Skip to main content
Log in

Noncoding RNAs in the regulation of skeletal muscle biology in health and disease

  • Review
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Skeletal muscle is composed of multinucleated myofibers that arise from the fusion of myoblasts during development. Skeletal muscle is essential for various body functions such as maintaining posture, locomotion, breathing, and metabolism. Skeletal muscle undergoes remarkable adaptations in response to environmental stimuli leading to atrophy or hypertrophy. Moreover, degeneration of skeletal muscle is a common feature in a number of muscular disorders including muscular dystrophy. Emerging evidence suggests that noncoding RNAs, such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are critical for skeletal muscle physiology. Several miRNAs and lncRNAs have now been found to control skeletal muscle development and regeneration. Noncoding RNAs also play an important role in the regulation of skeletal muscle mass in adults. Furthermore, aberrant expression of miRNAs and lncRNAs has been observed in several muscular disorders. In this article, we discuss the mechanisms of action of miRNAs and lncRNAs in skeletal muscle formation, growth, regeneration, and disease. We further highlight potential therapeutic strategies for utilizing noncoding RNAs to improve skeletal muscle function.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Buckingham M, Bajard L, Chang T, Daubas P, Hadchouel J, Meilhac S, Montarras D, Rocancourt D, Relaix F (2003) The formation of skeletal muscle: from somite to limb. J Anat 202:59–68

    Article  PubMed  PubMed Central  Google Scholar 

  2. Mauro A (1961) Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol 9:493–495

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Relaix F, Zammit PS (2012) Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns centre stage. Development 139:2845–2856

    Article  CAS  PubMed  Google Scholar 

  4. Yin H, Price F, Rudnicki MA (2013) Satellite cells and the muscle stem cell niche. Physiol Rev 93:23–67

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bentzinger CF, Wang YX, Rudnicki MA (2012) Building muscle: molecular regulation of myogenesis. Cold Spring Harb Perspect Biol 4. DOI 10.1101/cshperspect.a008342

  6. Kuang S, Rudnicki MA (2008) The emerging biology of satellite cells and their therapeutic potential. Trends Mol Med 14:82–91

    Article  CAS  PubMed  Google Scholar 

  7. Sandri M (2008) Signaling in muscle atrophy and hypertrophy. Physiology (Bethesda) 23:160–170

    Article  CAS  Google Scholar 

  8. Jackman RW, Kandarian SC (2004) The molecular basis of skeletal muscle atrophy. Am J Physiol Cell Physiol 287:C834–C843

    Article  CAS  PubMed  Google Scholar 

  9. Emery AE (1998) The muscular dystrophies. BMJ 317:991–995

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75:843–854

    Article  CAS  PubMed  Google Scholar 

  11. Krol J, Loedige I, Filipowicz W (2010) The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet 11:597–610

    CAS  PubMed  Google Scholar 

  12. Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297

    Article  CAS  PubMed  Google Scholar 

  13. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG et al (2012) The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res 22:1775–1789

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Quinn JJ, Chang HY (2015) Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet 17:47–62

    Article  Google Scholar 

  15. Pang KC, Frith MC, Mattick JS (2006) Rapid evolution of noncoding RNAs: lack of conservation does not mean lack of function. Trends Genet 22:1–5

    Article  CAS  PubMed  Google Scholar 

  16. Ma L, Bajic VB, Zhang Z (2013) On the classification of long non-coding RNAs. RNA Biol 10:925–933

    PubMed  Google Scholar 

  17. Rinn JL, Chang HY (2012) Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166

    Article  CAS  PubMed  Google Scholar 

  18. Mercer TR, Dinger ME, Mattick JS (2009) Long non-coding RNAs: insights into functions. Nat Rev Genet 10:155–159

    Article  CAS  PubMed  Google Scholar 

  19. Sempere LF, Freemantle S, Pitha-Rowe I, Moss E, Dmitrovsky E, Ambros V (2004) Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol 5:R13

