Skip to main content

Non-coding RNAs in Physiological Cardiac Hypertrophy

  • Chapter
  • First Online:
Book cover Non-coding RNAs in Cardiovascular Diseases

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1229))

Abstract

Non-coding RNA (ncRNA) is a class of RNAs that are not act as translational protein templates. They are involved in the regulation of gene transcription, RNA maturation and protein translation, participating in a variety of physiological and physiological processes. NcRNAs have important functions, and are recently one of the hotspots in biomedical research. Cardiac hypertrophy is classified into physiological cardiac hypertrophy and pathological cardiac hypertrophy. Different from pathological cardiac hypertrophy, physiological cardiac hypertrophy usually developed during exercise, pregnancy, normal postnatal growth, accompanied with preservation or improvement of systolic function, while no cardiac fibrosis. In this chapter, we will briefly introduce the definition, characteristics, and functions of ncRNAs, including miRNAs, lncRNAs, and circRNAs, as well as a summary of the existing bioinformatics online databases which commonly used in the study of ncRNAs. Specially, this chapter will be focused on the characteristics and the underlying mechanisms about physiological cardiac hypertrophy. Furthermore, the regulatory mechanism of ncRNAs in physiological hypertrophy and the latest research progress will be summarized. Taken together, exploring physiologic cardiac hypertrophy-specific ncRNAs might be a unique research perspective that provides new point of view for interventions in heart failure and other cardiovascular diseases.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Chi KR. The dark side of the human genome. Nature. 2016;538(7624):275–7.

    Article  CAS  PubMed  Google Scholar 

  2. Li Y, Liang Y, Zhu Y, Zhang Y, Bei Y. Noncoding RNAs in cardiac hypertrophy. J Cardiovasc Transl Res. 2018;11(6):439–49.

    Article  PubMed  Google Scholar 

  3. Kopp F, Mendell JT. Functional classification and experimental dissection of long noncoding RNAs. Cell. 2018;172(3):393–407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Treiber T, Treiber N, Meister G. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol. 2019;20(1):5–20.

    Article  CAS  PubMed  Google Scholar 

  5. Sullenger BA, Nair S. From the RNA world to the clinic. Science. 2016;352(6292):1417–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Viereck J, Thum T. Circulating noncoding RNAs as biomarkers of cardiovascular disease and injury. Circ Res. 2017;120(2):381–99.

    Article  CAS  PubMed  Google Scholar 

  7. Barwari T, Joshi A, Mayr M. MicroRNAs in cardiovascular disease. J Am Coll Cardiol. 2016;68(23):2577–84.

    Article  CAS  PubMed  Google Scholar 

  8. Lucas T, Dimmeler S. RNA therapeutics for treatment of cardiovascular diseases: promises and challenges. Circ Res. 2016;119(7):794–7.

    Article  CAS  PubMed  Google Scholar 

  9. Creemers EE, van Rooij E. Function and therapeutic potential of noncoding RNAs in cardiac fibrosis. Circ Res. 2016;118(1):108–18.

    Article  CAS  PubMed  Google Scholar 

  10. Sallam T, Sandhu J, Tontonoz P. Long noncoding RNA discovery in cardiovascular disease: decoding form to function. Circ Res. 2018;122(1):155–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nakamura M, Sadoshima J. Mechanisms of physiological and pathological cardiac hypertrophy. Nat Rev Cardiol. 2018;15(7):387–407.

    Article  CAS  PubMed  Google Scholar 

  12. Schiattarella GG, Hill JA. Inhibition of hypertrophy is a good therapeutic strategy in ventricular pressure overload. Circulation. 2015;131(16):1435–47.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Maillet M, van Berlo JH, Molkentin JD. Molecular basis of physiological heart growth: fundamental concepts and new players. Nat Rev Mol Cell Biol. 2013;14(1):38–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bernardo BC, Weeks KL, Pretorius L, McMullen JR. Molecular distinction between physiological and pathological cardiac hypertrophy: experimental findings and therapeutic strategies. Pharmacol Ther. 2010;128(1):191–227.

    Article  CAS  PubMed  Google Scholar 

  15. Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding key mechanisms of exercise-induced cardiac protection to mitigate disease: current knowledge and emerging concepts. Phys Rev. 2018;98(1):419–75.

    CAS  Google Scholar 

  16. Bezzerides VJ, Platt C, Lerchenmuller C, Paruchuri K, Oh NL, Xiao C, Cao Y, Mann N, Spiegelman BM, Rosenzweig A. CITED4 induces physiologic hypertrophy and promotes functional recovery after ischemic injury. J Clin Investig. 2016;1(9)

    Google Scholar 

  17. Liu X, Xiao J, Zhu H, Wei X, Platt C, Damilano F, Xiao C, Bezzerides V, Bostrom P, Che L, Zhang C, Spiegelman BM, Rosenzweig A. miR-222 is necessary for exercise-induced cardiac growth and protects against pathological cardiac remodeling. Cell Metab. 2015;21(4):584–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bostrom P, Mann N, Wu J, Quintero PA, Plovie ER, Panakova D, Gupta RK, Xiao C, MacRae CA, Rosenzweig A, Spiegelman BM. C/EBPbeta controls exercise-induced cardiac growth and protects against pathological cardiac remodeling. Cell. 2010;143(7):1072–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Vega RB, Konhilas JP, Kelly DP, Leinwand LA. Molecular mechanisms underlying cardiac adaptation to exercise. Cell Metab. 2017;25(5):1012–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the Microprocessor complex. Nature. 2004;432(7014):231–5.

