Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Long noncoding RNAs in cardiac development and ageing

Key Points

  • Long noncoding RNAs (lncRNAs) comprise a vast family of noncoding RNAs that regulate gene expression through various epigenetic mechanisms mainly related to chromatin regulation

  • LncRNAs regulate multiple biological pathways in the heart

  • Deep RNA sequencing has identified hundreds of lncRNAs that are dysregulated in diseased hearts, although this change does not necessarily imply a functional effect

  • LncRNAs control the differentiation of pluripotent stem cells and cardiac precursors into cardiomyocytes and, therefore, might be useful for cardiac regeneration

  • LncRNAs are involved in cellular senescence and might be used to limit ageing-associated disease processes

  • The therapeutic or biomarker utility of lncRNAs remains to be validated

Abstract

A large part of the mammalian genome is transcribed into noncoding RNAs. Long noncoding RNAs (lncRNAs) have emerged as critical epigenetic regulators of gene expression. Distinct molecular mechanisms allow lncRNAs either to activate or to repress gene expression, thereby participating in the regulation of cellular and tissue function. LncRNAs, therefore, have important roles in healthy and diseased hearts, and might be targets for therapeutic intervention. In this Review, we summarize the current knowledge of the roles of lncRNAs in cardiac development and ageing. After describing the definition and classification of lncRNAs, we present an overview of the mechanisms by which lncRNAs regulate gene expression. We discuss the multiple roles of lncRNAs in the heart, and focus on the regulation of embryonic stem cell differentiation, cardiac cell fate and development, and cardiac ageing. We emphasize the importance of chromatin remodelling in this regulation. Finally, we discuss the therapeutic and biomarker potential of lncRNAs.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Classification of lncRNAs according to their genomic location.
Figure 2: Mechanisms of action of lncRNAs.
Figure 3: Regulation of cardiac pathways by lncRNAs.
Figure 4: Mhrt inhibits chromatin targeting and gene regulation by Brg1.

Similar content being viewed by others

References

  1. Comings, D. E. The structure and function of chromatin. Adv. Hum. Genet. 3, 237–431 (1972).

    Article  CAS  PubMed  Google Scholar 

  2. Olson, E. N. MicroRNAs as therapeutic targets and biomarkers of cardiovascular disease. Sci. Transl. Med. 6, 239ps3 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Goretti, E., Wagner, D. R. & Devaux, Y. miRNAs as biomarkers of myocardial infarction: a step forward towards personalized medicine? Trends Mol. Med. 20, 716–725 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Thum, T. Noncoding RNAs and myocardial fibrosis. Nat. Rev. Cardiol. 11, 655–663 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Ucar, A. et al. The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy. Nat. Commun. 3, 1078 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Kapusta, A. & Feschotte, C. Volatile evolution of long noncoding RNA repertoires: mechanisms and biological implications. Trends Genet. 30, 439–452 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Erdmann, V. A., Szymanski, M., Hochberg, A., de Groot, N. & Barciszewski, J. Collection of mRNA-like non-coding RNAs. Nucleic Acids Res. 27, 192–195 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Guttman, M. et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477, 295–300 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Mercer, T. R., Dinger, M. E. & Mattick, J. S. Long non-coding RNAs: insights into functions. Nat. Rev. Genet. 10, 155–159 (2009).