    Article  PubMed  PubMed Central  Google Scholar 

  20. Baskerville S, Bartel DP (2005) Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. RNA 11:241–247

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kim HK, Lee YS, Sivaprasad U, Malhotra A, Dutta A (2006) Muscle-specific microRNA miR-206 promotes muscle differentiation. J Cell Biol 174:677–687

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM, Conlon FL, Wang DZ (2006) The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet 38:228–233

    Article  CAS  PubMed  Google Scholar 

  23. Chen JF, Tao Y, Li J, Deng Z, Yan Z, Xiao X, Wang DZ (2010) microRNA-1 and microRNA-206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. J Cell Biol 190:867–879

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rao PK, Kumar RM, Farkhondeh M, Baskerville S, Lodish HF (2006) Myogenic factors that regulate expression of muscle-specific microRNAs. Proc Natl Acad Sci U S A 103:8721–8726

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Yuasa K, Hagiwara Y, Ando M, Nakamura A, Takeda S, Hijikata T (2008) MicroRNA-206 is highly expressed in newly formed muscle fibers: implications regarding potential for muscle regeneration and maturation in muscular dystrophy. Cell Struct Funct 33: 163-169. DOI Doi 10.1247/Csf.08022

  26. Sweetman D, Goljanek K, Rathjen T, Oustanina S, Braun T, Dalmay T, Munsterberg A (2008) Specific requirements of MRFs for the expression of muscle specific microRNAs, miR-1, miR-206 and miR-133. Dev Biol 321:491–499

    Article  CAS  PubMed  Google Scholar 

  27. Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A, Bozzoni I (2011) A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 147:358–369

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lu J, McKinsey TA, Zhang CL, Olson EN (2000) Regulation of skeletal myogenesis by association of the MEF2 transcription factor with class II histone deacetylases. Mol Cell 6:233–244

    Article  CAS  PubMed  Google Scholar 

  29. McKinsey TA, Zhang CL, Lu J, Olson EN (2000) Signal-dependent nuclear export of a histone deacetylase regulates muscle differentiation. Nature 408:106–111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Soulez M, Rouviere CG, Chafey P, Hentzen D, Vandromme M, Lautredou N, Lamb N, Kahn A, Tuil D (1996) Growth and differentiation of C2 myogenic cells are dependent on serum response factor. Mol Cell Biol 16:6065–6074

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Weintraub H, Tapscott SJ, Davis RL, Thayer MJ, Adam MA, Lassar AB, Miller AD (1989) Activation of muscle-specific genes in pigment, nerve, fat, liver, and fibroblast cell lines by forced expression of MyoD. Proc Natl Acad Sci U S A 86:5434–5438

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rosenberg MI, Georges SA, Asawachaicharn A, Analau E, Tapscott SJ (2006) MyoD inhibits Fstl1 and Utrn expression by inducing transcription of miR-206. J Cell Biol 175:77–85

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hindi SM, Kumar A (2016) TRAF6 regulates satellite stem cell self-renewal and function during regenerative myogenesis. J Clin Invest 126:151–168

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  36. Alexander MS, Casar JC, Motohashi N, Myers JA, Eisenberg I, Gonzalez RT, Estrella EA, Kang PB, Kawahara G, Kunkel LM (2011) Regulation of DMD pathology by an ankyrin-encoded miRNA. Skelet Muscle 1. DOI Artn 27 10.1186/2044-5040-1-27

  37. Small EM, O’Rourke JR, Moresi V, Sutherland LB, McAnally J, Gerard RD, Richardson JA, Olson EN (2010) Regulation of PI3-kinase/Akt signaling by muscle-enriched microRNA-486. Proc Natl Acad Sci U S A 107:4218–4223

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hitachi K, Nakatani M, Tsuchida K (2014) Myostatin signaling regulates Akt activity via the regulation of miR-486 expression. Int J Biochem Cell Biol 47:93–103