    Article  CAS  PubMed  Google Scholar 

  22. Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S, Kim VN. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425(6956):415–9.

    Article  CAS  PubMed  Google Scholar 

  23. Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol. 2019;20(1):21–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Pasquinelli AE. MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship. Nat Rev Genet. 2012;13(4):271–82.

    Article  CAS  PubMed  Google Scholar 

  25. Tatsuguchi M, Seok HY, Callis TE, Thomson JM, Chen JF, Newman M, Rojas M, Hammond SM, Wang DZ. Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol. 2007;42(6):1137–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cheng Y, Zhang C. MicroRNA-21 in cardiovascular disease. J Cardiovasc Transl Res. 2010;3(3):251–5.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Goretti E, Wagner DR, Devaux Y. miRNAs as biomarkers of myocardial infarction: a step forward towards personalized medicine? Trends Mol Med. 2014;20(12):716–25.

    Article  CAS  PubMed  Google Scholar 

  28. Schober A, Weber C. Mechanisms of MicroRNAs in Atherosclerosis. Annu Rev Phytopathol. 2016;11:583–616.

    Article  CAS  Google Scholar 

  29. Zhou S, Lei D, Bu F, Han H, Zhao S, Wang Y. MicroRNA-29b-3p Targets SPARC gene to protect cardiocytes against autophagy and apoptosis in hypoxic-induced H9c2 cells. J Cardiovasc Trans Res. 2019;12(4):358–65.

    Article  Google Scholar 

  30. Yin H, Zhao L, Zhang S, Zhang Y, Lei S. MicroRNA1 suppresses cardiac hypertrophy by targeting nuclear factor of activated T cells cytoplasmic 3. Mol Med Rep. 2015;12(6):8282–8.

    Article  CAS  PubMed  Google Scholar 

  31. Hua Y, Zhang Y, Ren J. IGF-1 deficiency resists cardiac hypertrophy and myocardial contractile dysfunction: role of microRNA-1 and microRNA-133a. J Cell Mol Med. 2012;16(1):83–95.

    Article  CAS  PubMed  Google Scholar 

  32. Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang ML, Segnalini P, Gu Y, Dalton ND, Elia L, Latronico MV, Hoydal M, Autore C, Russo MA, Dorn GW 2nd, Ellingsen O, Ruiz-Lozano P, Peterson KL, Croce CM, Peschle C, Condorelli G. MicroRNA-133 controls cardiac hypertrophy. Nat Med. 2007;13(5):613–8.

    Article  CAS  PubMed  Google Scholar 

  33. Li R, Yan G, Zhang Q, Jiang Y, Sun H, Hu Y, Sun J, Xu B. miR-145 inhibits isoproterenol-induced cardiomyocyte hypertrophy by targeting the expression and localization of GATA6. FEBS Lett. 2013;587(12):1754–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cha MJ, Jang JK, Ham O, Song BW, Lee SY, Lee CY, Park JH, Lee J, Seo HH, Choi E, Jeon WM, Hwang HJ, Shin HT, Choi E, Hwang KC. MicroRNA-145 suppresses ROS-induced Ca2+ overload of cardiomyocytes by targeting CaMKIIdelta. Biochem Biophys Res Commun. 2013;435(4):720–6.

    Article  CAS  PubMed  Google Scholar 

  35. Zaglia T, Ceriotti P, Campo A, Borile G, Armani A, Carullo P, Prando V, Coppini R, Vida V, Stolen, TO, Ulrik W, Cerbai E, Stellin G, Faggian G, De Stefani D, Sandri M, Rizzuto R, Di Lisa F, Pozzan T, Catalucci D, Mongillo M. Content of mitochondrial calcium uniporter (MCU) in cardiomyocytes is regulated by microRNA-1 in physiologic and pathologic hypertrophy. Proc Natl Acad Sci USA. 2017;114(43):E9006–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ooi JYY, Bernardo BC, Singla S, Patterson NL, Lin RCY, McMullen JR. Identification of miR-34 regulatory networks in settings of disease and antimiR-therapy: Implications for treating cardiac pathology and other diseases. RNA Biol. 2017;14(5):500–13.

    Article  PubMed  Google Scholar 

  37. Tony H, Meng K, Wu B, Yu A, Zeng Q, Yu K, Zhong Y. MicroRNA-208a dysregulates apoptosis genes expression and promotes cardiomyocyte apoptosis during ischemia and its silencing improves cardiac function after myocardial infarction. Mediat Inflamm. 2015;2015:479123.

