Abstract

Chronic kidney disease (CKD) patients often exhibit a low muscle mass and strength, leading to physical impairment and an increased mortality. Two major signalling pathways control protein synthesis, the insulin-like growth factor-1/Akt (IGF-1/Akt) pathway, acting as a positive regulator, and the myostatin (Mstn) pathway, acting as a negative regulator. Mstn, also known as the growth development factor-8 (GDF-8), is a member of the transforming growth factor-β superfamily, which is secreted by mature muscle cells. Mstn inhibits satellite muscle cell proliferation and differentiation and induces a proteolytic phenotype of muscle cells by activating the ubiquitin–proteasome system. Recent advances have been made in the comprehension of the Mstn pathway disturbance and its role in muscle wasting during CKD. Most studies report higher Mstn concentrations in CKD and dialysis patients than in healthy subjects. Several factors increase Mstn production in uraemic conditions: low physical activity, chronic or acute inflammation and oxidative stress, uraemic toxins, angiotensin II, metabolic acidosis and glucocorticoids. Mstn seems to be only scarcely removed during haemodialysis or peritoneal dialysis, maybe because of its large molecule size in plasma where it is linked to its prodomain. In dialysis patients, Mstn has been proposed as a biomarker of muscle mass, muscle strength or physical performances, but more studies are needed in this field. This review outlines the interconnection between Mstn activation, muscle dysfunction and CKD. We discuss mechanisms of action and efficacy of pharmacological Mstn pathway inhibition that represents a promising treatment approach of striated muscle dysfunction. Many approaches and molecules are in development but until now, no study has proved a benefit in CKD.

INTRODUCTION

The association of skeletal muscle atrophy and impaired muscle function, named sarcopenia, is associated with lower health-related quality of life, and greater disability and mortality risk in chronic kidney disease (CKD) patients [1, 2]. Its prevalence is estimated to be between 12% and 29% and it represents an important medico-economic burden [3, 4].

In healthy adults, a constant muscle mass is maintained through the balance of the insulin-like growth factor-1/Akt (IGF-1/Akt) pathway inducing protein synthesis and recruitment of muscle satellite cells on one hand, and the myostatin (Mstn) pathway inducing protein breakdown by activation of the ubiquitin–proteasome pathway and caspase3 as well as inhibiting satellite cell recruitment on the other hand [5–7]. These two pathways are highly regulated and interact with one another. Muscle loss comes from a disequilibrium between these two systems [8].

A better understanding of this system in CKD-associated muscle dysfunction is crucial. Recent advances in the role of Mstn as well as development of potential therapeutic molecules give rise to great hope for future treatments.

MOLECULAR PATHWAYS OF MUSCLE MASS REGULATION

The main muscle growth pathway is the IGF-1/Akt (Figure 1). Activation of this signalling pathway is initiated by the interaction of IGF-1—a hepatic synthetized growth factor after stimulation of the growth hormone (GH)—with its receptor (IGF-1R) at muscle cell surface [9]. The binding of IGF-1 to its receptor leads to Akt phosphorylation into p-Akt. Akt, also called protein kinase B, regulates more than 2000 genes of mammalian target of rapamycin complex 1 (mTORC1). mTORC1 and its downstream effectors are an important protein synthesis pathway. In muscle cells, it can be activated by p-Akt as well as by other mechanical or hormonal stimuli in order to activate muscle growth.

Activation and signalling pathway explaining muscle wasting and cachexia in patients with CKD. CKD is associated with an increase in circulating inflammatory cytokines, uraemic toxins and oxidative stress combined with reduction of physical activity, which sets the stage for the activation of Mstn. Mstn binds to ActRIIB/IIB on muscle cell membrane. Subsequent phosphorylation of Smad2/3 leads to its binding with Smad4 and translocation of the complex to the nucleus, where it blocks the transcription of genes responsible for the myogenesis. Mstn also reduces Akt activity and decreases FoxO phosphorylation. Dephosphorylated FoxO enters the nucleus to activate transcription of MuRF1 and Atrogin-1, which cause muscle protein ubiquitination and degradation by the proteasome and autophagy. Mstn also active p38/caspase pathways that are involved in apoptosis. Mstn also suppresses the mTORC1 pathway and protein synthesis. This leads to muscle wasting and cachexia. ActRIIA, activin receptor type IIA; ActRIIB, activin receptor type IIB; ALK, anaplastic lymphoma receptor tyrosine kinase; eIF4G, eukaryotic translation initiation factor 4 G; IL, interleukin; JAK, Janus kinase; mTORC1, mammalian target of rapamycin complex 1; P, phosphorylation; SMAD, Sma- and Mad-related protein; TNF-α, tumor necrosis factor alpha.
FIGURE 1

Activation and signalling pathway explaining muscle wasting and cachexia in patients with CKD. CKD is associated with an increase in circulating inflammatory cytokines, uraemic toxins and oxidative stress combined with reduction of physical activity, which sets the stage for the activation of Mstn. Mstn binds to ActRIIB/IIB on muscle cell membrane. Subsequent phosphorylation of Smad2/3 leads to its binding with Smad4 and translocation of the complex to the nucleus, where it blocks the transcription of genes responsible for the myogenesis. Mstn also reduces Akt activity and decreases FoxO phosphorylation. Dephosphorylated FoxO enters the nucleus to activate transcription of MuRF1 and Atrogin-1, which cause muscle protein ubiquitination and degradation by the proteasome and autophagy. Mstn also active p38/caspase pathways that are involved in apoptosis. Mstn also suppresses the mTORC1 pathway and protein synthesis. This leads to muscle wasting and cachexia. ActRIIA, activin receptor type IIA; ActRIIB, activin receptor type IIB; ALK, anaplastic lymphoma receptor tyrosine kinase; eIF4G, eukaryotic translation initiation factor 4 G; IL, interleukin; JAK, Janus kinase; mTORC1, mammalian target of rapamycin complex 1; P, phosphorylation; SMAD, Sma- and Mad-related protein; TNF-α, tumor necrosis factor alpha.

Besides its central role in proteo-synthesis activation, p-Akt also suppresses the proteolytic cell activity by inactivating and maintaining in the cytoplasm the forkhead box O (FoxO) transcription factors resulting in the ubiquitin–proteasome system (UPS) as well as the autophagy–lysosomal proteolytic pathways activation [10].

