039110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Emery PW, Rothwell NJ, Stock MJ, Winter PD. Chronic effects of beta 2-adrenergic agonists on body composition and protein synthesis in the rat. Bioscience reports. 1984 Jan;4(1):83–91. doi: 10.1007/BF01120827. [DOI] [PubMed] [Google Scholar]
  • 143.von Maltzahn J, Bentzinger CF, Rudnicki MA. Wnt7a-Fzd7 signalling directly activates the Akt/mTOR anabolic growth pathway in skeletal muscle. Nature cell biology. 2012 Feb;14(2):186–91. doi: 10.1038/ncb2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Porporato PE, Filigheddu N, Reano S, Ferrara M, Angelino E, Gnocchi VF, et al. Acylated and unacylated ghrelin impair skeletal muscle atrophy in mice. The Journal of clinical investigation. 2013 Feb 1;123(2):611–22. doi: 10.1172/JCI39920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Minetti GC, Feige JN, Rosenstiel A, Bombard F, Meier V, Werner A, et al. Galphai2 signaling promotes skeletal muscle hypertrophy, myoblast differentiation, and muscle regeneration. Science signaling. 2011;4(201):RA80. doi: 10.1126/scisignal.2002038. [DOI] [PubMed] [Google Scholar]
  • 146.Ryall JG, Sillence MN, Lynch GS. Systemic administration of beta2-adrenoceptor agonists, formoterol and salmeterol, elicit skeletal muscle hypertrophy in rats at micromolar doses. British journal of pharmacology. 2006 Mar;147(6):587–95. doi: 10.1038/sj.bjp.0706669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Kline WO, Panaro FJ, Yang H, Bodine SC. Rapamycin inhibits the growth and muscle-sparing effects of clenbuterol. J Appl Physiol. 2007 Feb;102(2):740–7. doi: 10.1152/japplphysiol.00873.2006. [DOI] [PubMed] [Google Scholar]
  • 148.Soppa GKR, Smolenski RT, Latif N, Yuen AHY, Malik A, Karbowska J, et al. Effects of chronic administration of clenbuterol on function and metabolism of adult rat cardiac muscle. American Journal of Physiology - Heart and Circulatory Physiology. 2005;288(3):H1468–H76. doi: 10.1152/ajpheart.00624.2004. [DOI] [PubMed] [Google Scholar]
  • 149.Burks TN, Andres-Mateos E, Marx R, Mejias R, Van Erp C, Simmers JL, et al. Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Science Translational Medicine. 2011;3(82):82RA37. doi: 10.1126/scitranslmed.3002227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tries RS, Chen T, Da Vies MV, Tomkinson KN, Pearson AA, Shakey QA, et al. Gdf-8 propeptide binds to GDF-8 and antagonizes biological activity by inhibiting GDF-8 receptor binding. Growth Factors. 2001;18(4):251–9. doi: 10.3109/08977190109029114. [DOI] [PubMed] [Google Scholar]
  • 151.Hu S, Chen C, Sheng J, Sun Y, Cao X, Qiao J. Enhanced muscle growth by plasmid-mediated delivery of myostatin propeptide. Journal of Biomedicine and Biotechnology. 2010;2010 doi: 10.1155/2010/862591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Cadena SM, Tomkinson KN, Monnell TE, Spaits MS, Kumar R, Underwood KW, et al. Administration of a soluble activin type IIB receptor promotes skeletal muscle growth independent of fiber type. Journal of Applied Physiology. 2010 Sep 1;109(3):635–42. doi: 10.1152/japplphysiol.00866.2009. 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lawlor MW, Read BP, Edelstein R, Yang N, Pierson CR, Stein MJ, et al. Inhibition of Activin Receptor Type IIB increases strength and lifespan in Myotubularin-deficient mice. The American Journal Of Pathology. 2011;178(2):784–93. doi: 10.1016/j.ajpath.2010.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lee SJ. Quadrupling muscle mass in mice by targeting TGF-ß signaling pathways. PLoS ONE. 2007;2(8):E789. doi: 10.1371/journal.pone.0000789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Lee SJ, McPherron AC. Regulation of myostatin activity and muscle growth. Proceedings of the National Academy of Sciences. 2001 Jul 31;98(16):9306–11. doi: 10.1073/pnas.151270098. 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Nakatani M, Takehara Y, Sugino H, Matsumoto M, Hashimoto O, Hasegawa Y, et al. Transgenic expression of a myostatin inhibitor derived from follistatin increases skeletal muscle mass and ameliorates dystrophic pathology in mdx mice. The FASEB Journal. 2008;22(2):477–87. doi: 10.1096/fj.07-8673com. [DOI] [PubMed] [Google Scholar]