    Article  CAS  Google Scholar 

  38. Diniz GP, Takano AP, Barreto-Chaves ML. MiRNA-208a and miRNA-208b are triggered in thyroid hormone-induced cardiac hypertrophy – role of type 1 Angiotensin II receptor (AT1R) on miRNA-208a/alpha-MHC modulation. J Mol Endocrinol. 2013;374(1–2):117–24.

    Article  CAS  Google Scholar 

  39. Seok HY, Chen J, Kataoka M, Huang ZP, Ding J, Yan J, Hu X, Wang DZ. Loss of MicroRNA-155 protects the heart from pathological cardiac hypertrophy. Circ Res. 2014;114(10):1585–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kong C, Sun L, Zhang M, Ding L, Zhang Q, Cheng X, Diao Z, Yan Q, Zhang H, Fang T, Zhen X, Hu Y, Sun H, Yan G. miR-133b reverses the hydrosalpinx-induced impairment of embryo attachment through down-regulation of SGK1. J Clin Endocrinol Metab. 2016;101(4):1478–89.

    Article  CAS  PubMed  Google Scholar 

  41. Li Z, Song Y, Liu L, Hou N, An X, Zhan D, Li Y, Zhou L, Li P, Yu L, Xia J, Zhang Y, Wang J, Yang X. miR-199a impairs autophagy and induces cardiac hypertrophy through mTOR activation. Cell Death Differ. 2017;24(7):1205–13.

    Article  CAS  PubMed  Google Scholar 

  42. Yang Z, Kaye DM. Mechanistic insights into the link between a polymorphism of the 3′UTR of the SLC7A1 gene and hypertension. Hum Mutat. 2009;30(3):328–33.

    Article  PubMed  CAS  Google Scholar 

  43. Aryal B, Singh AK, Rotllan N, Price N, Fernandez-Hernando C. MicroRNAs and lipid metabolism. Curr Opin Lipidol. 2017;28(3):273–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Novak J, Olejnickova V, Tkacova N, Santulli G. Mechanistic role of microRNAs in coupling lipid metabolism and atherosclerosis. Adv Exp Med Biol. 2015;887:79–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Creemers EE, Tijsen AJ, Pinto YM. Circulating microRNAs: novel biomarkers and extracellular communicators in cardiovascular disease? Circ Res. 2012;110(3):483–95.

    Article  CAS  PubMed  Google Scholar 

  46. Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol Rev. 2016;96(4):1297–325.

    Article  CAS  PubMed  Google Scholar 

  47. Huang JZ, Chen M, Chen GXC, Zhu S, Huang H, Hu M, Zhu H, Yan GR. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 2017;68(1):171–84.. e176

    Article  CAS  PubMed  Google Scholar 

  48. Nelson BR, Makarewich CA, Anderson DM, Winders BR, Troupes CD, Wu F, Reese AL, McAnally JR, Chen X, Kavalali ET, Cannon SC, Houser SR, Bassel-Duby R, Olson EN. A peptide encoded by a transcript annotated as long noncoding RNA enhances SERCA activity in muscle. Science. 2016;351(6270):271–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Devaux Y, Zangrando J, Schroen B, Creemers EE, Pedrazzini T, Chang CP, Dorn GW 2nd, Thum T, Heymans S, Cardiolinc n. Long noncoding RNAs in cardiac development and ageing. Nat Rev Cardiol. 2015;12(7):415–25.

    Article  CAS  PubMed  Google Scholar 

  50. Viereck J, Thum T. Long Noncoding RNAs in Pathological Cardiac Remodeling. Circ Res. 2017;120(2):262–4.

    Article  CAS  PubMed  Google Scholar 

  51. Zhang D, Wang B, Ma M, Yu K, Zhang Q, Zhang X. lncRNA HOTAIR protects myocardial infarction rat by sponging miR-519d-3p. J Cardiovasc Transl Res. 2019;12(3):171–83.

    Article  PubMed  Google Scholar 

  52. Wang Z, Zhang XJ, Ji YX, Zhang P, Deng KQ, Gong J, Ren S, Wang X, Chen I, Wang H, Gao C, Yokota T, Ang YS, Li S, Cass A, Vondriska TM, Li G, Deb A, Srivastava D, Yang HT, Xiao X, Li H, Wang Y. The long noncoding RNA Chaer defines an epigenetic checkpoint in cardiac hypertrophy. Nat Med. 2016;22(10):1131–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang K, Liu F, Zhou LY, Long B, Yuan SM, Wang Y, Liu CY, Sun T, Zhang XJ, Li PF. The long noncoding RNA CHRF regulates cardiac hypertrophy by targeting miR-489. Circ Res. 2014;114(9):1377–88.