    Article  CAS  PubMed  Google Scholar 

  39. Egerman MA, Glass DJ (2014) Signaling pathways controlling skeletal muscle mass. Crit Rev Biochem Mol Biol 49:59–68

    Article  CAS  PubMed  Google Scholar 

  40. McCarthy JJ (2011) The MyomiR network in skeletal muscle plasticity. Exerc Sport Sci Rev 39:150–154

    Article  PubMed  PubMed Central  Google Scholar 

  41. van Rooij E, Quiat D, Johnson BA, Sutherland LB, Qi X, Richardson JA, Kelm RJ Jr, Olson EN (2009) A family of microRNAs encoded by myosin genes governs myosin expression and muscle performance. Dev Cell 17:662–673

    Article  PubMed  PubMed Central  Google Scholar 

  42. Wang L, Chen X, Zheng Y, Li F, Lu Z, Chen C, Liu J, Wang Y, Peng Y, Shen Z et al (2012) MiR-23a inhibits myogenic differentiation through down regulation of fast myosin heavy chain isoforms. Exp Cell Res 318:2324–2334

    Article  CAS  PubMed  Google Scholar 

  43. Wang H, Garzon R, Sun H, Ladner KJ, Singh R, Dahlman J, Cheng A, Hall BM, Qualman SJ, Chandler DS et al (2008) NF-kappaB-YY1-miR-29 regulatory circuitry in skeletal myogenesis and rhabdomyosarcoma. Cancer Cell 14:369–381

    Article  CAS  PubMed  Google Scholar 

  44. Wei W, He HB, Zhang WY, Zhang HX, Bai JB, Liu HZ, Cao JH, Chang KC, Li XY, Zhao SH (2013) miR-29 targets Akt3 to reduce proliferation and facilitate differentiation of myoblasts in skeletal muscle development. Cell Death Dis 4. DOI 10.1038/cddis.2013.184

  45. Wang XH, Hu Z, Klein JD, Zhang L, Fang F, Mitch WE (2011) Decreased miR-29 suppresses myogenesis in CKD. J Am Soc Nephrol 22:2068–2076

    Article  PubMed  PubMed Central  Google Scholar 

  46. Khanna N, Ge Y, Chen J (2014) MicroRNA-146b promotes myogenic differentiation and modulates multiple gene targets in muscle cells. PLoS One 9. DOI 10.1371/journal.pone.0100657

  47. Naguibneva I, Ameyar-Zazoua M, Polesskaya A, Ait-Si-Ali S, Groisman R, Souidi M, Cuvellier S, Harel-Bellan A (2006) The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nat Cell Biol 8:278–284

    Article  CAS  PubMed  Google Scholar 

  48. Antoniou A, Mastroyiannopoulos NP, Uney JB, Phylactou LA (2014) miR-186 inhibits muscle cell differentiation through myogenin regulation. J Biol Chem 289:3923–3935

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. McFarlane C, Vajjala A, Arigela H, Lokireddy S, Ge X, Bonala S, Manickam R, Kambadur R, Sharma M (2014) Negative auto-regulation of myostatin expression is mediated by Smad3 and microRNA-27. PLoS One 9:e87687

    Article  PubMed  PubMed Central  Google Scholar 

  50. Crist CG, Montarras D, Pallafacchina G, Rocancourt D, Cumano A, Conway SJ, Buckingham M (2009) Muscle stem cell behavior is modified by microRNA-27 regulation of Pax3 expression. Proc Natl Acad Sci U S A 106:13383–13387

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Crist CG, Montarras D, Buckingham M (2012) Muscle satellite cells are primed for myogenesis but maintain quiescence with sequestration of Myf5 mRNA targeted by microRNA-31 in mRNP granules. Cell Stem Cell 11:118–126

    Article  CAS  PubMed  Google Scholar 

  52. Sun Q, Zhang Y, Yang G, Chen X, Zhang Y, Cao G, Wang J, Sun Y, Zhang P, Fan M et al (2008) Transforming growth factor-beta-regulated miR-24 promotes skeletal muscle differentiation. Nucleic Acids Res 36:2690–2699