  • 157.Kota J, Handy CR, Haidet AM, Montgomery CL, Eagle A, Rodino-Klapac LR, et al. Follistatin gene delivery enhances muscle growth and strength in nonhuman primates. Science Translational Medicine. 2009;1(6):6RA15. doi: 10.1126/scitranslmed.3000112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Akpan I, Goncalves MD, Dhir R, Yin X, Pistilli EE, Bogdanovich S, et al. The effects of a soluble activin type IIB receptor on obesity and insulin sensitivity. International Journal of Obesity. 2009;33(11):1265–73. doi: 10.1038/ijo.2009.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Koncarevic A, Kajimura S, Cornwall-Brady M, Andreucci A, Pullen A, Sako D, et al. A Novel Therapeutic Approach to Treating Obesity through Modulation of TGFB Signaling. Endocrinology. 2012 Jul 1;153(7):3133–46. doi: 10.1210/en.2012-1016. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Nagler A, Ohana M, Leiba M, Levdansky L, Gorodetsky R. Effect of halofuginone, a collagen A1(I) inhibitor, on wound healing in normal and irradiated skin: Implication for hematopoietic stem cell transplantation. Acta Haematologica: Acta Haematologica. 2007 doi: 10.1159/000104897. [DOI] [PubMed] [Google Scholar]
  • 161.McGaha TL, Phelps RG, Spiera H, Bona C. Halofuginone, an inhibitor of type-I collagen synthesis and skin sclerosis, blocks transforming-growth-factor-beta-mediated Smad3 activation in fibroblasts. Journal Of Investigative Dermatology. 2002 doi: 10.1046/j.0022-202x.2001.01690.x. [DOI] [PubMed] [Google Scholar]
  • 162.Turgeman T, Hagai Y, Huebner K, Jassal DS, Anderson JE, Genin O, et al. Prevention of muscle fibrosis and improvement in muscle performance in the mdx mouse by halofuginone. Neuromuscular disorders: NMD. 2008;18(11):857–68. doi: 10.1016/j.nmd.2008.06.386. [DOI] [PubMed] [Google Scholar]
  • 163.Huebner KD, Jassal DS, Halevy O, Pines M, Anderson JE. Functional resolution of fibrosis in mdx mouse dystrophic heart and skeletal muscle by halofuginone. American Journal of Physiology - Heart and Circulatory Physiology. 2008 Apr 01;294(4):H1550–H61. doi: 10.1152/ajpheart.01253.2007. 2008. 16:09:07. [DOI] [PubMed] [Google Scholar]
  • 164.Pines M, H O. Halofuginone and muscular dystrophy. Histol Histopathol. 2011 doi: 10.14670/HH-26.135. [DOI] [PubMed] [Google Scholar]
  • 165.Neilan TG, Jassal DS, Perez-Sanz TM, Raher MJ, Pradhan AD, Buys ES, et al. Tissue Doppler imaging predicts left ventricular dysfunction and mortality in a murine model of cardiac injury. European Heart Journal. 2006 Aug 1;27(15):1868–75. doi: 10.1093/eurheartj/ehl013. 2006. [DOI] [PubMed] [Google Scholar]
  • 166.Roffe S, Hagai Y, Pines M, Halevy O. Halofuginone inhibits Smad3 phosphorylation via the PI3K/Akt and MAPK/ERK pathways in muscle cells: Effect on myotube fusion. Experimental Cell Research. 2010;316(6):1061–9. doi: 10.1016/j.yexcr.2010.01.003. 4/1. [DOI] [PubMed] [Google Scholar]
  • 167.Winbanks CE, Chen JL, Qian H, Liu Y, Bernardo BC, Beyer C, et al. The bone morphogenetic protein axis is a positive regulator of skeletal muscle mass. The Journal of Cell Biology. 2013 Oct 28;203(2):345–57. doi: 10.1083/jcb.201211134. 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Mohler ML, Bohl CE, Jones A, Coss CC, Narayanan R, He Y, et al. Nonsteroidal Selective Androgen Receptor Modulators (SARMs): Dissociating the Anabolic and Androgenic Activities of the Androgen Receptor for Therapeutic Benefit. Journal of Medicinal Chemistry. 2009 Jun 25;52(12):3597–617. doi: 10.1021/jm900280m. 2009. [DOI] [PubMed] [Google Scholar]
  • 169.Yin D, Gao W, Kearbey JD, Xu H, Chung K, He Y, et al. Pharmacodynamics of Selective Androgen Receptor Modulators. Journal of Pharmacology and Experimental Therapeutics. 2003 Mar 1;304(3):1334–40. doi: 10.1124/jpet.102.040840. 2003. [DOI] [PubMed] [Google Scholar]