    Article  CAS  PubMed  Google Scholar 

  54. Han P, Li W, Lin CH, Yang J, Shang C, Nuernberg ST, Jin KK, Xu W, Lin CY, Lin CJ, Xiong Y, Chien H, Zhou B, Ashley E, Bernstein D, Chen PS, Chen HV, Quertermous T, Chang CP. A long noncoding RNA protects the heart from pathological hypertrophy. Nature. 2014;514(7520):102–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Sallam T, Jones M, Thomas BJ, Wu X, Gilliland T, Qian K, Eskin A, Casero D, Zhang Z, Sandhu J, Salisbury D, Rajbhandari P, Civelek M, Hong C, Ito A, Liu X, Daniel B, Lusis AJ, Whitelegge J, Nagy L, Castrillo A, Smale S, Tontonoz P. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat Med. 2018;24(3):304–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wu G, Cai J, Han Y, Chen J, Huang ZP, Chen C, Cai Y, Huang H, Yang Y, Liu Y, Xu Z, He D, Zhang X, Hu X, Pinello L, Zhong D, He F, Yuan GC, Wang DZ, Zeng C. LincRNA-p21 regulates neointima formation, vascular smooth muscle cell proliferation, apoptosis, and atherosclerosis by enhancing p53 activity. Circulation. 2014;130(17):1452–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Piccoli MT, Gupta SK, Viereck J, Foinquinos A, Samolovac S, Kramer FL, Garg A, Remke J, Zimmer K, Batkai S, Thum T. Inhibition of the cardiac fibroblast-enriched lncRNA Meg3 prevents cardiac fibrosis and diastolic dysfunction. Circ Res. 2017;121(5):575–83.

    Article  CAS  PubMed  Google Scholar 

  58. Qu X, Du Y, Shu Y, Gao M, Sun F, Luo S, Yang T, Zhan L, Yuan Y, Chu W, Pan Z, Wang Z, Yang B, Lu Y. MIAT Is a pro-fibrotic long non-coding RNA governing cardiac fibrosis in post-infarct myocardium. Sci Rep. 2017;7:42657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. de Gonzalo-Calvo D, Kenneweg F, Bang C, Toro R, van der Meer RW, Rijzewijk LJ, Smit JW, Lamb HJ, Llorente-Cortes V, Thum T. Circulating long-non coding RNAs as biomarkers of left ventricular diastolic function and remodelling in patients with well-controlled type 2 diabetes. Sci Rep. 2016;6:37354.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z, Sharpless NE. Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk. PLoS Genet. 2010;6(12):e1001233.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Guo JU, Agarwal V, Guo H, Bartel DP. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15(7):409.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs. Nat Biotechnol. 2014;32(5):453–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495(7441):384–8.

    Article  CAS  PubMed  Google Scholar 

  64. Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, Loewer A, Ziebold U, Landthaler M, Kocks C, le Noble F, Rajewsky N. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.

    Article  CAS  PubMed  Google Scholar 

  65. Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L, Chen LL. Circular intronic long noncoding RNAs. Mol Cell. 2013;51(6):792–806.

    Article  CAS  PubMed  Google Scholar 

  66. Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, Zhu P, Chang Z, Wu Q, Zhao Y, Jia Y, Xu P, Liu H, Shan G. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22(3):256–64.

    Article  PubMed  CAS  Google Scholar 

  67. Ebbesen KK, Hansen TB, Kjems J. Insights into circular RNA biology. RNA Biol. 2017;14(8):1035–45.

    Article  PubMed  Google Scholar 

  68. Li X, Yang L, Chen LL. The biogenesis, functions, and challenges of circular RNAs. Mol Cell. 2018;71(3):428–42.

    Article  CAS  PubMed  Google Scholar 

  69. Zheng Q, Bao C, Guo W, Li S, Chen J, Chen B, Luo Y, Lyu D, Li Y, Shi G, Liang L, Gu J, He X, Huang S. Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs. Nat Commun. 2016;7:11215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, Kohlmaier A, Herbst A, Northoff BH, Nicolaou A, Gabel G, Beutner F, Scholz M, Thiery J, Musunuru K, Krohn K, Mann M, Teupser D. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat Commun. 2016;7:12429.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Zhu P, Zhu X, Wu J, He L, Lu T, Wang Y, Liu B, Ye B, Sun L, Fan D, Wang J, Yang L, Qin X, Du Y, Li C, He L, Ren W, Wu X, Tian Y, Fan Z. IL-13 secreted by ILC2s promotes the self-renewal of intestinal stem cells through circular RNA circPan3. Nat Immunol. 2019;20(2):183–94.

    Article  CAS  PubMed  Google Scholar 

  72. Yang Y, Gao X, Zhang M, Yan S, Sun C, Xiao F, Huang N, Yang X, Zhao K, Zhou H, Huang S, Xie B, Zhang N. Novel role of FBXW7 circular RNA in repressing glioma tumorigenesis. J Natl Cancer Inst. 2018;110(3):304–15.

    Article  CAS  Google Scholar 

  73. Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, Fatica A, Santini T, Andronache A, Wade M, Laneve P, Rajewsky N, Bozzoni I. Circ-ZNF609 Is a circular RNA that can be translated and functions in myogenesis. Mol Cell. 2017;66(1):22–37.. e29

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, Hanan M, Wyler E, Perez-Hernandez D, Ramberger E, Shenzis S, Samson M, Dittmar G, Landthaler M, Chekulaeva M, Rajewsky N, Kadener S. Translation of CircRNAs. Mol Cell. 2017;66(1):9–21.. e27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kristensen LS, Hansen TB, Veno MT, Kjems J. Circular RNAs in cancer: opportunities and challenges in the field. Oncogene. 2018;37(5):555–65.