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Winbanks CE, Wang B, Beyer C, Koh P, White L, Kantharidis P, Gregorevic P (2011) TGF-beta regulates miR-206 and miR-29 to control myogenic differentiation through regulation of HDAC4. J Biol Chem 286:13805–13814

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sato MM, Nashimoto M, Katagiri T, Yawaka Y, Tamura M (2009) Bone morphogenetic protein-2 down-regulates miR-206 expression by blocking its maturation process. Biochem Biophys Res Commun 383:125–129

    Article  CAS  PubMed  Google Scholar 

  55. Gabory A, Jammes H, Dandolo L (2010) The H19 locus: role of an imprinted non-coding RNA in growth and development. BioEssays 32:473–480

    Article  CAS  PubMed  Google Scholar 

  56. Onyango P, Feinberg AP (2011) A nucleolar protein, H19 opposite tumor suppressor (HOTS), is a tumor growth inhibitor encoded by a human imprinted H19 antisense transcript. Proc Natl Acad Sci U S A 108:16759–16764

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Dey BK, Pfeifer K, Dutta A (2014) The H19 long noncoding RNA gives rise to microRNAs miR-675-3p and miR-675-5p to promote skeletal muscle differentiation and regeneration. Genes Dev 28:491–501

    Article  PubMed  PubMed Central  Google Scholar 

  58. Mueller AC, Cichewicz MA, Dey BK, Layer R, Reon BJ, Gagan JR, Dutta A (2015) MUNC, a long noncoding RNA that facilitates the function of MyoD in skeletal myogenesis. Mol Cell Biol 35:498–513

    Article  PubMed  Google Scholar 

  59. Ballarino M, Cazzella V, D’Andrea D, Grassi L, Bisceglie L, Cipriano A, Santini T, Pinnaro C, Morlando M, Tramontano A et al (2015) Novel long noncoding RNAs (lncRNAs) in myogenesis: a miR-31 overlapping lncRNA transcript controls myoblast differentiation. Mol Cell Biol 35:728–736

    PubMed  Google Scholar 

  60. Gong C, Li Z, Ramanujan K, Clay I, Zhang Y, Lemire-Brachat S, Glass DJ (2015) A long non-coding RNA, LncMyoD, regulates skeletal muscle differentiation by blocking IMP2-mediated mRNA translation. Dev Cell 34:181–191

    Article  CAS  PubMed  Google Scholar 

  61. Watts R, Johnsen VL, Shearer J, Hittel DS (2013) Myostatin-induced inhibition of the long noncoding RNA Malat1 is associated with decreased myogenesis. Am J Physiol Cell Physiol 304:C995–C1001

    Article  CAS  PubMed  Google Scholar 

  62. Han X, Yang F, Cao H, Liang Z (2015) Malat1 regulates serum response factor through miR-133 as a competing endogenous RNA in myogenesis. FASEB J 29:3054–3064

    Article  CAS  PubMed  Google Scholar 

  63. Mousavi K, Zare H, Dell’orso S, Grontved L, Gutierrez-Cruz G, Derfoul A, Hager GL, Sartorelli V (2013) eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol Cell 51:606–617

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Caretti G, Schiltz RL, Dilworth FJ, Di Padova M, Zhao P, Ogryzko V, Fuller-Pace FV, Hoffman EP, Tapscott SJ, Sartorelli V (2006) The RNA helicases p68/p72 and the noncoding RNA SRA are coregulators of MyoD and skeletal muscle differentiation. Dev Cell 11:547–560

    Article  CAS  PubMed  Google Scholar 

  65. Wang J, Gong C, Maquat LE (2013) Control of myogenesis by rodent SINE-containing lncRNAs. Genes Dev 27:793–804

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Lu L, Sun K, Chen X, Zhao Y, Wang L, Zhou L, Sun H, Wang H (2013) Genome-wide survey by ChIP-seq reveals YY1 regulation of lincRNAs in skeletal myogenesis. EMBO J 32:2575–2588