On the other side, the main negative regulator of muscle cells is the Mstn pathway, which activates the UPS in mature muscle cells and inhibits muscle satellite cell recruitment (Figure 1). Mstn, or growth development factor-8 (GDF-8), is a member of the transforming growth factor-β (TGF-β) superfamily. It is mainly secreted by mature muscle cells, and to a lesser extent in smooth muscle cells, heart, adipocytes, mammary glands and haematopoietic stem cells [11]. Mstn is produced by muscle cells as a pre-pro-myostatin, then secreted in the intercellular space and blood as pro-myostatin. After a two-step cleavage from its prodomain, called latency-associated protein (LAP) in its active dimeric form, Mstn acts as a potent autocrine, paracrine and endocrine negative regulator of muscle growth [11].

At muscle cell surface, Mstn binds to its receptor activin type II B receptor (ActRIIB) leading to phosphorylation and activation of transcription factors Smad2 and Smad3 [12] (Figure 1). The Smads are then imported into the nucleus where they modulate transcription of target genes. They increase the transcription of Atrogin-1 (or muscle atrophy F-box, MAFbx) and muscle RING finger 1 (Murf-1), two E3 ubiquitin ligases, stimulating protein breakdown via the UPS [13]. Smad2 and -3 also inhibit Akt phosphorylation and the Janus kinase/Stat pathway [12]. Other members of the TGF-β superfamily such as Activin A or GDF-11 are also able to bind to ActRIIB and induce the same cell signal. Mstn is not the only negative regulator of muscle mass during CKD. Metabolic acidosis or endogenous glucocorticoids inhibit phosphoinositide 3-kinase activation and therefore Akt phosphorylation [14, 15]. Inflammation as well, and notably interleukin (IL)-6, inhibits muscle protein synthesis via Stat3 activation and possibly increases Mstn transcription [16, 17].

Satellite muscle cells are partially differentiated quiescent stem cells living at the periphery of muscle fibres. When muscle fibres are wounded, satellite cells activated by growth factors proliferate, differentiate and fuse, leading to new muscle fibers. Alteration of the IGF-1/Akt pathway as well as binding of Mstn to satellite cells inhibit their proliferation and differentiation [18].

ANATOMIC AND METABOLIC EFFECTS OF MYOSTATIN

The numerous natural or experimental Mstn knock-out animal models of cattle, mice or dog exert an impressive muscle mass increase and confirm the relationship between Mstn and muscle growth inhibition [19]. Interestingly, muscle strength and physical performance of these models are proportionally less increased than their muscle mass, and are associated with earlier muscle fatigue. This fatigue is linked to a shift of an aerobic to anaerobic energetic metabolism and consequently lactic acid production during exercise. In human, an observation of a loss-of-function mutation in the Mstn gene has been reported in a 4-year-old boy who showed a significant muscle hypertrophy since birth [20]. Interestingly, his heterozygous mother also had a high muscle mass, but not as high as his son. Mstn polymorphisms have been associated with muscle strength in human [21].

Opposite this, administration of Mstn or Activin A in mice induces a 30% muscle mass reduction. Mstn is increased in various animal models of muscle dystrophy, cachexia and immobilization. In human, Mstn expression increases with age. In sarcopenic elderly, Mstn is positively correlated with muscle atrophy. This correlation is also found in young individuals after immobilization as well as in several pathologic states: corticosteroid therapy, cancer, acquired immunodeficiency syndrome, heart failure and chronic obstructive pulmonary disease [22].

Besides its negative regulator of muscle growth, Mstn also interferes with several metabolic pathways including lipid storage, insulin sensitivity and thermogenic regulation. Mstn is upregulated in obese patients and correlates with insulin resistance [23]. Bariatric surgery lowers Mstn expression and improves insulin sensitivity. These results suggest that the association of sarcopenia, obesity and type II diabetes could constitute a target for Mstn inhibitors.

Insulin sensitivity is not only due to an increase in muscle mass. The phosphorylation of Akt increases the muscle fibre expression of glucose transporters GLUT-1 and GLUT-4, and glycogen synthesis, improving glucose muscle uptake [24]. These observations are supplemental arguments to developing Mstn inhibitors to increase insulin sensitivity.

MYOSTATIN DURING CKD

Low physical activity, chronic or acute inflammation and oxidative stress, accumulation of uraemic toxins, angiotensin II, metabolic acidosis and glucocorticoids observed in CKD patients are all factors that may contribute to increase in Mstn production [8, 25] (Figure 1); although, limited data are available on the role of Mstn upregulation in the alterations in skeletal muscle mass in the field of CKD. In human, most data are observational and only few animal models have been studied.

In a rat subtotal nephrectomy CKD model, muscle Mstn mRNA was increased compared with pair-fed rats and, after 2–7 days of exercise returned to normal values. At the same time, muscle mass and IGF-1 mRNA were increased [26]. In a 5/6 nephrectomy mice model, CKD mice display higher IL-6, Mstn and Atrogin-1 mRNA in gastrocnemius muscle and lower Akt phosphorylation [27]. The same team reported in vitro in myotubes that, indoxyl sulfate(IS), an indolic uraemic toxin, was involved in the expression of IL-6, Mstn and Atrogin-1. They confirmed these results in vivo and chronic treatment of IS in half-nephrectomized mice was able to increase the same genes in muscle, but curiously they did not find any change in Akt phosphorylation [28]. These data suggest that IS could upregulate Mstn secretion and be a major player of muscle dysfunction during CKD, but this link must be confirmed in CKD patients.

In human, a vastus lateralis transcriptome study performed in five haemodialysis (HD) patients before their dialysis session did not report any change in Mstn or Atrogene mRNA expression compared with healthy subjects [29]. Another study performed in 51 younger (mean age 42 years old) HD patients did not show any increase in Mstn mRNA in vastus lateralis compared with healthy subjects [30]. Nevertheless, in rectus abdominis muscle biopsies performed at the time of peritoneal catheter insertion in 22 CKD Stage 5 patients, Verzola et al. reported that Mstn mRNA was 10 times higher than in 22 paired healthy subjects and Mstn protein concentration 4 times higher [31]. Enzyme-linked immunosorbent assay (ELISA) and western blot can be performed to measure tissue Mstn concentrations, but as for plasma Mstn dosing, the specificity of available antibodies for each of the Mstn forms (active Mstn, monomer or dimer form, pro-myostatin) is not so clear. Overall, muscle tissue Mstn concentration seems to be increased during CKD, at least in some patients, but more studies are needed to better describe how Mstn acts as a local signal to promote atrophy.