    Article  CAS  PubMed  Google Scholar 

  76. Lu D, Xu AD. Mini review: circular RNAs as potential clinical biomarkers for disorders in the central nervous system. Front Genet. 2016;7:53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Devaux Y, Creemers EE, Boon RA, Werfel S, Thum T, Engelhardt S, Dimmeler S, Squire I, Cardiolinc n. Circular RNAs in heart failure. Eur J Heart Fail. 2017;19(6):701–9.

    Article  CAS  PubMed  Google Scholar 

  78. Li M, Ding W, Sun T, Tariq MA, Xu T, Li P, Wang J. Biogenesis of circular RNAs and their roles in cardiovascular development and pathology. FEBS J. 2018;285(2):220–32.

    Article  CAS  PubMed  Google Scholar 

  79. Geng HH, Li R, Su YM, Xiao J, Pan M, Cai XX, Ji XP. The circular RNA Cdr1as promotes myocardial infarction by mediating the regulation of miR-7a on its target genes expression. PLoS One. 2016;11(3):e0151753.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Wang K, Gan TY, Li N, Liu CY, Zhou LY, Gao JN, Chen C, Yan KW, Ponnusamy M, Zhang YH, Li PF. Circular RNA mediates cardiomyocyte death via miRNA-dependent upregulation of MTP18 expression. Cell Death Differ. 2017;24(6):1111–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Cai L, Qi B, Wu X, Peng S, Zhou G, Wei Y, Xu J, Chen S, Liu S. Circular RNA Ttc3 regulates cardiac function after myocardial infarction by sponging miR-15b. J Mol Cell Cardiol. 2019;130:10–22.

    Article  CAS  PubMed  Google Scholar 

  82. Wang K, Long B, Liu F, Wang JX, Liu CY, Zhao B, Zhou LY, Sun T, Wang M, Yu T, Gong Y, Liu J, Dong YH, Li N, Li PF. A circular RNA protects the heart from pathological hypertrophy and heart failure by targeting miR-223. Eur Heart J. 2016;37(33):2602–11.

    Article  CAS  PubMed  Google Scholar 

  83. Tang CM, Zhang M, Huang L, Hu ZQ, Zhu JN, Xiao Z, Zhang Z, Lin QX, Zheng XL, Yang M, Wu SL, Cheng JD, Shan ZX. CircRNA_000203 enhances the expression of fibrosis-associated genes by derepressing targets of miR-26b-5p, Col1a2 and CTGF, in cardiac fibroblasts. Sci Rep. 2017;7:40342.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Zhou B, Yu JW. A novel identified circular RNA, circRNA_010567, promotes myocardial fibrosis via suppressing miR-141 by targeting TGF-beta1. Biochem Biophys Res Commun. 2017;487(4):769–75.

    Article  CAS  PubMed  Google Scholar 

  85. Zeng Y, Du WW, Wu Y, Yang Z, Awan FM, Li X, Yang W, Zhang C, Yang Q, Yee A, Chen Y, Yang F, Sun H, Huang R, Yee AJ, Li RK, Wu Z, Backx PH, Yang BB. A circular RNA binds to and activates AKT phosphorylation and nuclear localization reducing apoptosis and enhancing cardiac repair. Theranostics. 2017;7(16):3842–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Du WW, Yang W, Chen Y, Wu ZK, Foster FS, Yang Z, Li X, Yang BB. Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses. Eur Heart J. 2017;38(18):1402–12.

    CAS  PubMed  Google Scholar 

  87. Huang S, Li X, Zheng H, Si X, Li B, Wei G, Li C, Chen Y, Chen Y, Liao W, Liao Y, Bin J. Loss of super-enhancer-regulated CircRNA Nfix induces cardiac regeneration after myocardial infarction in adult mice. Circulation. 2019;139(25):2857–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wang L, Lv Y, Li G, Xiao J. MicroRNAs in heart and circulation during physical exercise. J Sport Health Sci. 2018;7(4):433–41.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Mach N, Fuster-Botella D. Endurance exercise and gut microbiota: A review. J Sport Health Sci. 2017;6(2):179–97.

    Article  PubMed  Google Scholar 

  90. Fiuza-Luces C, Santos-Lozano A, Joyner M, Carrera-Bastos P, Picazo O, Zugaza JL, Izquierdo M, Ruilope LM, Lucia A. Exercise benefits in cardiovascular disease: beyond attenuation of traditional risk factors. Nat Rev Cardiol. 2018;15(12):731–43.

    Article  CAS  PubMed  Google Scholar 

  91. Nieman DC, Wentz LM. The compelling link between physical activity and the body’s defense system. J Sport Health Sci. 2019;8(3):201–17.

    Article  PubMed  Google Scholar 

  92. Weeks KL, McMullen JR. The athlete’s heart vs. the failing heart: can signaling explain the two distinct outcomes? Physiology (Bethesda). 2011;26(2):97–105.