    Article  CAS  PubMed  Google Scholar 

  11. Amaral, P. P., Clark, M. B., Gascoigne, D. K., Dinger, M. E. & Mattick, J. S. lncRNAdb: a reference database for long noncoding RNAs. Nucleic Acids Res. 39, D146–D151 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Xie, C. et al. NONCODEv4: exploring the world of long non-coding RNA genes. Nucleic Acids Res. 42, D98–D103 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Han, P. et al. A long noncoding RNA protects the heart from pathological hypertrophy. Nature 514, 102–106 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Klattenhoff, C. A. et al. Braveheart, a long noncoding RNA required for cardiovascular lineage commitment. Cell 152, 570–583 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Matkovich, S. J., Edwards, J. R., Grossenheider, T. C., de Guzman Strong, C. & Dorn, G. W. 2nd. Epigenetic coordination of embryonic heart transcription by dynamically regulated long noncoding RNAs. Proc. Natl Acad. Sci. USA 111, 12264–12269 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ounzain, S. et al. Genome-wide profiling of the cardiac transcriptome after myocardial infarction identifies novel heart-specific long non-coding RNAs. Eur. Heart J. 36, 353–368 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Pang, K. C., Frith, M. C. & Mattick, J. S. Rapid evolution of noncoding RNAs: lack of conservation does not mean lack of function. Trends Genet. 22, 1–5 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Guo, H., Ingolia, N. T., Weissman, J. S. & Bartel, D. P. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 466, 835–840 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kapranov, P. et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 316, 1484–1488 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Clemson, C. M. et al. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol. Cell 33, 717–726 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Quinodoz, S. & Guttman, M. Long noncoding RNAs: an emerging link between gene regulation and nuclear organization. Trends Cell Biol. 24, 651–663 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, K. C. & Chang, H. Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell 43, 904–914 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mohammad, F., Mondal, T., Guseva, N., Pandey, G. K. & Kanduri, C. Kcnq1ot1 noncoding RNA mediates transcriptional gene silencing by interacting with Dnmt1. Development 137, 2493–2499 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Mancini-Dinardo, D., Steele, S. J., Levorse, J. M., Ingram, R. S. & Tilghman, S. M. Elongation of the Kcnq1ot1 transcript is required for genomic imprinting of neighboring genes. Genes Dev. 20, 1268–1282 (2006).

    CAS  Google Scholar 

  25. Pandey, R. R. et al. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol. Cell 32, 232–246 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Peters, A. H. et al. Partitioning and plasticity of repressive histone methylation states in mammalian chromatin. Mol. Cell 12, 1577–1589 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Czermin, B. et al. Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell 111, 185–196 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Mohammad, F. et al. Long noncoding RNA-mediated maintenance of DNA methylation and transcriptional gene silencing. Development 139, 2792–2803 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Korostowski, L., Sedlak, N. & Engel, N. The Kcnq1ot1 long non-coding RNA affects chromatin conformation and expression of Kcnq1, but does not regulate its imprinting in the developing heart. PLoS Genet. 8, e1002956 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tripathi, V. et al. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol. Cell 39, 925–938 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Michalik, K. M. et al. Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth. Circ. Res. 114, 1389–1397 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Ishii, N. et al. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J. Hum. Genet. 51, 1087–1099 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Ishizuka, A., Hasegawa, Y., Ishida, K., Yanaka, K. & Nakagawa, S. Formation of nuclear bodies by the lncRNA Gomafu-associating proteins Celf3 and SF1. Genes Cells 19, 704–721 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wang, K. et al. The long noncoding RNA CHRF regulates cardiac hypertrophy by targeting miR-489. Circ. Res. 114, 1377–1388 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, K. et al. CARL lncRNA inhibits anoxia-induced mitochondrial fission and apoptosis in cardiomyocytes by impairing miR-539-dependent PHB2 downregulation. Nat. Commun. 5, 3596 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Bell, R. D. et al. Identification and initial functional characterization of a human vascular cell-enriched long noncoding RNA. Arterioscler. Thromb. Vasc. Biol. 34, 1249–1259 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Grote, P. et al. The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse. Dev. Cell 24, 206–214 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Aguilo, F., Zhou, M. M. & Walsh, M. J. Long noncoding RNA, polycomb, and the ghosts haunting INK4b-ARF-INK4a expression. Cancer Res. 71, 5365–5369 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Consortium, C. A. D. et al. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat. Genet. 45, 25–33 (2013).

    Article  CAS  Google Scholar 

  40. Samani, N. J. et al. Genomewide association analysis of coronary artery disease. N. Engl. J. Med. 357, 443–453 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Vausort, M., Wagner, D. R. & Devaux, Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ. Res. 115, 668–677 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Bochenek, G. et al. The large non-coding RNA ANRIL, which is associated with atherosclerosis, periodontitis and several forms of cancer, regulates ADIPOR1, VAMP3 and C11ORF10. Hum. Mol. Genet. 22, 4516–4527 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhou, X., Chen, J. & Tang, W. The molecular mechanism of HOTAIR in tumorigenesis, metastasis, and drug resistance. Acta Biochim. Biophys. Sin. (Shanghai) 46, 1011–1015 (2014).