Inflammation could be the trigger of Mstn overexpression during CKD. Indeed, Verzola et al. observed that CKD patients had a higher IL-6 mRNA in muscle and that Mstn gene overexpression was correlated with inflammation parameters in this population [31]. In good concordance, Zhang et al. reported high levels of pro-inflammatory cytokines including IL-6 in muscle biopsies of 18 CKD patients, which activate transcription factors including Stat3. Stat3 expression is increased in animals and human during CKD and upregulate Mstn gene expression. Interestingly, Stat3 knockout mice in muscle exert no inflammation and no muscle atrophy after CKD induction [16]. Physical activity could also be a clue: a study in HD reported a 50% decrease in Mstn mRNA in vastus lateralis muscle after only 9 weeks of endurance exercise training [32].

In human, the few studies having reported plasma concentrations of Mstn during CKD are described in Table 1. Concentrations and results are highly influenced by the dosing technique, age and gender. Nevertheless, most studies report higher Mstn concentrations in CKD, HD and peritoneal dialysis (PD) patients than in healthy subjects.

Table 1

Studies of myostatin plasma concentrations in CKD and dialysis patients

PopulationAssay techniquesMstn concentration in populationResultsStudy
Non-dialysis CKD
93 CKD patientsR&D Systems, Abingdon, UK3.34 ng/mL
  • Negative correlation with

  • eGFR (β = −0.31, P < 0.01) and ClCr (β = −0.35, P < 0.01)

  • Higher than in healthy subjects

Yano et al. [33]
78 CKD Stage 1 diabetic nephropathy patientsUsc life Science & Technology Co., Ltd, Wuhan, China8.1 ± 1.3 ng/mLHigher than in healthy subjects (5.3 ± 0.9 ng/mL; P < 0.001)Yilmaz et al. [34]
  • Group A: 26 ADPKD CKD patients (eGFR 45–70 mL/min/1.73 m2)

  • Group B: 26 ADPKD without CKD patients

R&D Systems, Abingdon, UK
  • 4.62 ± 0.59 ng/mL in group A

  • 3.06 ± 0.35 ng/mL in group B (P < 0.001 compared with group A)

Higher in group A and B than in healthy subjects (2.18 ± 0.45 ng/mL; P < 0.001 compared with each group)Raptis et al. [35]
HD
10 HD patientsR&D Systems, Abingdon, UK3376 ± 1723 pg/mLNo difference compared with healthy subjectsCavalier et al. [36]
10 HD patientsR&D Systems, Minneapolis, MN, USA6 ± 3.4 ng/mLHigher than in healthy subjects (3.1 ± 0.6 ng/mL) although not significant (P = 0.07)Esposito et al. [37]
140 HD patientsCloud-Clone Corp., USA40.18 ± 8.36 ng/mLHigher than in healthy subjects (2.5 ± 2.4 ng/mL; P < 0.001)Koyun et al. [38]
  • 37 HD patients

  • 34 PD patients

R&D Systems, Minneapolis, MN, USA5.3 ± 2.6 ng/mLNo control groupLee et al. [39]
60 HD patientsHomemade ELISA25.7 ± 12.8 µg/mLNo difference compared with healthy subjectsHan et al. [40]
204 HD patientsR&D Systems, Minneapolis, MN, USA2573 (1662–3703) pg/mLNo control groupDelanaye et al. [41]
69 PD patientsR&D Systems, Minneapolis, MN, USA7.59 ± 3.37 ng/mL
  • No control group

  • No difference according to residual renal function (r = 0.079, P = 0.510)

Yamada et al. [42]
PopulationAssay techniquesMstn concentration in populationResultsStudy
Non-dialysis CKD
93 CKD patientsR&D Systems, Abingdon, UK3.34 ng/mL
  • Negative correlation with

  • eGFR (β = −0.31, P < 0.01) and ClCr (β = −0.35, P < 0.01)

  • Higher than in healthy subjects

Yano et al. [33]
78 CKD Stage 1 diabetic nephropathy patientsUsc life Science & Technology Co., Ltd, Wuhan, China8.1 ± 1.3 ng/mLHigher than in healthy subjects (5.3 ± 0.9 ng/mL; P < 0.001)Yilmaz et al. [34]
  • Group A: 26 ADPKD CKD patients (eGFR 45–70 mL/min/1.73 m2)

  • Group B: 26 ADPKD without CKD patients

R&D Systems, Abingdon, UK
  • 4.62 ± 0.59 ng/mL in group A

  • 3.06 ± 0.35 ng/mL in group B (P < 0.001 compared with group A)

Higher in group A and B than in healthy subjects (2.18 ± 0.45 ng/mL; P < 0.001 compared with each group)Raptis et al. [35]
HD
10 HD patientsR&D Systems, Abingdon, UK3376 ± 1723 pg/mLNo difference compared with healthy subjectsCavalier et al. [36]
10 HD patientsR&D Systems, Minneapolis, MN, USA6 ± 3.4 ng/mLHigher than in healthy subjects (3.1 ± 0.6 ng/mL) although not significant (P = 0.07)Esposito et al. [37]
140 HD patientsCloud-Clone Corp., USA40.18 ± 8.36 ng/mLHigher than in healthy subjects (2.5 ± 2.4 ng/mL; P < 0.001)Koyun et al. [38]
  • 37 HD patients

  • 34 PD patients

R&D Systems, Minneapolis, MN, USA5.3 ± 2.6 ng/mLNo control groupLee et al. [39]
60 HD patientsHomemade ELISA25.7 ± 12.8 µg/mLNo difference compared with healthy subjectsHan et al. [40]
204 HD patientsR&D Systems, Minneapolis, MN, USA2573 (1662–3703) pg/mLNo control groupDelanaye et al. [41]
69 PD patientsR&D Systems, Minneapolis, MN, USA7.59 ± 3.37 ng/mL
  • No control group

  • No difference according to residual renal function (r = 0.079, P = 0.510)

Yamada et al. [42]

eGFR, estimated glomerular filtration rate; ClCr, creatinine clearance; ADPKD, autosomal dominant polycystic kidney disease.