    CAS  Google Scholar 

  93. McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Kang PM, Izumo S. Phosphoinositide 3-kinase(p110alpha) plays a critical role for the induction of physiological, but not pathological, cardiac hypertrophy. Proc Natl Acad Sci U S A. 2003;100(21):12355–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. DeBosch B, Treskov I, Lupu TS, Weinheimer C, Kovacs A, Courtois M, Muslin AJ. Akt1 is required for physiological cardiac growth. Circulation. 2006;113(17):2097–104.

    Article  CAS  PubMed  Google Scholar 

  95. Neri Serneri GG, Boddi M, Modesti PA, Cecioni I, Coppo M, Padeletti L, Michelucci A, Colella A, Galanti G. Increased cardiac sympathetic activity and insulin-like growth factor-I formation are associated with physiological hypertrophy in athletes. Circ Res. 2001;89(11):977–82.

    Article  CAS  PubMed  Google Scholar 

  96. Olszanecka A, Dragan A, Kawecka-Jaszcz K, Fedak D, Czarnecka D. Relationships of insulin-like growth factor-1, its binding proteins, and cardiometabolic risk in hypertensive perimenopausal women. Metabolism. 2017;69:96–106.

    Article  CAS  PubMed  Google Scholar 

  97. Riehle C, Wende AR, Zhu Y, Oliveira KJ, Pereira RO, Jaishy BP, Bevins J, Valdez S, Noh J, Kim BJ, Moreira AB, Weatherford ET, Manivel R, Rawlings TA, Rech M, White MF, Abel ED. Insulin receptor substrates are essential for the bioenergetic and hypertrophic response of the heart to exercise training. Mol Cell Biol. 2014;34(18):3450–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Kim J, Wende AR, Sena S, Theobald HA, Soto J, Sloan C, Wayment BE, Litwin SE, Holzenberger M, LeRoith D, Abel ED. Insulin-like growth factor I receptor signaling is required for exercise-induced cardiac hypertrophy. J Mol Endocrinol. 2008;22(11):2531–43.

    Article  CAS  Google Scholar 

  99. Weeks KL, Gao X, Du XJ, Boey EJ, Matsumoto A, Bernardo BC, Kiriazis H, Cemerlang N, Tan JW, Tham YK, Franke TF, Qian H, Bogoyevitch MA, Woodcock EA, Febbraio MA, Gregorevic P, McMullen JR. Phosphoinositide 3-kinase p110alpha is a master regulator of exercise-induced cardioprotection and PI3K gene therapy rescues cardiac dysfunction. Circ Heart Fail. 2012;5(4):523–34.

    Article  CAS  PubMed  Google Scholar 

  100. McMullen JR, Amirahmadi F, Woodcock EA, Schinke-Braun M, Bouwman RD, Hewitt KA, Mollica JP, Zhang L, Zhang Y, Shioi T, Buerger A, Izumo S, Jay PY, Jennings GL. Protective effects of exercise and phosphoinositide 3-kinase(p110alpha) signaling in dilated and hypertrophic cardiomyopathy. Proc Natl Acad Sci U S A. 2007;104(2):612–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Gosselin H, Beliveau L, Burelle Y, Clement R, Lajoie C, El-Helou V, Calderone A. Disparate regulation of signaling proteins after exercise and myocardial infarction. Med Sci Sports Exerc. 2006;38(3):455–62.

    Article  CAS  PubMed  Google Scholar 

  102. Gonzalez AM, Osorio JC, Manlhiot C, Gruber D, Homma S, Mital S. Hypertrophy signaling during peripartum cardiac remodeling. Am J Phys Heart Circ Phys. 2007;293(5):H3008–13.

    CAS  Google Scholar 

  103. O’Neill BT, Kim J, Wende AR, Theobald HA, Tuinei J, Buchanan J, Guo A, Zaha VG, Davis DK, Schell JC, Boudina S, Wayment B, Litwin SE, Shioi T, Izumo S, Birnbaum MJ, Abel ED. A conserved role for phosphatidylinositol 3-kinase but not Akt signaling in mitochondrial adaptations that accompany physiological cardiac hypertrophy. Cell Metab. 2007;6(4):294–306.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Fisher DA, Klein AH. Thyroid development and disorders of thyroid function in the newborn. N Engl J Med. 1981;304(12):702–12.

    Article  CAS  PubMed  Google Scholar 

  105. Chang KC, Figueredo VM, Schreur JH, Kariya K, Weiner MW, Simpson PC, Camacho SA. Thyroid hormone improves function and Ca2+ handling in pressure overload hypertrophy. Association with increased sarcoplasmic reticulum Ca2+-ATPase and alpha-myosin heavy chain in rat hearts. J Clin Investig. 1997;100(7):1742–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Trivieri MG, Oudit GY, Sah R, Kerfant BG, Sun H, Gramolini AO, Pan Y, Wickenden AD, Croteau W, Morreale de Escobar G, Pekhletski R, St Germain D, Maclennan DH, Backx PH. Cardiac-specific elevations in thyroid hormone enhance contractility and prevent pressure overload-induced cardiac dysfunction. Proc Natl Acad Sci U S A. 2006;103(15):6043–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Belke DD, Gloss B, Swanson EA, Dillmann WH. Adeno-associated virus-mediated expression of thyroid hormone receptor isoforms-alpha1 and -beta1 improves contractile function in pressure overload-induced cardiac hypertrophy. Endocrinology. 2007;148(6):2870–7.