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Nie M, Deng ZL, Liu J, Wang DZ (2015) Noncoding RNAs, Emerging Regulators of Skeletal Muscle Development and Diseases. Biomed Res Int 2015: 676575. doi: 10.1155/2015/676575

  68. Legnini I, Morlando M, Mangiavacchi A, Fatica A, Bozzoni I (2014) A feedforward regulatory loop between HuR and the long noncoding RNA linc-MD1 controls early phases of myogenesis. Mol Cell 53:506–514

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gong C, Kim YK, Woeller CF, Tang Y, Maquat LE (2009) SMD and NMD are competitive pathways that contribute to myogenesis: effects on PAX3 and myogenin mRNAs. Genes Dev 23:54–66

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Gleghorn ML, Gong C, Kielkopf CL, Maquat LE (2013) Staufen1 dimerizes through a conserved motif and a degenerate dsRNA-binding domain to promote mRNA decay. Nat Struct Mol Biol 20:515–524

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Park E, Gleghorn ML, Maquat LE (2013) Staufen2 functions in Staufen1-mediated mRNA decay by binding to itself and its paralog and promoting UPF1 helicase but not ATPase activity. Proc Natl Acad Sci U S A 110:405–412

    Article  CAS  PubMed  Google Scholar 

  72. Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA et al (2010) The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell 39:925–938

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Sartori R, Gregorevic P, Sandri M (2014) TGFbeta and BMP signaling in skeletal muscle: potential significance for muscle-related disease. Trends Endocrinol Metab 25:464–471

    Article  CAS  PubMed  Google Scholar 

  74. Hube F, Velasco G, Rollin J, Furling D, Francastel C (2011) Steroid receptor RNA activator protein binds to and counteracts SRA RNA-mediated activation of MyoD and muscle differentiation. Nucleic Acids Res 39:513–525

    Article  CAS  PubMed  Google Scholar 

  75. Giovarelli M, Bucci G, Ramos A, Bordo D, Wilusz CJ, Chen CY, Puppo M, Briata P, Gherzi R (2014) H19 long noncoding RNA controls the mRNA decay promoting function of KSRP. Proc Natl Acad Sci U S A 111:E5023–E5028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi JS, Zhang H et al (2013) The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell 52:101–112

    Article  CAS  PubMed  Google Scholar 

  77. Bodine SC, Baehr LM (2014) Skeletal muscle atrophy and the E3 ubiquitin ligases MuRF1 and MAFbx/atrogin-1. Am J Physiol Endocrinol Metab 307:E469–E484

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Sato S, Ogura Y, Kumar A (2014) TWEAK/Fn14 Signaling Axis Mediates Skeletal Muscle Atrophy and Metabolic Dysfunction. Front Immunol 5:18

    Article  PubMed  PubMed Central  Google Scholar 

  79. Panguluri SK, Bhatnagar S, Kumar A, McCarthy JJ, Srivastava AK, Cooper NG, Lundy RF, Kumar A (2010) Genomic profiling of messenger RNAs and microRNAs reveals potential mechanisms of TWEAK-induced skeletal muscle wasting in mice. PLoS One 5. doi: 10.1371/journal.pone.0008760

  80. McCarthy JJ, Esser KA, Peterson CA, Dupont-Versteegden EE (2009) Evidence of MyomiR network regulation of beta-myosin heavy chain gene expression during skeletal muscle atrophy. Physiol Genomics 39:219–226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. van Rooij E, Liu N, Olson EN (2008) MicroRNAs flex their muscles. Trends Genet 24:159–166

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Xu J, Li R, Workeneh B, Dong Y, Wang X, Hu Z (2012) Transcription factor FoxO1, the dominant mediator of muscle wasting in chronic kidney disease, is inhibited by microRNA-486. Kidney Int 82:401–411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Sharma M, Juvvuna PK, Kukreti H, McFarlane C (2014) Mega roles of microRNAs in regulation of skeletal muscle health and disease. Front Physiol 5:239