Table 1

Studies of myostatin plasma concentrations in CKD and dialysis patients

PopulationAssay techniquesMstn concentration in populationResultsStudy
Non-dialysis CKD
93 CKD patientsR&D Systems, Abingdon, UK3.34 ng/mL
  • Negative correlation with

  • eGFR (β = −0.31, P < 0.01) and ClCr (β = −0.35, P < 0.01)

  • Higher than in healthy subjects

Yano et al. [33]
78 CKD Stage 1 diabetic nephropathy patientsUsc life Science & Technology Co., Ltd, Wuhan, China8.1 ± 1.3 ng/mLHigher than in healthy subjects (5.3 ± 0.9 ng/mL; P < 0.001)Yilmaz et al. [34]
  • Group A: 26 ADPKD CKD patients (eGFR 45–70 mL/min/1.73 m2)

  • Group B: 26 ADPKD without CKD patients

R&D Systems, Abingdon, UK
  • 4.62 ± 0.59 ng/mL in group A

  • 3.06 ± 0.35 ng/mL in group B (P < 0.001 compared with group A)

Higher in group A and B than in healthy subjects (2.18 ± 0.45 ng/mL; P < 0.001 compared with each group)Raptis et al. [35]
HD
10 HD patientsR&D Systems, Abingdon, UK3376 ± 1723 pg/mLNo difference compared with healthy subjectsCavalier et al. [36]
10 HD patientsR&D Systems, Minneapolis, MN, USA6 ± 3.4 ng/mLHigher than in healthy subjects (3.1 ± 0.6 ng/mL) although not significant (P = 0.07)Esposito et al. [37]
140 HD patientsCloud-Clone Corp., USA40.18 ± 8.36 ng/mLHigher than in healthy subjects (2.5 ± 2.4 ng/mL; P < 0.001)Koyun et al. [38]
  • 37 HD patients

  • 34 PD patients

R&D Systems, Minneapolis, MN, USA5.3 ± 2.6 ng/mLNo control groupLee et al. [39]
60 HD patientsHomemade ELISA25.7 ± 12.8 µg/mLNo difference compared with healthy subjectsHan et al. [40]
204 HD patientsR&D Systems, Minneapolis, MN, USA2573 (1662–3703) pg/mLNo control groupDelanaye et al. [41]
69 PD patientsR&D Systems, Minneapolis, MN, USA7.59 ± 3.37 ng/mL
  • No control group

  • No difference according to residual renal function (r = 0.079, P = 0.510)

Yamada et al. [42]
PopulationAssay techniquesMstn concentration in populationResultsStudy
Non-dialysis CKD
93 CKD patientsR&D Systems, Abingdon, UK3.34 ng/mL
  • Negative correlation with

  • eGFR (β = −0.31, P < 0.01) and ClCr (β = −0.35, P < 0.01)

  • Higher than in healthy subjects

Yano et al. [33]
78 CKD Stage 1 diabetic nephropathy patientsUsc life Science & Technology Co., Ltd, Wuhan, China8.1 ± 1.3 ng/mLHigher than in healthy subjects (5.3 ± 0.9 ng/mL; P < 0.001)Yilmaz et al. [34]
  • Group A: 26 ADPKD CKD patients (eGFR 45–70 mL/min/1.73 m2)

  • Group B: 26 ADPKD without CKD patients

R&D Systems, Abingdon, UK
  • 4.62 ± 0.59 ng/mL in group A

  • 3.06 ± 0.35 ng/mL in group B (P < 0.001 compared with group A)

Higher in group A and B than in healthy subjects (2.18 ± 0.45 ng/mL; P < 0.001 compared with each group)Raptis et al. [35]
HD
10 HD patientsR&D Systems, Abingdon, UK3376 ± 1723 pg/mLNo difference compared with healthy subjectsCavalier et al. [36]
10 HD patientsR&D Systems, Minneapolis, MN, USA6 ± 3.4 ng/mLHigher than in healthy subjects (3.1 ± 0.6 ng/mL) although not significant (P = 0.07)Esposito et al. [37]
140 HD patientsCloud-Clone Corp., USA40.18 ± 8.36 ng/mLHigher than in healthy subjects (2.5 ± 2.4 ng/mL; P < 0.001)Koyun et al. [38]
  • 37 HD patients

  • 34 PD patients

R&D Systems, Minneapolis, MN, USA5.3 ± 2.6 ng/mLNo control groupLee et al. [39]
60 HD patientsHomemade ELISA25.7 ± 12.8 µg/mLNo difference compared with healthy subjectsHan et al. [40]
204 HD patientsR&D Systems, Minneapolis, MN, USA2573 (1662–3703) pg/mLNo control groupDelanaye et al. [41]
69 PD patientsR&D Systems, Minneapolis, MN, USA7.59 ± 3.37 ng/mL
  • No control group

  • No difference according to residual renal function (r = 0.079, P = 0.510)

Yamada et al. [42]

eGFR, estimated glomerular filtration rate; ClCr, creatinine clearance; ADPKD, autosomal dominant polycystic kidney disease.

Active Mstn is a small 25 kDa dimer and could theoretically be removed during HD or haemodiafiltration (HDF). However, in plasma, active Mstn is mainly linked to its prodomain, forming a 43 kDa protein or 86 kDa protein when dimerized. Thus, dialysis modality could only partly influence Mstn concentration. In a study comparing 60 HD patients, those who were dialysed with high-flux membrane had higher grip strength and lower Mstn concentrations. Mstn concentrations decreased to 36% during the dialysis session in patients with a high-flux membrane but increased to 25% in those with low-flux membranes; however, the dosing of the assay was not described [40]. Delanaye et al. report a 12% decrease in Mstn concentrations during a 4 h high-flux dialyser HD session, but did not find any effect of HDF on conventional HD [41]. Another study in 32 HD patients reports similar removal rates between the two techniques [43]. However, in a crossover study comparing Mstn concentrations after 3 months HD and 3 months HDF, Mstn was significantly lower after the HDF period [37]. Mstn concentrations seem not to be influenced by peritoneal clearance [42].

In HD patients, the correlation between Mstn concentrations and muscle function and mass is contradictory [38–42, 44] (Table 2). Mstn is positively associated with muscle mass or strength in most studies. Since Mstn is produced by muscle cells, its concentration might only be a surrogate marker of muscle mass [41]. Supporting this hypothesis, Zhou et al. report an increase in serum myostatin in CKD patients after 12 months of strength exercise [44]. More recently, an observational study reported an independent correlation between Mstn concentrations and vascular calcifications in PD and HD patients [39].

Table 2

Studies of myostatin plasma concentrations in CKD and dialysis patients and association with muscle parameters

PopulationResultsStudy
140 HD patientsMstn not correlated with ASMI (r = 0.042, P = 0.624)Koyun et al. [38]
20 PD or HD patientsMstn positively correlated with ASMI (r = 0.516, P = 0.020)Lee et al.[39]
60 HD patientsMstn inversely correlated with handgrip strength (r = −0.36, P < 0.01)Han et al. [40]
69 PD patientsMstn positively correlated with LBM (r = 0.57, P < 0.001)Yamada et al. [42]
204 HD patients
  • Mstn positively correlated with handgrip strength (r = 0.37, 95% CI 0.25–0.48, P < 0.0001)

  • Mstn positively correlated with muscle mass index (r = 0.30, 95% CI 0.07–0.50, P = 0.01)

Delanaye et al. [41]
151 CKD patients
  • Mstn is positively correlated with handgrip strength (P < 0.001), isometric quadriceps strength (P < 0.001)

  • Mstn is positively correlated with arm lean mass (P < 0.001), leg lean mass (P < 0.001) and trunk lean mass (P = 0.001)

Zhou et al. [44]
PopulationResultsStudy
140 HD patientsMstn not correlated with ASMI (r = 0.042, P = 0.624)Koyun et al. [38]
20 PD or HD patientsMstn positively correlated with ASMI (r = 0.516, P = 0.020)Lee et al.[39]
60 HD patientsMstn inversely correlated with handgrip strength (r = −0.36, P < 0.01)Han et al. [40]
69 PD patientsMstn positively correlated with LBM (r = 0.57, P < 0.001)Yamada et al. [42]
204 HD patients
  • Mstn positively correlated with handgrip strength (r = 0.37, 95% CI 0.25–0.48, P < 0.0001)

  • Mstn positively correlated with muscle mass index (r = 0.30, 95% CI 0.07–0.50, P = 0.01)

Delanaye et al. [41]
151 CKD patients
  • Mstn is positively correlated with handgrip strength (P < 0.001), isometric quadriceps strength (P < 0.001)

  • Mstn is positively correlated with arm lean mass (P < 0.001), leg lean mass (P < 0.001) and trunk lean mass (P = 0.001)

Zhou et al. [44]

ASMI, appendicular skeletal muscle mass index; CI, confidence interval; LBM, lean body mass.

Table 2

Studies of myostatin plasma concentrations in CKD and dialysis patients and association with muscle parameters

PopulationResultsStudy
140 HD patientsMstn not correlated with ASMI (r = 0.042, P = 0.624)Koyun et al. [38]
20 PD or HD patientsMstn positively correlated with ASMI (r = 0.516, P = 0.020)Lee et al.[39]
60 HD patientsMstn inversely correlated with handgrip strength (r = −0.36, P < 0.01)Han et al. [40]
69 PD patientsMstn positively correlated with LBM (r = 0.57, P < 0.001)Yamada et al. [42]
204 HD patients
  • Mstn positively correlated with handgrip strength (r = 0.37, 95% CI 0.25–0.48, P < 0.0001)

  • Mstn positively correlated with muscle mass index (r = 0.30, 95% CI 0.07–0.50, P = 0.01)

Delanaye et al. [41]
151 CKD patients
  • Mstn is positively correlated with handgrip strength (P < 0.001), isometric quadriceps strength (P < 0.001)

  • Mstn is positively correlated with arm lean mass (P < 0.001), leg lean mass (P < 0.001) and trunk lean mass (P = 0.001)

Zhou et al. [44]
PopulationResultsStudy
140 HD patientsMstn not correlated with ASMI (r = 0.042, P = 0.624)Koyun et al. [38]
20 PD or HD patientsMstn positively correlated with ASMI (r = 0.516, P = 0.020)Lee et al.[39]
60 HD patientsMstn inversely correlated with handgrip strength (r = −0.36, P < 0.01)Han et al. [40]
69 PD patientsMstn positively correlated with LBM (r = 0.57, P < 0.001)Yamada et al. [42]
204 HD patients
  • Mstn positively correlated with handgrip strength (r = 0.37, 95% CI 0.25–0.48, P < 0.0001)

  • Mstn positively correlated with muscle mass index (r = 0.30, 95% CI 0.07–0.50, P = 0.01)

Delanaye et al. [41]
151 CKD patients
  • Mstn is positively correlated with handgrip strength (P < 0.001), isometric quadriceps strength (P < 0.001)

  • Mstn is positively correlated with arm lean mass (P < 0.001), leg lean mass (P < 0.001) and trunk lean mass (P = 0.001)

Zhou et al. [44]

ASMI, appendicular skeletal muscle mass index; CI, confidence interval; LBM, lean body mass.

Overall, several issues may lead to an increase in Mstn in CKD: an increased production under physical inactivity, inflammation, and maybe uraemic toxin triggers and Mstn removal decreases. However, data are lacking on the potential role of kidney on Mstn removal. All these phenomena participate in sarcopenia in CKD [12].

INHIBITION OF THE MYOSTATIN PATHWAY

Mstn pathway inhibition represents a promising therapeutic approach in situations associated with striated muscle dysfunction such as myopathy, cancer or sarcopenia associated with chronic diseases including CKD. Many strategies are proposed to inhibit the Mstn pathway. The majority of them act extracellularly to block Mstn engaging with the ActRIIB receptor complex, either by binding directly to Mstn itself or by binding to components of this receptor complex (Figure 2).

Inhibitors of myostatin used in experimental and clinical trials. ActRII, activin receptor type II; ALK, anaplastic lymphoma receptor tyrosine kinase.
FIGURE 2

Inhibitors of myostatin used in experimental and clinical trials. ActRII, activin receptor type II; ALK, anaplastic lymphoma receptor tyrosine kinase.

Follistatin, the main natural Mstn inhibitor, not only inhibits Mstn’s ligation to its receptor, but is also able to link to other TGF-β family members including Activin A that also regulate negatively muscle growth. As a result, in animal models, follistatin increases satellite cell recruitment, muscle strength and muscle mass [45]. Of note, follistatin plasma concentration is positively correlated with age and inflammatory markers in CKD, but is not associated with renal function [46]. Other natural proteins are involved in Mstn regulation: follistatin-related gene and follistatin-like 3, which inhibit Activin A; growth and differentiation factor-associated serum protein-1 (GASP-1), which shares structural similarities with follistatin; and the soluble ActRIIB, which binds to Activin A or Mstn in the plasma and inhibits their activity on target cells. Therefore, in healthy mice, treatments with these natural inhibitors (follistatin, GASP-1) confirm an increase in muscle mass [47, 48].

Most exogenous Mstn pathway inhibitors are Mstn, Activin A or ActRIIB neutralizing antibodies. In wild-type mice, treatment with monoclonal antibody directed against Mstn [49] or using a mutant inactive form of the Mstn propeptide [50] is capable of increasing muscle mass by almost 25%. As natural molecules, most of these inhibitors reduce protein catabolism, increase Akt phosphorylation and reduce concentrations of inflammatory cytokines including IL-6. They mainly increase protein synthesis in myofibrils, and to a lesser extent promote satellite cell recruitment and myogenesis. Recently, a novel approach with an antibody (SRK-015) targeting the pro-myostatin and blocking the release of the active Mstn has also been developed [11].

In different experiment models of skeletal muscle disease like amyotrophic lateral sclerosis or muscular dystrophy, efficay of first developed anti-Mstn antibodies [49] is contradictory to survival improvements and muscle mass depending on the strategy to inhibit Mstn. Therefore, more specific anti-Mstn antibodies have been developed [11]. Special care has been taken so that these treatments do not activate other TGF-β pathways that could lead to undesirable effects, and previously led a therapeutic trial to premature stop [51]. Anti-Mstn antibody (REGN1033) prevented the loss of muscle mass induced by immobilization, glucocorticoid treatment or with age in mice [52]. Other anti-Mstn antibodies, PF-354 or ATA 842 [53], prevented muscle loss due to ageing and cancer cachexia. In a model of stroke, a better recovery of skeletal muscle mass was observed in mice treated with the anti-Mstn protein PINTA745 [54]. Finally, ActRIIB antagonist or anti-ActRIIB antibody (bimagrumab/BYM338) has positive effects on muscle mass in mouse models of cancer cachexia or glucocorticoid-induced atrophy [55]. Encouraging results are observed with double inhibition of Mstn with Mstn-targeting siRNA and ActRIIB antagonist [56]. Follistatin confirms its therapeutic potential in models of skeletal muscle atrophy but GASP1/2 has not yet been tested in models of muscle wasting.

In CKD, preliminary study by Zhang et al. demonstrated that injections of an anti-Mstn peptibody into uraemic mice reversed loss of body weight and increased muscle mass by 10% [17]. Counteracting Mstn was able to decrease protein degradation rate and inflammation, and prevent fibrosis in skeletal muscles and suppress the proliferation of fibro/adipogenic progenitor cells. An inhibitor of Stat3 (C188-9) prevented activation of Mstn and CKD-induced muscle wasting [16]. Until now, no other pharmacology strategy in CKD has been tested.

Several Phase 1 or Phase 2 studies have been performed or are currently ongoing with different Mstn inhibitors, in the field of geriatrics, muscle dystrophy or oncology. For example, landogrozumab (LY2495655) increases lean mass and might improve functional measures of muscle power in the elderly [57]. This antibody is actually being tested in patients with advanced cancer (NCT01524224). Stamulumab (MYO-029) was tested in Duchenne muscular dystrophy, but the results were not as good as expected and the trial was stopped. In the same disease, antibody domagrozumab (PF-06252616) (NCT02907619) and an anti-Mstn Adnectin fusion protein talditercept alpha (BMS-986089/RG6206) (NCT03039686) have actually been studied. A Phase 2 study (NCT01963598) on safety and efficacy of antibody trevogrumab (REGN1033) has been completed in sarcopenic subjects, although the results are not yet available. Antibodies anti-ActRIIB (bimagrumab/BYM-338) increased muscle mass and strength in older adults with sarcopenia and improved mobility in those with slow walking speed [58]. Also, ACE031 (soluble activin receptor type IIB) showed an increase in lean body mass and attenuation of declines in 6-min walk distance in Duchenne muscular dystrophy [51]. Different activin inhibitors are used in ongoing trials like STM 434 in cancer cachexia (NCT02262455). AMG745/PINTA745, a peptibody against Mstn has been tested in age-associated muscle loss (NCT00975104) and in patients with inclusion myositis (NCT01925209).

In CKD patients, only one Phase 2 randomized, placebo-controlled trial has been performed with AMG745/PINTA745. The encouraging improvement of muscle strength and mass in mice led to a 12-week pilot trial in 51 HD patients with protein-energy wasting syndrome (NCT01958970). Unfortunately, the results of the trial have not been published and development of drugs was stopped because the trial did not meet its primary endpoint, defined as the percent change from baseline in lean body mass as measured by dual energy X-ray absorptiometry at Week 12 following weekly treatment. PINTA 745 also did not improve physical function, measures of glycaemic control and markers of inflammation.

Safety questions were raised about the long-term effect of exposure to Mstn inhibition. ACE-031, a soluble ActRIIB, induced bleeding and telangiectasias and led to a premature stop of a clinical trial [51]. Also, Mstn is expressed in heart, which raises the possibility of cardiomyopathy with sustained Mstn inhibition [59]. On the other hand, the increase in insulin sensitivity combined with an advantageous lipid profile and a possible anti-fibrotic effect on cardiac tissue would come as a welcome side effect [60].

Improvement of factors favouring Mstn overexpression is also an important clue: reducing metabolic acidosis, inflammation, angiotensin II, malnutrition or lack of exercise is a first-line therapy. Physical exercise has been shown to improve muscle mass and function, but to reduce Mstn production in uraemic mice. In HD patients, a 21-week training course combining endurance and resistance exercises led to an increase in IGF-1 mRNA in vastus lateralis muscle biopsies, and a reduction in Mstn mRNA. At the same time, muscle mass was increased and fat mass decreased [61]. Follistatin production is also increased during physical exercise. More studies are needed to better understand the effect of physical exercise on Mstn production and activity during CKD.

CONCLUSION

Muscle mass and function impairment during CKD is a major morbimortality contributor, especially in elderly who associate age sarcopenia with uraemic sarcopenia. Mstn, which is a major stakeholder of muscle protein catabolism in muscle cells and inhibits muscle repair and satellite cell recruitment, is increased in plasma of CKD patients for several reasons, including decreased Mstn clearance. This accumulation participates in uraemic muscle dysfunction, although its pathophysiological comprehension requires further studies. Mstn inhibitors could constitute a promising therapeutic approach in this field, in addition to physical activity and conventional CKD treatments. Much remains to be discovered regarding the biology of the Mstn pathway to inform the development of specific inhibitors and its efficiency in uraemic conditions.

CONFLICT OF INTEREST STATEMENT

The authors have no competing interests to declare. The results presented in this paper have not been published previously in whole or part.

REFERENCES

1

Carrero
JJ
Johansen
KL
Lindholm
B
et al.
Screening for muscle wasting and dysfunction in patients with chronic kidney disease
.
Kidney Int
2016
;
90
:
53
66

2

Cruz-Jentoft
AJ
Baeyens
JP
Bauer
JM
et al. .
Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People
.
Age Ageing
2010
;
39
:
412
423

3

de Souza
VA
Oliveira
D
Barbosa
SR
et al.
Sarcopenia in patients with chronic kidney disease not yet on dialysis: analysis of the prevalence and associated factors
.
PLoS One
2017
;
12
:
e0176230

4

Bataille
S
Serveaux
M
Carreno
E
et al. .
The diagnosis of sarcopenia is mainly driven by muscle mass in hemodialysis patients
.
Clin Nutr
2017
;
36
:
1654
1660

5

Mitch
WE
Goldberg
AL.
Mechanisms of muscle wasting—the role of the ubiquitin–proteasome pathway
.
N Engl J Med
1996
;
335
:
1897
1905

6

Ohsawa
Y
Hagiwara
H
Nakatani
M
et al. .
Muscular atrophy of caveolin-3-deficient mice is rescued by myostatin inhibition
.
J Clin Invest
2006
;
116
:
2924
2934

7

Workeneh
BT
Rondon-Berrios
H
Zhang
L
et al. .
Development of a diagnostic method for detecting increased muscle protein degradation in patients with catabolic conditions
.
J Am Soc Nephrol
2006
;
17
:
3233
3239

8

Wang
XH
Mitch
WE.
Mechanisms of muscle wasting in chronic kidney disease
.
Nat Rev Nephrol
2014
;
10
:
504
516

9

Gao
Y
Arfat
Y
Wang
H
Goswami
N.
Muscle atrophy induced by mechanical unloading: mechanisms and potential countermeasures
.
Front Physiol
2018
;
9
:
235

10

Sanchez
AMJ
Candau
RB
Bernardi
H.
FoxO transcription factors: their roles in the maintenance of skeletal muscle homeostasis
.
Cell Mol Life Sci
2014
;
71
:
1657
1671

11

Pirruccello-Straub
M
Jackson
J
Wawersik
S
et al. .
Blocking extracellular activation of myostatin as a strategy for treating muscle wasting
.
Sci Rep
2018
;
8
:
2292

12

Verzola
D
Barisione
C
Picciotto
D
et al. .
Emerging role of myostatin and its inhibition in the setting of chronic kidney disease
.
Kidney Int
2019
;
95
:
506
517

13

Schiaffino
S
Dyar
KA
Ciciliot
S
et al. .
Mechanisms regulating skeletal muscle growth and atrophy
.
FEBS J
2013
;
280
:
4294
4314

14

Mitch
WE
Medina
R
Grieber
S
et al. .
Metabolic acidosis stimulates muscle protein degradation by activating the adenosine triphosphate-dependent pathway involving ubiquitin and proteasomes
.
J Clin Invest
1994
;
93
:
2127
2133

15

Hu
Z
Wang
H
Lee
IH
et al. .
Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice
.
J Clin Invest
2009
;
119
:
3059
3069

16

Zhang
L
Pan
J
Dong
Y
et al. .
Stat3 activation links a C/EBPδ to myostatin pathway to stimulate loss of muscle mass
.
Cell Metab
2013
;
18
:
368
379

17

Zhang
L
Rajan
V
Lin
E
et al. .
Pharmacological inhibition of myostatin suppresses systemic inflammation and muscle atrophy in mice with chronic kidney disease
.
FASEB J
2011
;
25
:
1653
1663

18

Zhang
L
Wang
XH
Wang
H
et al. .
Satellite cell dysfunction and impaired IGF-1 signaling cause CKD-induced muscle atrophy
.
J Am Soc Nephrol
2010
;
21
:
419
427

19

Mosher
DS
Quignon
P
Bustamante
CD
et al. .
A mutation in the myostatin gene increases muscle mass and enhances racing performance in heterozygote dogs
.
PLoS Genet
2007
;
3
:
e79

20

Schuelke
M
Wagner
KR
Stolz
LE
et al. .
Myostatin mutation associated with gross muscle hypertrophy in a child
.
N Engl J Med
2004
;
350
:
2682
2688

21

Seibert
MJ
Xue
QL
Fried
LP
et al. .
Polymorphic variation in the human myostatin (GDF-8) gene and association with strength measures in the Women’s Health and Aging Study II cohort
.
J Am Geriatr Soc
2001
;
49
:
1093
1096

22

Han
HQ
Mitch
WE.
Targeting the myostatin signaling pathway to treat muscle wasting diseases
.
Curr Opin Support Palliat Care
2011
;
5
:
334
341

23

Hittel
DS
Berggren
JR
Shearer
J
et al. .
Increased secretion and expression of myostatin in skeletal muscle from extremely obese women
.
Diabetes
2009
;
58
:
30
38

24

Cleasby
ME
Jamieson
PM
Atherton
PJ.
Insulin resistance and sarcopenia: mechanistic links between common co-morbidities
.
J Endocrinol
2016
;
229
:
R67
–R
81

25

Price
SR
Gooch
JL
Donaldson
SK
et al.
Muscle atrophy in chronic kidney disease results from abnormalities in insulin signaling
.
J Ren Nutr
2010
;
20
:
S24
–S2
8

26

Sun
DF
Chen
Y
Rabkin
R.
Work-induced changes in skeletal muscle IGF-1 and myostatin gene expression in uremia
.
Kidney Int
2006
;
70
:
453
459

27

Enoki
Y
Watanabe
H
Arake
R
et al. .
Potential therapeutic interventions for chronic kidney disease-associated sarcopenia via indoxyl sulfate-induced mitochondrial dysfunction: Therapy for CKD-associated muscle dysfunction
.
J Cachexia Sarcopenia Muscle
2017
;
8
:
735
747

28

Enoki
Y
Watanabe
H
Arake
R
et al. .
Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress-mediated expression of myostatin and atrogin-1
.
Sci Rep
2016
;
6
:
32084

29

Shah
VO
Dominic
EA
Moseley
P
et al. .
Hemodialysis modulates gene expression profile in skeletal muscle
.
Am J Kidney Dis
2006
;
48
:
616
628

30

Wang
H
Casaburi
R
Taylor
WE
et al. .
Skeletal muscle mRNA for IGF-IEa, IGF-II, and IGF-I receptor is decreased in sedentary chronic hemodialysis patients
.
Kidney Int
2005
;
68
:
352
361

31

Verzola
D
Procopio
V
Sofia
A
et al. .
Apoptosis and myostatin mRNA are upregulated in the skeletal muscle of patients with chronic kidney disease
.
Kidney Int
2011
;
79
:
773
782

32

Kopple
JD
Cohen
AH
Wang
H
et al. .
Effect of exercise on mRNA levels for growth factors in skeletal muscle of hemodialysis patients
.
J Ren Nutr
2006
;
16
:
312
324

33

Yano
S
Nagai
A
Isomura
M
et al. .
Relationship between blood myostatin levels and kidney function: Shimane CoHRE study
.
PLoS One
2015
;
10
:
e0141035

34

Yilmaz
MI
Sonmez
A
Saglam
M
et al. .
Reduced proteinuria using ramipril in diabetic CKD stage 1 decreases circulating cell death receptor activators concurrently with ADMA
.
A novel pathophysiological pathway? Nephrol Dial Transplant
2010
;
25
:
3250
3256