    Article  CAS  PubMed  Google Scholar 

  108. Fernandes T, Hashimoto NY, Magalhaes FC, Fernandes FB, Casarini DE, Carmona AK, Krieger JE, Phillips MI, Oliveira EM. Aerobic exercise training-induced left ventricular hypertrophy involves regulatory MicroRNAs, decreased angiotensin-converting enzyme-angiotensin ii, and synergistic regulation of angiotensin-converting enzyme 2-angiotensin (1-7). Hypertension. 2011;58(2):182–9.

    Article  CAS  PubMed  Google Scholar 

  109. Ramasamy S, Velmurugan G, Shanmugha Rajan K, Ramprasath T, Kalpana K. MiRNAs with apoptosis regulating potential are differentially expressed in chronic exercise-induced physiologically hypertrophied hearts. PLoS One. 2015;10(3):e0121401.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Ma Z, Qi J, Meng S, Wen B, Zhang J. Swimming exercise training-induced left ventricular hypertrophy involves microRNAs and synergistic regulation of the PI3K/AKT/mTOR signaling pathway. Eur J Appl Physiol. 2013;113(10):2473–86.

    Article  CAS  PubMed  Google Scholar 

  111. Martinelli NC, Cohen CR, Santos KG, Castro MA, Biolo A, Frick L, Silvello D, Lopes A, Schneider S, Andrades ME, Clausell N, Matte U, Rohde LE. An analysis of the global expression of microRNAs in an experimental model of physiological left ventricular hypertrophy. PLoS One. 2014;9(4):e93271.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Shi J, Bei Y, Kong X, Liu X, Lei Z, Xu T, Wang H, Xuan Q, Chen P, Xu J, Che L, Liu H, Zhong J, Sluijter JP, Li X, Rosenzweig A, Xiao J. miR-17-3p contributes to exercise-induced cardiac growth and protects against myocardial ischemia-reperfusion injury. Theranostics. 2017;7(3):664–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Yang L, Li Y, Wang X, Mu X, Qin D, Huang W, Alshahrani S, Nieman M, Peng J, Essandoh K, Peng T, Wang Y, Lorenz J, Soleimani M, Zhao ZQ, Fan GC. Overexpression of miR-223 tips the balance of pro- and anti-hypertrophic signaling cascades toward physiologic cardiac hypertrophy. J Biol Chem. 2016;291(30):15700–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Li Z, Liu L, Hou N, Song Y, An X, Zhang Y, Yang X, Wang J. miR-199-sponge transgenic mice develop physiological cardiac hypertrophy. Cardiovasc Res. 2016;110(2):258–67.

    Article  CAS  PubMed  Google Scholar 

  115. Xu Y, Bei Y, Shen S, Zhang J, Lu Y, Xiao J, Li X. MicroRNA-222 promotes the proliferation of pulmonary arterial smooth muscle cells by targeting P27 and TIMP3. Cell Physiol Biochem. 2017;43(1):282–92.

    Article  CAS  PubMed  Google Scholar 

  116. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34(Database issue):D140–4.

    Article  CAS  PubMed  Google Scholar 

  117. Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic Acids Res. 2019;47(D1):D155–62.

    Article  CAS  PubMed  Google Scholar 

  118. Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 2011;39(Database issue):D152–7.

    Article  CAS  PubMed  Google Scholar 

  119. Kruger J, Rehmsmeier M. RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic Acids Res. 2006;34(Web Server issue):W451–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42(Database issue):D92–7.

    Article  CAS  PubMed  Google Scholar 

  121. Yang JH, Li JH, Shao P, Zhou H, Chen YQ, Qu LH. starBase: a database for exploring microRNA-mRNA interaction maps from Argonaute CLIP-Seq and Degradome-Seq data. Nucleic Acids Res. 2011;39(Database issue):D202–9.

    Article  CAS  PubMed  Google Scholar 

  122. Yang JH, Li JH, Jiang S, Zhou H, Qu LH. ChIPBase: a database for decoding the transcriptional regulation of long non-coding RNA and microRNA genes from ChIP-Seq data. Nucleic Acids Res. 2013;41(Database issue):D177–87.

    Article  CAS  PubMed  Google Scholar 

  123. Zhou KR, Liu S, Sun WJ, Zheng LL, Zhou H, Yang JH, Qu LH. ChIPBase v2.0: decoding transcriptional regulatory networks of non-coding RNAs and protein-coding genes from ChIP-seq data. Nucleic Acids Res. 2017;45(D1):D43–50.

    Article  CAS  PubMed  Google Scholar 

  124. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15–20.

    Article  CAS  PubMed  Google Scholar 

  125. Volders PJ, Anckaert J, Verheggen K, Nuytens J, Martens L, Mestdagh P, Vandesompele J. LNCipedia 5: towards a reference set of human long non-coding RNAs. Nucleic Acids Res. 2019;47(D1):D135–9.