  • 170.Kicman AT. Pharmacology of anabolic steroids. British Journal of Pharmacology. 2008;154(3):502–21. doi: 10.1038/bjp.2008.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Bhasin S, Jasuja R. Selective androgen receptor modulators as function promoting therapies. Current Opinion in Clinical Nutrition & Metabolic Care. 2009;12(3):232–40. doi: 10.1097/MCO.0b013e32832a3d79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Cirak S, Arechavala-Gomeza V, Guglieri M, Feng L, Torelli S, Anthony K, et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. The Lancet. 2011;378(9791):595–605. doi: 10.1016/S0140-6736(11)60756-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Jarmin S, Kymalainen H, Popplewell L, Dickson G. New developments in the use of gene therapy to treat Duchenne muscular dystrophy. Expert Opinion on Biological Therapy. 2014;14(2):209–30. doi: 10.1517/14712598.2014.866087. [DOI] [PubMed] [Google Scholar]
  • 174.Long C, McAnally JR, Shelton JM, Mireault AA, Bassel-Duby R, Olson EN. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science. 2014;345(6201):1184–8. doi: 10.1126/science.1254445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Okada T, Ichi Takeda S. Current Challenges and Future Directions in Recombinant AAV-Mediated Gene Therapy of Duchenne Muscular Dystrophy. Pharmaceuticals. 2013;6(7):813–36. doi: 10.3390/ph6070813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Gregorevic P, Allen JM, Minami E, Blankinship MJ, Haraguchi M, Meuse L, et al. Raav6-microdystrophin Preserves Muscle Function and Extends Lifespan in Severely Dystrophic Mice. Nature Medicine. 2006;12(7):787–9. doi: 10.1038/nm1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Gregorevic P, Blankinship MJ, Allen JM, Chamberlain JS. Systemic Microdystrophin Gene Delivery Improves Skeletal Muscle Structure and Function in Old Dystrophic Mdx Mice. Molecular Therapy. 2008;16(4):657–64. doi: 10.1038/mt.2008.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Odom GL, Gregorevic P, Allen JM, Finn E, Chamberlain JS. Microutrophin Delivery through Raav6 Increases Lifespan and Improves Muscle Function in Dystrophic Dystrophin/utrophin-deficient Mice. Molecular Therapy. 2008;16(9):1539–45. doi: 10.1038/mt.2008.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tramontana JM, U U, Molloy A, Akarasewi P, Burroughs M, Makonkawkeyoon S, Johnson B, Klausner JD, Rom W, Kaplan G. Thalidomide treatment reduces tumor necrosis factor alpha production and enhances weight gain in patients with pulmonary tuberculosis. Mol Med. 1995;1(4):384–97. [PMC free article] [PubMed] [Google Scholar]
  • 180.Gordon JN, Trebble TM, Ellis RD, Duncan HD, Johns T, Goggin PM. Thalidomide in the treatment of cancer cachexia: a randomised placebo controlled trial. Gut. 2005 Apr 1;54(4):540–5. doi: 10.1136/gut.2004.047563. 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Fearon KC, Von Meyenfeldt MF, Moses AG, Van Geenen R, Roy A, Gouma DJ, et al. Effect of a protein and energy dense N-3 fatty acid enriched oral supplement on loss of weight and lean tissue in cancer cachexia: a randomised double blind trial. Gut. 2003 Oct;52(10):1479–86. doi: 10.1136/gut.52.10.1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Fearon KC, Barber MD, Moses AG, Ahmedzai SH, Taylor GS, Tisdale MJ, et al. Double-blind, placebo-controlled, randomized study of eicosapentaenoic acid diester in patients with cancer cachexia. J Clin Oncol. 2006 Jul 20;24(21):3401–7. doi: 10.1200/JCO.2005.04.5724. [DOI] [PubMed] [Google Scholar]
  • 183.Van Norren K, Kegler D, Argiles JM, Luiking Y, Gorselink M, Laviano A, et al. Dietary supplementation with a specific combination of high protein, leucine, and fish oil improves muscle function and daily activity in tumour-bearing cachectic mice. Br J Cancer. 2009 doi: 10.1038/sj.bjc.6604905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Argilés JM, Busquets S, López-Soriano FJ. Anti-inflammatory therapies in cancer cachexia. European Journal of Pharmacology. 2011;668(Supplement 1)(0):S81–S6. doi: 10.1016/j.ejphar.2011.07.007. [DOI] [PubMed] [Google Scholar]
  • 185.Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell. 2010;142(4):531–43. doi: 10.1016/j.cell.2010.07.011. [DOI] [PubMed] [Google Scholar]
  • 186.Dobs AS, Boccia RV, Croot CC, Gabrail NY, Dalton JT, Hancock ML, et al. Effects of enobosarm on muscle wasting and physical function in patients with cancer: a double-blind, randomised controlled phase 2 trial. The Lancet Oncology. 2013;14(4):335–45. doi: 10.1016/S1470-2045(13)70055-X. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES