Critical role of miRNAs in mediating skeletal muscle atrophy (Review)

  • Authors:
    • Yonghui Yu
    • Wanli Chu
    • Jiake Chai
    • Xiao Li
    • Lingying Liu
    • Li Ma
  • View Affiliations

  • Published online on: December 30, 2015     https://doi.org/10.3892/mmr.2015.4748
  • Pages: 1470-1474
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Skeletal muscle atrophy, a conventional clinical feature in patients with cancer, chronic obstructive pulmonary disease, sepsis and severe burns, is defined as a reduction in muscle mass. During atrophy, the protein degradation is abnormally activated and the aberrance between protein synthesis and protein degradation results in muscle atrophy. Previous studies have demonstrated that miRNAs, small non‑coding RNA molecules, serve an important role in the regulation of muscle atrophy. Further studies have indicated the implications of the ubiquitin‑proteasome and PI3K/Akt/FoxO signaling pathways and myogenic regulatory factors in miRNA‑mediated muscle atrophy. Therefore, in this review, the effects and molecular mechanisms of miRNAs on muscle atrophy are summarized, leading to the suggestion that miRNAs may serve as potential therapeutic targets in muscle atrophy.

1. Introduction

Skeletal muscle makes up approximately 40% of the body weight and is essential for locomotion (1). Skeletal muscle atrophy, predominantly resulting from excessive protein degradation, occurs in various conditions including starvation (2), aging (3), sepsis (4), cancer cachexia (5), severe burns (6,7) and chronic kidney disease (8). Muscle atrophy results in reductions in mobility of the patients and an increased risk of mortality (9). In patients with severe burns, skeletal muscle atrophy occurs as a result of prolongation of time spent bed-bound and suppression of wound healing (7). In general, skeletal muscle atrophy predicts poor prognosis of patients.

The ubiquitin-proteasome pathway and cell apoptosis are involved in regulating skeletal muscle atrophy (1012). The ubiquitin-proteasome pathway contributes to protein degradation, as the targeted proteins for degradation are substrates that can be identified and bound to ubiquitin. Subsequently, poly-ubiquitinated substrates are targeted for degradation by proteasomes (11). Activation of the ubiquitin-proteasome pathway may serve an important role in the mediation of skeletal muscle atrophy (10,11). Previous studies have additionally demonstrated that increased cell apoptosis is accompanied by stress-induced skeletal muscle atrophy (12), and that apoptosis is also a critical factor which leads to muscle atrophy (13,14).

MicroRNAs (miRNAs), the small non-coding RNAs, were first identified in C. elegans and are highly conserved in eukaryotes (15). At present, greater than 1,700 miRNAs have been identified, which serve critical roles in regulating proliferation, differentiation and the development of various diseases (16). miRNA exerts its biological activation via binding to the 3′-untranslated region (3′-UTR) of targeted mRNA (17). Accelerating target mRNA degradation or inhibiting its translation are two key ways in which miRNA mediates the control of gene expression (18). The miRNA (miR)-1/206 family, miR-133, miR-208 and miR-488 are identified as muscle-specific miRNAs and serve essential roles in regulating normal myoblast differentiation, proliferation and muscle remodeling in response to stress (1921). In addition, muscle-specific miRNAs, miR-128a and miR-351, are involved in the regulation of myogenesis (21,22).

The present review will focus upon the miRNAs involved in the regulation of skeletal muscle atrophy and the potential molecular mechanisms. Further studies are required in order to elucidate the specific miRNAs implicated in stress-induced skeletal muscle atrophy, which may lead to the development of novel targets for clinical therapy.

2. The effect of the ubiquitin-proteasome pathway on miR-NA-mediated muscle atrophy

Aberrant activation of protein degradation is the key factor that leads to muscle atrophy, and the ubiquitin-proteasome pathway serves a pivotal role in the mediation of protein degradation (11). The proteasome can identify the poly-ubiq-uitinated protein and trigger the degradation procedure (23). E3 ligase is the critical mediator of protein ubiquitination. Muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx) are two muscle specific E3 ligases (24). During muscle atrophy, MuRF1 and MAFbx are overexpressed in muscle, and inhibiting the function of MuRF1 and MAFbx has been demonstrated to suppress muscle loss and subsequently attenuate muscle atrophy (25,26). Previous studies (2729) have additionally indicated that miRNAs are implicated in the regulation of MuRF1 and MAFbx expression (Fig. 1). miR-23a is able to inhibit the translational activation of MuRF1 and MAFbx via binding with their 3′-UTR, and miR-23a transgenic mice exert resistance against glucocorticoid-induced muscle atrophy (27). In a dexamethasone (Dex)-induced mouse model of atrophy, muscle-specific miR-1 expression is upregulated. miR-1 has been previously reported to induce MuRF1 and MAFbx expression via the HSP70/protein kinase B(Akt)/forkhead box (Fox) O3 signaling pathway and is responsible for Dex-induced muscle atrophy (28). The miR-199/214 cluster is also involved in regulating the ubiquitin-proteasome pathway (29). Taken together, miRNA-dependent activation of the ubiquitin-proteasome pathway is responsible for the promotion of muscle atrophy by directly or indirectly targeting the muscle specific E3 ligases of MuRF1 and MAFbx.

3. miRNAs mediate muscle atrophy via the regulation of myogenesis

In addition to enhancing muscle proteolysis, aberrant myogenesis is also a critical factor during muscle atrophy. Myogenesis is impaired in models of mice with cancer (30), and pigs with chronic obstructive pulmonary disease (31). Inactivation of myogenesis is also observed in diseases such as Duchenne muscular dystrophy and spinal and bulbar muscular atrophy (32,33). Muscle satellite cells are stem cells with self-renewal and differentiation potency, and when muscle disruption occurs, proliferative satellite cells could differentiate into myotubes and contribute to muscle regeneration (34). Defects in post-natal myogenesis and muscle regeneration result in muscle atrophy, and miRNAs are implicated in the regulation of myogenesis and muscle atrophy (35,36). As presented in Fig. 2, paired-box transcription factor (Pax) is essential for satellite cell proliferation and differentiation. miR-1, miR-206 and miR-486 restrict satellite cell proliferation and promote its differentiation through suppression of Pax7 expression (3739). Pax3 is also the critical factor required to trigger satellite cell proliferation; suppressing miR-27b, miR-1 and miR-206 expression suppresses satellite cell differentiation via enhancement of Pax3 activation (40,41). The myogenic regulatory factor (MRF) family, which includes MyoD, Myf5, myogenin and Myf6, has the pivotal role in myogenic differentiation. MyoD is expressed in activated satellite cells, and miR-27a overexpression elevates the MyoD protein level and enhances myoblast differentiation (42). In C2C12 myoblast cells, miR-26a upregulates MyoD expression and promotes the myogenic process (43). Subsequent to injury, miR-26a is induced during muscle regeneration, and blocking miR-26a expression enhances Smad-dependent muscle differentiation (44). miR-186 suppresses C2C12 myoblast cell myogenic differentiation via targeting myogenin (45). In addition to the MRF family, miRNAs also regulate myogenesis through targeting a variety of proteins. Myostatin is the negative mediator of myogenesis; miR-27a and miR-27b promote satellite cell proliferation and post-natal myogenesis by suppressing myostatin expression (4648). miR-125b, miR-133 and miR-199a-3p are involved in the regulation of the insulin-like growth factor/insulin-like growth factor receptor signaling pathway and inhibit cell differentiation and muscle regeneration (4951). miR-203 functions as the suppressor of myoblast differentiation by repressing c-Jun and myocyte enhancer factor 2C (MEF2C) expression (52). miR-155 inhibits MEF2A expression and suppresses the myogenic process (53). miR-29 is a pro-myogenic factor, which acts through downregulation of Akt3 or RING1 and YY1-binding protein (54,55). Thus, miRNAs have critical roles in regulating satellite cell proliferation, myogenic differentiation and muscle regeneration.

4. Implications of miRNAs in cell apoptosis-mediated muscle atrophy

Cell apoptosis is programmed cell death and is a promoting factor of muscle atrophy (14,56). Studies using a mouse model have demonstrated that cell apoptosis is involved in the progression of heart failure, severe burns and age-associated muscle atrophy (5760). The mitochondria and caspase-mediated apoptotic pathways are some of the mechanisms of burn, age or stress-induced muscle atrophy (12,57,61,62). miRNA is an important mediator of myoblast cell apoptosis (63). In skeletal muscle, pre-conditional activation of interleukin (IL)-11/signal transducer and activator of transcription (STAT)3 pathway protects human skeletal myoblasts from oxidant-induced apoptosis (64), and miR-21 is a key regulator of extracellular signal-related kinase 1/2-STAT3 signaling downstream of IL-11 and inhibits the apoptosis of skeletal myoblasts (65). Skeletal muscle loss in cancer cachexia is partially associated with cell apoptosis, and a previous study indicated that miR-21 in microvesicles of cancer cachexia triggers muscle cell apoptosis via enhancement of c-Jun N-terminal kinase activation (66). In acute muscle injury, myogenic progenitor cell apoptosis is triggered by miR-351 knockdown (21). MyoD is a critical factor in the regulation muscle differentiation; MyoD knockout in myoblasts decelerates miR-1 and miR-206 expression and results in resistance to apoptosis (67). Forced MyoD expression in MyoD knockout myoblasts enhances the expression of miR-1 and miR-206 and triggers cell apoptosis via Pax3 downregulation (67). Thus, it is suggested that miRNA is a critical mediator in regulating myoblast apoptosis and implicated in muscle atrophic process.

5. PI3K/Akt/FoxO signaling pathway in miRNA-mediated muscle atrophy

The PI3K/Akt/FoxO signal pathway serves an important role in muscle atrophy (Fig. 1). Attenuated activation of the PI3K/Akt signaling pathway results in rat skeletal muscle atrophy (13,68). Akt inactivation functions as the promoter of burn-induced muscle atrophy (69). FoxO is phosphorylated and exported into the cytoplasm by the upstream kinase Akt, and cytoplasmic FoxO is degraded with loss of transactivation (70). FoxO is additionally implicated in promoting muscle atrophy (71). Tumor necrosis factor receptor-associated factor 6 promotes starvation-induced atrophy in an Akt/FoxO3a-dependent manner (2). FoxO1, the dominant mediator of muscle atrophy, serves a critical role in chronic kidney disease or burn-induced muscle atrophy (8,72). miR-486, the regulator of PTEN, is overexpressed in Duchenne muscular dystrophy; and miR-486 transgenic mice exert the impairment of muscle regeneration in a PTEN/Akt dependent manner (73). In patients with breast patients, miR-486 is downregulated, and the expression of its target genes PTEN and FoxO1A are elevated (74). Myostatin is well known as a negative regulator of muscle mass by reducing protein synthesis. Overexpression of miR-486 is observed in skeletal muscle of myostatin knockout mice and is essential to maintain skeletal muscle size through the Akt/mTOR signaling pathway (75). In the C2C12 myotube cells, miR-182 was reported to suppress FoxO3a protein expression via binding to the 3′-UTR of FoxO3a mRNA, and prevent glucocorticoid-induced rat muscle atrophy (76). Muscle-specific miR-1 is involved in dephos-phorylating and activating FoxO3a in an HSP70/Akt dependent manner and promotes Dex- or myostatin-induced atrophy in skeletal muscle (28). In summary, PI3K/Akt inactivation reduces FoxO protein phosphorylation and dephosphorylated FoxO enters into the nucleus and promotes muscle atrophy.

6. Conclusion

Aberrant muscle protein degradation, impairment of myogenesis, and promotion of muscle cell apoptosis are all important factors that contribute to muscle atrophy. miRNAs are critical mediators of protein degradation and myogenesis through regulation of the ubiquitin-proteasome and PI3K/Akt/FoxO signaling pathways and other myogenic regulatory factors. Thus, miRNAs may be potential and effective therapeutic targets for muscle atrophy.

Acknowledgments

The authors would like to thank Dr Hongli Wu for her critical reading of this review. The current study was supported by the National Science Foundation of China (grant nos. NSFC81120108014, NSFC81471873, NSFC81501694, NSFC81571894 and NSFC81171807), the Beijing Natural Science Foundation (grant no. 7144250) and the Nursery Fund of People's Liberation Army General Hospital (grant nos. 14KMM22 and 13KMM31).

References

1 

Hitachi K and Tsuchida K: Role of microRNAs in skeletal muscle hypertrophy. Front Physiol. 4:4082014. View Article : Google Scholar : PubMed/NCBI

2 

Paul PK, Bhatnagar S, Mishra V, Srivastava S, Darnay BG, Choi Y and Kumar A: The E3 Kubiquitin ligase TRAF6 intercedes in starvation-induced skeletal muscle atrophy through multiple mechanisms. Mol Cell Biol. 32:1248–1259. 2012. View Article : Google Scholar : PubMed/NCBI

3 

McGregor RA, Poppitt SD and Cameron-Smith D: Role of microRNAs in the age-related changes in skeletal muscle and diet or exercise interventions to promote healthy aging in humans. Ageing Res Rev. 17:25–33. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Nystrom G, Pruznak A, Huber D, Frost RA and Lang CH: Local insulin-like growth factor I prevents sepsis-induced muscle atrophy. Metabolism. 58:787–797. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Wang H, Lai YJ, Chan YL, Li TL and Wu CJ: Epigallocatechin-3-gallate effectively attenuates skeletal muscle atrophy caused by cancer cachexia. Cancer Lett. 305:40–49. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Bertsch S, Lang CH and Vary TC: Inhibition of glycogen synthase kinase 3[beta] activity with lithium in vitro attenuates sepsis-induced changes in muscle protein turnover. Shock. 35:266–274. 2011. View Article : Google Scholar

7 

Hart DW, Wolf SE, Chinkes DL, Gore DC, Mlcak RP, Beauford RB, Obeng MK, Lal S, Gold WF, Wolfe RR and Herndon DN: Determinants of skeletal muscle catabolism after severe burn. Ann Surg. 232:455–465. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Xu J, Li R, Workeneh B, Dong Y, Wang X and Hu Z: Transcription factor FoxO1, the dominant mediator of muscle wasting in chronic kidney disease, is inhibited by microRNA-486. Kidney Int. 82:401–411. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Metter EJ, Talbot LA, Schrager M and Conwit R: Skeletal muscle strength as a predictor of all-cause mortality in healthy men. J Gerontol A Biol Sci Med Sci. 57:B359–B365. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Chai J, Wu Y and Sheng ZZ: Role of ubiquitin-proteasome pathway in skeletal muscle wasting in rats with endotoxemia. Crit Care Med. 31:1802–1807. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Attaix D, Combaret L, Bechet D and Taillandier D: Role of the ubiquitin-proteasome pathway in muscle atrophy in cachexia. Curr Opin Support Palliat Care. 2:262–266. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Sishi B, Loos B, Ellis B, Smith W, du Toit EF and Engelbrecht AM: Diet-induced obesity alters signalling pathways and induces atrophy and apoptosis in skeletal muscle in a prediabetic rat model. Exp Physiol. 96:179–193. 2011. View Article : Google Scholar

13 

Engelbrecht AM, Smith C, Neethling I, Thomas M, Ellis B, Mattheyse M and Myburgh KH: Daily brief restraint stress alters signaling pathways and induces atrophy and apoptosis in rat skeletal muscle. Stress. 13:132–141. 2010. View Article : Google Scholar

14 

Dupont-Versteegden EE: Apoptosis in skeletal muscle and its relevance to atrophy. World J Gastroenterol. 12:7463–7466. 2006.PubMed/NCBI

15 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar : PubMed/NCBI

16 

Sayed D and Abdellatif M: MicroRNAs in development and disease. Physiol Rev. 91:827–887. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Didiano D and Hobert O: Molecular architecture of a miRNA-regulated 3′ UTR. RNA. 14:1297–1317. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Travaglini L, Vian L, Billi M, Grignani F and Nervi C: Epigenetic reprogramming of breast cancer cells by valproic acid occurs regardless of estrogen receptor status. Int J Biochem Cell Biol. 41:225–234. 2009. View Article : Google Scholar

20 

Taulli R, Bersani F, Foglizzo V, Linari A, Vigna E, Ladanyi M, Tuschl T and Ponzetto C: The muscle-specific microRNA miR-206 blocks human rhabdomyosarcoma growth in xeno-transplanted mice by promoting myogenic differentiation. J Clin Invest. 119:2366–2378. 2009.PubMed/NCBI

21 

Chen Y, Melton DW, Gelfond JA, McManus LM and Shireman PK: MiR-351 transiently increases during muscle regeneration and promotes progenitor cell proliferation and survival upon differentiation. Physiol Genomics. 44:1042–1051. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Motohashi N, Alexander MS, Shimizu-Motohashi Y, Myers JA, Kawahara G and Kunkel LM: Regulation of IRS1/Akt insulin signaling by microRNA-128a during myogenesis. J Cell Sci. 126:2678–2691. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Hartmann-Petersen R and Gordon C: Proteins interacting with the 26S proteasome. Cell Mol Life Sci. 61:1589–1595. 2004.PubMed/NCBI

24 

Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, Poueymirou WT, Panaro FJ, Na E, Dharmarajan K, et al: Identification of ubiquitin ligases required for skeletal muscle atrophy. Science. 294:1704–1708. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Eddins MJ, Marblestone JG, Suresh Kumar KG, Leach CA, Sterner DE, Mattern MR and Nicholson B: Targeting the ubiquitin E3 ligase MuRF1 to inhibit muscle atrophy. Cell Biochem Biophys. 60:113–118. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Clavel S, Coldefy AS, Kurkdjian E, Salles J, Margaritis I and Derijard B: Atrophy-related ubiquitin ligases, atrogin-1 and MuRF1 are up-regulated in aged rat Tibialis Anterior muscle. Mech Ageing Dev. 127:794–801. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Wada S, Kato Y, Okutsu M, Miyaki S, Suzuki K, Yan Z, Schiaffino S, Asahara H, Ushida T and Akimoto T: Translational suppression of atrophic regulators by microRNA-23a integrates resistance to skeletal muscle atrophy. J Biol Chem. 286:38456–38465. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Kukreti H, Amuthavalli K, Harikumar A, Sathiyamoorthy S, Feng PZ, Anantharaj R, Tan SL, Lokireddy S, Bonala S, Sriram S, et al: Muscle-specific microRNA1 (miR1) targets heat shock protein 70 (HSP70) during dexamethasone-mediated atrophy. J Biol Chem. 288:6663–6678. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Baumgarten A, Bang C, Tschirner A, Engelmann A, Adams V, von Haehling S, Doehner W, Pregla R, Anker MS, Blecharz K, et al: TWIST1 regulates the activity of ubiquitin proteasome system via the miR-199/214 cluster in human end-stage dilated cardiomyopathy. Int J Cardiol. 168:1447–1452. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Penna F, Costamagna D, Fanzani A, Bonelli G, Baccino FM and Costelli P: Muscle wasting and impaired myogenesis in tumor bearing mice are prevented by ERK inhibition. PLoS One. 5:e136042010. View Article : Google Scholar : PubMed/NCBI

31 

Verhees KJ, Pansters NA, Baarsma HA, Remels AH, Haegens A, de Theije CC, Schols AM, Gosens R and Langen RC: Pharmacological inhibition of GSK-3 in a guinea pig model of LPS-induced pulmonary inflammation: II. Effects on skeletal muscle atrophy. Respir Res. 14:1172013. View Article : Google Scholar : PubMed/NCBI

32 

Shi H, Verma M, Zhang L, Dong C, Flavell RA and Bennett AM: Improved regenerative myogenesis and muscular dystrophy in mice lacking Mkp5. J Clin Invest. 123:2064–2077. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Malena A, Pennuto M, Tezze C, Querin G, D'Ascenzo C, Silani V, Cenacchi G, Scaramozza A, Romito S, Morandi L, et al: Androgen-dependent impairment of myogenesis in spinal and bulbar muscular atrophy. Acta Neuropathol. 126:109–121. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Sacco A, Doyonnas R, Kraft P, Vitorovic S and Blau HM: Self-renewal and expansion of single transplanted muscle stem cells. Nature. 456:502–506. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Dachs E, Hereu M, Piedrafita L, Casanovas A, Calderó J and Esquerda JE: Defective neuromuscular junction organization and postnatal myogenesis in mice with severe spinal muscular atrophy. J Neuropathol Exp Neurol. 70:444–461. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Wang XH: MicroRNA in myogenesis and muscle atrophy. Curr Opin Clin Nutr Metab Care. 16:258–266. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Chen JF, Tao Y, Li J, Deng Z, Yan Z, Xiao X and Wang DZ: microRNA-1 and microRNA-206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. J Cell Biol. 190:867–879. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Dey BK, Gagan J and Dutta A: miR-206 and -486 induce myoblast differentiation by downregulating Pax7. Mol Cell Biol. 31:203–214. 2011. View Article : Google Scholar :

39 

Liu N, Williams AH, Maxeiner JM, Bezprozvannaya S, Shelton JM, Richardson JA, Bassel-Duby R and Olson EN: microRNA-206 promotes skeletal muscle regeneration and delays progression of Duchenne muscular dystrophy in mice. J Clin Invest. 122:2054–2065. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Goljanek-Whysall K, Sweetman D, Abu-Elmagd M, Chapnik E, Dalmay T, Hornstein E and Münsterberg A: MicroRNA regulation of the paired-box transcription factor Pax3 confers robustness to developmental timing of myogenesis. Proc Natl Acad Sci USA. 108:11936–11941. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Crist CG, Montarras D, Pallafacchina G, Cumano A, Conway SJ and Buckingham M: Muscle stem cell behavior is modified by microRNA-27 regulation of Pax3 expression. Proc Natl Acad Sci USA. 106:13383–13387. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Chen X, Huang Z, Chen D, Yang T and Liu G: Role of microRNA-27a in myoblast differentiation. Cell Biol Int. 38:266–271. 2014. View Article : Google Scholar

43 

Wong CF and Tellam RL: MicroRNA-26a targets the histone methyltransferase Enhancer of Zeste homolog 2 during myogenesis. J Biol Chem. 283:9836–9843. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Dey BK, Gagan J, Yan Z and Dutta A: miR-26a is required for skeletal muscle differentiation and regeneration in mice. Genes Dev. 26:2180–2191. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Antoniou A, Mastroyiannopoulos NP, Uney JB and Phylactou LA: miR-186 inhibits muscle cell differentiation through myogenin regulation. J Biol Chem. 289:3923–3935. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Huang Z, Chen X, Yu B, He J and Chen D: MicroRNA-27a promotes myoblast proliferation by targeting myostatin. Biochem Biophys Res Commun. 423:265–269. 2012. View Article : Google Scholar : PubMed/NCBI

47 

McFarlane C, Vajjala A, Arigela H, Lokireddy S, Ge X, Bonala S, Manickam R, Kambadur R and Sharma M: Negative auto-regulation of myostatin expression is mediated by Smad3 and microRNA-27. PLoS One. 9:e876872014. View Article : Google Scholar : PubMed/NCBI

48 

Yang T, Chen XL, Huang ZQ, Wen WX, Xu M, Chen DW, Yu B, He J, Luo JQ, Yu J, et al: MicroRNA-27a promotes porcine myoblast proliferation by downregulating myostatin expression. Animal. 8:1867–1872. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Ge Y, Sun Y and Chen J: IGF-II is regulated by microRNA-125b in skeletal myogenesis. J Cell Biol. 192:69–81. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Huang MB, Xu H, Xie SJ, Zhou H and Qu LH: Insulin-like growth factor-1 receptor is regulated by microRNA-133 during skeletal myogenesis. PLoS One. 6:e291732011. View Article : Google Scholar : PubMed/NCBI

51 

Jia L, Li YF, Wu GF, Song ZY, Lu HZ, Song CC, Zhang QL, Zhu JY, Yang GS and Shi XE: MiRNA-199a-3p regulates C2C12 myoblast differentiation through IGF-1/AKT/mTOR signal pathway. Int J Mol Sci. 15:296–308. 2013. View Article : Google Scholar

52 

Luo W, Wu H, Ye Y, Li Z, Hao S, Kong L, Zheng X, Lin S, Nie Q and Zhang X: The transient expression of miR-203 and its inhibiting effects on skeletal muscle cell proliferation and differentiation. Cell Death Dis. 5:e13472014. View Article : Google Scholar : PubMed/NCBI

53 

Seok HY, Tatsuguchi M, Callis TE, He A, Pu WT and Wang DZ: miR-155 inhibits expression of the MEF2A protein to repress skeletal muscle differentiation. J Biol Chem. 286:35339–35346. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Wei W, He HB, Zhang WY, Zhang HX, Bai JB, Liu HZ, Cao JH, Chang KC, Li XY and Zhao SH: miR-29 targets Akt3 to reduce proliferation and facilitate differentiation of myoblasts in skeletal muscle development. Cell Death Dis. 4:e6682013. View Article : Google Scholar : PubMed/NCBI

55 

Zhou L, Wang L, Lu L, Jiang P, Sun H and Wang H: A novel target of microRNA-29, Ring1 and YY1-binding protein (Rybp), negatively regulates skeletal myogenesis. J Biol Chem. 287:25255–25265. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Dupont-Versteegden EE: Apoptosis in muscle atrophy: Relevance to sarcopenia. Exp Gerontol. 40:473–481. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Dirks AJ and Leeuwenburgh C: The role of apoptosis in age-related skeletal muscle atrophy. Sports Med. 35:473–483. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Lee HY, Kaneki M, Andreas J, Tompkins RG and Martyn JA: Novel mitochondria-targeted antioxidant peptide ameliorates burn-induced apoptosis and endoplasmic reticulum stress in the skeletal muscle of mice. Shock. 36:580–585. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Fanzani A, Conraads VM, Penna F and Martinet W: Molecular and cellular mechanisms of skeletal muscle atrophy: An update. J Cachexia Sarcopenia Muscle. 3:163–179. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Libera LD, Zennaro R, Sandri M, Ambrosio GB and Vescovo G: Apoptosis and atrophy in rat slow skeletal muscles in chronic heart failure. Am J Physiol. 277:C982–C986. 1999.PubMed/NCBI

61 

Yasuhara S, Perez ME, Kanakubo E, Yasuhara Y, Shin YS, Kaneki M, Fujita T and Martyn JA: Skeletal muscle apoptosis after burns is associated with activation of proapoptotic signals. Am J Physiol Endocrinol Metab. 279:E1114–E1121. 2000.PubMed/NCBI

62 

Marzetti E, Lawler JM, Hiona A, Manini T, Seo AY and Leeuwenburgh C: Modulation of age-induced apoptotic signaling and cellular remodeling by exercise and calorie restriction in skeletal muscle. Free Radic Biol Med. 44:160–168. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Callis TE, Chen JF and Wang DZ: MicroRNAs in skeletal and cardiac muscle development. DNA Cell Biol. 26:219–225. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Idris NM, Ashraf M, Ahmed RP, Shujia J and Haider KH: Activation of IL-11/STAT3 pathway in preconditioned human skeletal myoblasts blocks apoptotic cascade under oxidant stress. Regen Med. 7:47–57. 2012. View Article : Google Scholar

65 

Haider KH, Idris NM, Kim HW, Ahmed RP, Shujia J and Ashraf M: MicroRNA-21 is a key determinant in IL-11/Stat3 anti-apoptotic signalling pathway in preconditioning of skeletal myoblasts. Cardiovasc Res. 88:168–178. 2010. View Article : Google Scholar : PubMed/NCBI

66 

He WA, Calore F, Londhe P, Canella A, Guttridge DC and Croce CM: Microvesicles containing miRNAs promote muscle cell death in cancer cachexia via TLR7. Proc Natl Acad Sci USA. 111:4525–4529. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Hirai H, Verma M, Watanabe S, Tastad C, Asakura Y and Asakura A: MyoD regulates apoptosis of myoblasts through microRNA-mediated down-regulation of Pax3. J Cell Biol. 191:347–365. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO, Gonzalez M, Yancopoulos GD and Glass DJ: The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell. 14:395–403. 2004. View Article : Google Scholar : PubMed/NCBI

69 

Sugita H, Kaneki M, Sugita M, Yasukawa T, Yasuhara S and Martyn JA: Burn injury impairs insulin-stimulated Akt/PKB activation in skeletal muscle. Am J Physiol Endocrinol Metab. 288:E585–E591. 2005. View Article : Google Scholar

70 

Du K, Yu Y, Zhang D, Luo W, Huang H, Chen J, Gao J and Huang C: NFkappaB1 (p50) suppresses SOD2 expression by inhibiting FoxO3a transactivation in a miR190/PHLPP1/Akt-dependent axis. Mol Biol Cell. 24:3577–3583. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH and Goldberg AL: Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell. 117:399–412. 2004. View Article : Google Scholar : PubMed/NCBI

72 

Sheriff S, Kadeer N, Joshi R, Friend LA, James JH and Balasubramaniam A: Des-acyl ghrelin exhibits pro-anabolic and anti-catabolic effects on C2C12 myotubes exposed to cytokines and reduces burn-induced muscle proteolysis in rats. Mol Cell Endocrinol. 351:286–295. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Alexander MS, Casar JC, Motohashi N, Myers JA, Eisenberg I, Gonzalez RT, Estrella EA, Kang PB, Kawahara G and Kunkel LM: Regulation of DMD pathology by an ankyrin-encoded miRNA. Skelet Muscle. 1:272011. View Article : Google Scholar : PubMed/NCBI

74 

Chen D, Goswami CP, Burnett RM, Anjanappa M, Bhat-Nakshatri P, Muller W and Nakshatri H: Cancer affects microRNA expression, release and function in cardiac and skeletal muscle. Cancer Res. 74:4270–4281. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Hitachi K, Nakatani M and Tsuchida K: Myostatin signaling regulates Akt activity via the regulation of miR-486 expression. Int J Biochem Cell Biol. 47:93–103. 2014. View Article : Google Scholar

76 

Hudson MB, Rahnert JA, Zheng B, Woodworth-Hobbs ME, Franch HA and Price SR: miR-182 attenuates atrophy-related gene expression by targeting FoxO3 in skeletal muscle. Am J Physiol Cell Physiol. 307:C314–C319. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 13 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu Y, Chu W, Chai J, Li X, Liu L and Ma L: Critical role of miRNAs in mediating skeletal muscle atrophy (Review). Mol Med Rep 13: 1470-1474, 2016
APA
Yu, Y., Chu, W., Chai, J., Li, X., Liu, L., & Ma, L. (2016). Critical role of miRNAs in mediating skeletal muscle atrophy (Review). Molecular Medicine Reports, 13, 1470-1474. https://doi.org/10.3892/mmr.2015.4748
MLA
Yu, Y., Chu, W., Chai, J., Li, X., Liu, L., Ma, L."Critical role of miRNAs in mediating skeletal muscle atrophy (Review)". Molecular Medicine Reports 13.2 (2016): 1470-1474.
Chicago
Yu, Y., Chu, W., Chai, J., Li, X., Liu, L., Ma, L."Critical role of miRNAs in mediating skeletal muscle atrophy (Review)". Molecular Medicine Reports 13, no. 2 (2016): 1470-1474. https://doi.org/10.3892/mmr.2015.4748