    Article  CAS  PubMed  Google Scholar 

  126. Volders PJ, Helsens K, Wang X, Menten B, Martens L, Gevaert K, Vandesompele J, Mestdagh P. LNCipedia: a database for annotated human lncRNA transcript sequences and structures. Nucleic Acids Res. 2013;41(Database issue):D246–51.

    Article  CAS  PubMed  Google Scholar 

  127. Volders PJ, Verheggen K, Menschaert G, Vandepoele K, Martens L, Vandesompele J, Mestdagh P. An update on LNCipedia: a database for annotated human lncRNA sequences. Nucleic Acids Res. 2015;43(8):4363–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Liu K, Yan Z, Li Y, Sun Z. Linc2GO: a human LincRNA function annotation resource based on ceRNA hypothesis. Bioinformatics. 2013;29(17):2221–2.

    Article  CAS  PubMed  Google Scholar 

  129. Zhao Y, Li H, Fang S, Kang Y, Wu W, Hao Y, Li Z, Bu D, Sun N, Zhang MQ, Chen R. NONCODE 2016: an informative and valuable data source of long non-coding RNAs. Nucleic Acids Res. 2016;44(D1):D203–8.

    Article  CAS  PubMed  Google Scholar 

  130. Liu C, Bai B, Skogerbo G, Cai L, Deng W, Zhang Y, Bu D, Zhao Y, Chen R. NONCODE: an integrated knowledge database of non-coding RNAs. Nucleic Acids Res. 2005;33(Database issue):D112–5.

    Article  CAS  PubMed  Google Scholar 

  131. Xiyuan L, Dechao B, Liang S, Yang W, Shuangsang F, Hui L, Haitao L, Chunlong L, Wenzheng F, Runsheng C, Yi Z. Using the NONCODE database resource. Curr Protoc Bioinformatics. 2017;58:12 16 11–9.

    Article  Google Scholar 

  132. Fang S, Zhang L, Guo J, Niu Y, Wu Y, Li H, Zhao L, Li X, Teng X, Sun X, Sun L, Zhang MQ, Chen R, Zhao Y. NONCODEV5: a comprehensive annotation database for long non-coding RNAs. Nucleic Acids Res. 2018;46(D1):D308–14.

    Article  CAS  PubMed  Google Scholar 

  133. Bu D, Yu K, Sun S, Xie C, Skogerbo G, Miao R, Xiao H, Liao Q, Luo H, Zhao G, Zhao H, Liu Z, Liu C, Chen R, Zhao Y. NONCODE v3.0: integrative annotation of long noncoding RNAs. Nucleic Acids Res. 2012;40(Database issue):D210–5.

    Article  CAS  PubMed  Google Scholar 

  134. He S, Liu C, Skogerbo G, Zhao H, Wang J, Liu T, Bai B, Zhao Y, Chen R. NONCODE v2.0: decoding the non-coding. Nucleic Acids Res. 2008;36(Database issue):D170–2.

    CAS  PubMed  Google Scholar 

  135. Glazar P, Papavasileiou P, Rajewsky N. circBase: a database for circular RNAs. RNA. 2014;20(11):1666–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Dong R, Ma XK, Li GW, Yang L. CIRCpedia v2: an updated database for comprehensive circular RNA annotation and expression comparison. Genomics Proteomics Bioinformatics. 2018;16(4):226–33.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Panda AC, Dudekula DB, Abdelmohsen K, Gorospe M. Analysis of circular RNAs using the web tool circInteractome. Methods Mol Biol. 2018;1724:43–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Dudekula DB, Panda AC, Grammatikakis I, De S, Abdelmohsen K, Gorospe M. CircInteractome: A web tool for exploring circular RNAs and their interacting proteins and microRNAs. RNA Biol. 2016;13(1):34–42.

    Article  PubMed  Google Scholar 

  139. Liu M, Wang Q, Shen J, Yang BB, Ding X. Circbank: a comprehensive database for circRNA with standard nomenclature. RNA Biol. 2019;16(7):899–905.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the grants from National Natural Science Foundation of China (81722008, 91639101 and 81570362 to JJ Xiao, 81800358 to LJ Wang), Innovation Program of Shanghai Municipal Education Commission (2017-01-07-00-09-E00042 to JJ Xiao), the grant from Science and Technology Commission of Shanghai Municipality (17010500100, 18410722200 to JJ Xiao), Natural Science Foundation of Shanghai (19ZR1474100 to LJ Wang), National Key Research and Development Project (2018YFE0113500 to JJ Xiao), and the “Dawn” Program of Shanghai Education Commission (19SG34 to JJ Xiao).

Competing Financial Interests

The authors declare no competing financial interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Junjie Xiao .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2020 Springer Nature Singapore Pte Ltd.

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Wang, L., Wang, J., Li, G., Xiao, J. (2020). Non-coding RNAs in Physiological Cardiac Hypertrophy. In: Xiao, J. (eds) Non-coding RNAs in Cardiovascular Diseases. Advances in Experimental Medicine and Biology, vol 1229. Springer, Singapore. https://doi.org/10.1007/978-981-15-1671-9_8

Download citation

Publish with us

Policies and ethics