35

Raptis
V
Bakogiannis
C
Loutradis
C
et al. .
Serum Fas ligand, serum myostatin and urine TGF-β1 are elevated in autosomal dominant polycystic kidney disease patients with impaired and preserved renal function
.
Kidney Blood Press Res
2018
;
43
:
744
754

36

Cavalier
E
Beaudart
C
Buckinx
F
et al. .
Critical analytical evaluation of promising markers for sarcopenia
.
Eur Geriatr Med
2016
;
7
:
239
242

37

Esposito
P
La Porta
E
Calatroni
M
et al. .
Modulation of myostatin/hepatocyte growth factor balance by different hemodialysis modalities
.
BioMed Res Int
2017
;
2017
:
1
5

38

Koyun
D
Nergizoglu
G
Kir
KM.
Evaluation of the relationship between muscle mass and serum myostatin levels in chronic hemodialysis patients
.
Saudi J Kidney Dis Transpl
2018
;
29
:
809
815

39

Lee
SM
Kim
SE
Lee
JY
et al. .
Serum myostatin levels are associated with abdominal aortic calcification in dialysis patients
.
Kidney Res Clin Pract
2019
;
38
:
481
489

40

Han
D-S
Chen
Y-M
Lin
S-Y
et al. .
Serum myostatin levels and grip strength in normal subjects and patients on maintenance haemodialysis
.
Clin Endocrinol (Oxf)
2011
;
75
:
857
863

41

Delanaye
P
Bataille
S
Quinonez
K
et al. .
Myostatin and insulin-like growth factor 1 are biomarkers of muscle strength, muscle mass, and mortality in patients on hemodialysis
.
J Ren Nutr
2019
;
29
:
511
520

42

Yamada
S
Tsuruya
K
Yoshida
H
et al. .
Factors associated with the serum myostatin level in patients undergoing peritoneal dialysis: potential effects of skeletal muscle mass and vitamin D receptor activator use
.
Calcif Tissue Int
2016
;
99
:
13
22

43

Morena
M
Creput
C
Bouzernidj
M
et al. .
Randomised trial on clinical performances and biocompatibility of four high-flux hemodialyzers in two mode treatments: hemodialysis vs post dilution hemodiafiltration
.
Sci Rep
2019
;
9
:
18265

44

Zhou
Y
Hellberg
M
Hellmark
T
et al. .
Muscle mass and plasma myostatin after exercise training: a substudy of Renal Exercise (RENEXC)—a randomized controlled trial
.
Nephrol Dial Transplant
2019
; doi: 10.1093/ndt/gfz210

45

Hansen
J
Brandt
C
Nielsen
AR
et al. .
Exercise induces a marked increase in plasma follistatin: evidence that follistatin is a contraction-induced hepatokine
.
Endocrinology
2011
;
152
:
164
171

46

Miyamoto
T
Carrero
JJ
Rashid Qureshi
A
et al. .
Circulating follistatin in patients with chronic kidney disease: implications for muscle strength, bone mineral density, inflammation, and survival
.
Clin J Am Soc Nephrol
2011
;
6
:
1001
1008

47

Lee
S-J
Reed
LA
Davies
MV
et al. .
Regulation of muscle growth by multiple ligands signaling through activin type II receptors
.
Proc Natl Acad Sci USA
2005
;
102
:
18117
18122

48

Kralisch
S
Hoffmann
A
Klöting
N
et al. .
FSTL3 is increased in renal dysfunction
.
Nephrol Dial Transplant
2017
;
32
:
1637
1644

49

Bogdanovich
S
Krag
TOB
Barton
ER
et al. .
Functional improvement of dystrophic muscle by myostatin blockade
.
Nature
2002
;
420
:
418
421

50

Collins-Hooper
H
Sartori
R
Macharia
R
et al. .
Propeptide-mediated inhibition of myostatin increases muscle mass through inhibiting proteolytic pathways in aged mice
.
J Gerontol A Biol Sci Med Sci
2014
;
69
:
1049
1059

51

Campbell
C
McMillan
HJ
Mah
JK
et al. .
Myostatin inhibitor ACE-031 treatment of ambulatory boys with Duchenne muscular dystrophy: results of a randomized, placebo-controlled clinical trial
.
Muscle Nerve
2017
;
55
:
458
464

52

Latres
E
Pangilinan
J
Miloscio
L
et al. .
Myostatin blockade with a fully human monoclonal antibody induces muscle hypertrophy and reverses muscle atrophy in young and aged mice
.
Skelet Muscle
2015
;
5
:
34

53

Camporez
J-P
Petersen
MC
Abudukadier
A
et al. .
Anti-myostatin antibody increases muscle mass and strength and improves insulin sensitivity in old mice
.
Proc Natl Acad Sci USA
2016
;
113
:
2212
2217

54

Desgeorges
MM
Devillard
X
Toutain
J
et al. .
Pharmacological inhibition of myostatin improves skeletal muscle mass and function in a mouse model of stroke
.
Sci Rep
2017
;
7
:
14000

55

Lach-Trifilieff
E
Minetti
GC
Sheppard
K
et al. .
An antibody blocking activin type II receptors induces strong skeletal muscle hypertrophy and protects from atrophy
.
Mol Cell Biol
2014
;
34
:
606
618

56

Bayarsaikhan
O
Kawai
N
Mori
H
et al. .
Co-administration of myostatin-targeting siRNA and ActRIIB-Fc fusion protein increases masseter muscle mass and fiber size
.
J Nutr Sci Vitaminol
2017
;
63
:
244
248

57

Becker
C
Lord
SR
Studenski
SA
et al. .
Myostatin antibody (LY2495655) in older weak fallers: a proof-of-concept, randomised, phase 2 trial
.
Lancet Diabetes Endocrinol
2015
;
3
:
948
957

58

Rooks
D
Praestgaard
J
Hariry
S
et al. .
Treatment of sarcopenia with bimagrumab: results from a phase II, randomized, controlled, proof-of-concept study
.
J Am Geriatr Soc
2017
;
65
:
1988
1995

59

Oshima
Y
Ouchi
N
Shimano
M
et al. .
Activin A and follistatin-like 3 determine the susceptibility of heart to ischemic injury
.
Circulation
2009
;
120
:
1606
1615

60

Morissette
MR
Stricker
JC
Rosenberg
MA
et al. .
Effects of myostatin deletion in aging mice
.
Aging Cell
2009
;
8
:
573
583

61

Kopple
JD
Wang
H
Casaburi
R
et al. .
Exercise in maintenance hemodialysis patients induces transcriptional changes in genes favoring anabolic muscle
.
J Am Soc Nephrol
2007
;
18
:
2975
2986

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.