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Winbanks CE, Beyer C, Hagg A, Qian H, Sepulveda PV, Gregorevic P (2013) miR-206 represses hypertrophy of myogenic cells but not muscle fibers via inhibition of HDAC4. PLoS One 8

  87. McCarthy JJ, Esser KA (2007) MicroRNA-1 and microRNA-133a expression are decreased during skeletal muscle hypertrophy. J Appl Physiol 102:306–313

    Article  CAS  PubMed  Google Scholar 

  88. Lee SJ (2004) Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol 20:61–86

    Article  CAS  PubMed  Google Scholar 

  89. Eisenberg I, Eran A, Nishino I, Moggio M, Lamperti C, Amato AA, Lidov HG, Kang PB, North KN, Mitrani-Rosenbaum S et al (2007) Distinctive patterns of microRNA expression in primary muscular disorders. Proc Natl Acad Sci U S A 104:17016–17021

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Hoffman EP, Brown RH Jr, Kunkel LM (1987) Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 51:919–928

    Article  CAS  PubMed  Google Scholar 

  91. Blake DJ, Weir A, Newey SE, Davies KE (2002) Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev 82:291–329

    Article  CAS  PubMed  Google Scholar 

  92. Petrof BJ, Shrager JB, Stedman HH, Kelly AM, Sweeney HL (1993) Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proc Natl Acad Sci U S A 90:3710–3714

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Tawil R, Van Der Maarel SM (2006) Facioscapulohumeral muscular dystrophy. Muscle Nerve 34:1–15

    Article  CAS  PubMed  Google Scholar 

  94. Wang L, Zhou L, Jiang P, Lu L, Chen X, Lan H, Guttridge DC, Sun H, Wang H (2012) Loss of miR-29 in myoblasts contributes to dystrophic muscle pathogenesis. Mol Ther 20:1222–1233

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Alexander MS, Casar JC, Motohashi N, Vieira NM, Eisenberg I, Marshall JL, Gasperini MJ, Lek A, Myers JA, Estrella EA et al (2014) MicroRNA-486-dependent modulation of DOCK3/PTEN/AKT signaling pathways improves muscular dystrophy-associated symptoms. J Clin Invest 124:2651–2667

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Cacchiarelli D, Legnini I, Martone J, Cazzella V, D’Amico A, Bertini E, Bozzoni I (2011) miRNAs as serum biomarkers for Duchenne muscular dystrophy. Embo Mol Med 3:258–265

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Alexander MS, Kawahara G, Motohashi N, Casar JC, Eisenberg I, Myers JA, Gasperini MJ, Estrella EA, Kho AT, Mitsuhashi S et al (2013) MicroRNA-199a is induced in dystrophic muscle and affects WNT signaling, cell proliferation, and myogenic differentiation. Cell Death Differ 20:1194–1208

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Bovolenta M, Erriquez D, Valli E, Brioschi S, Scotton C, Neri M, Falzarano MS, Gherardi S, Fabris M, Rimessi P et al (2012) The DMD locus harbours multiple long non-coding RNAs which orchestrate and control transcription of muscle dystrophin mRNA isoforms. PLoS One 7:e45328

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Cabianca DS, Casa V, Bodega B, Xynos A, Ginelli E, Tanaka Y, Gabellini D (2012) A long ncRNA links copy number variation to a polycomb/trithorax epigenetic switch in FSHD muscular dystrophy. Cell 149:819–831

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We would like to apologize to the many researchers whose contributions were not cited due to space limitation or our oversight. This work was supported by funding from National Institute of Health (NIH, USA) grants AR059810, AR068313, and AG029623 to Ashok Kumar.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ashok Kumar.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Simionescu-Bankston, A., Kumar, A. Noncoding RNAs in the regulation of skeletal muscle biology in health and disease. J Mol Med 94, 853–866 (2016). https://doi.org/10.1007/s00109-016-1443-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-016-1443-y

Keywords

Navigation