    Article  CAS  Google Scholar 

  45. Liu, Y. et al. Expression profiling and ontology analysis of long noncoding RNAs in post-ischemic heart and their implied roles in ischemia/reperfusion injury. Gene 543, 15–21 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Yang, K. C. et al. Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support. Circulation 129, 1009–1021 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Werber, M., Wittler, L., Timmermann, B., Grote, P. & Herrmann, B. G. The tissue-specific transcriptomic landscape of the mid-gestational mouse embryo. Development 141, 2325–2330 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Zhang, L. et al. Identification of candidate long noncoding RNAs associated with left ventricular hypertrophy. Clin. Transl. Sci. http://dx.doi.org/10.1111/cts.12234.

  49. Zangrando, J. et al. Identification of candidate long non-coding RNAs in response to myocardial infarction. BMC Genomics 15, 460 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yang, L. et al. ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structures mediates gene activation programs. Cell 147, 773–788 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Azzalin, C. M., Reichenbach, P., Khoriauli, L., Giulotto, E. & Lingner, J. Telomeric repeat containing RNA and RNA surveillance factors at mammalian chromosome ends. Science 318, 798–801 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Porro, A. et al. Functional characterization of the TERRA transcriptome at damaged telomeres. Nat. Commun. 5, 5379 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Abdelmohsen, K. et al. Senescence-associated lncRNAs: senescence-associated long noncoding RNAs. Aging Cell 12, 890–900 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Bruneau, B. G. Signaling and transcriptional networks in heart development and regeneration. Cold Spring Harb. Perspect. Biol. 5, a008292 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bulger, M. & Groudine, M. Functional and mechanistic diversity of distal transcription enhancers. Cell 144, 327–339 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Consortium, E. P. et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  57. Yue, F. et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature 515, 355–364 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Blow, M. J. et al. ChIP-Seq identification of weakly conserved heart enhancers. Nat. Genet. 42, 806–810 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Narlikar, L. et al. Genome-wide discovery of human heart enhancers. Genome Res. 20, 381–392 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. May, D. et al. Large-scale discovery of enhancers from human heart tissue. Nat. Genet. 44, 89–93 (2012).

    Article  CAS  Google Scholar 

  61. Orom, U. A. et al. Long noncoding RNAs with enhancer-like function in human cells. Cell 143, 46–58 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang, K. C. et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472, 120–124 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Ounzain, S. et al. Functional importance of cardiac enhancer-associated noncoding RNAs in heart development and disease. J. Mol. Cell. Cardiol. 76, 55–70 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wu, H. et al. Tissue-specific RNA expression marks distant-acting developmental enhancers. PLoS Genet. 10, e1004610 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wamstad, J. A. et al. Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 151, 206–220 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Matkovich, S. J., Zhang, Y., Van Booven, D. J. & Dorn, G. W. 2nd. Deep mRNA sequencing for in vivo functional analysis of cardiac transcriptional regulators: application to Gαq. Circ. Res. 106, 1459–1467 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Batista, P. J. & Chang, H. Y. Long noncoding RNAs: cellular address codes in development and disease. Cell 152, 1298–1307 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Dorn, G. W. 2nd & Matkovich, S. J. Epitranscriptional regulation of cardiovascular development and disease. J. Physiol. http://dx.doi.org/10.1113/jphysiol.2014.283234.

  69. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hang, C. T. et al. Chromatin regulation by Brg1 underlies heart muscle development and disease. Nature 466, 62–67 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Gupta, S. K., Piccoli, M. T. & Thum, T. Non-coding RNAs in cardiovascular ageing. Ageing Res. Rev. 17, 79–85 (2014).

    Article  CAS  PubMed  Google Scholar 

  72. Zhang, B. et al. The lncRNA Malat1 is dispensable for mouse development but its transcription plays a cis-regulatory role in the adult. Cell Rep. 2, 111–123 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ratajczak, M. Z. Igf2-H19, an imprinted tandem gene, is an important regulator of embryonic development, a guardian of proliferation of adult pluripotent stem cells, a regulator of longevity, and a 'passkey' to cancerogenesis. Folia Histochem. Cytobiol 50, 171–179 (2012).

    Article  CAS  PubMed  Google Scholar 

  74. Wang, G. et al. Zbtb7a suppresses prostate cancer through repression of a Sox9-dependent pathway for cellular senescence bypass and tumor invasion. Nat. Genet. 45, 739–746 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kossack, N. et al. Isolation and characterization of pluripotent human spermatogonial stem cell-derived cells. Stem Cells 27, 138–149 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee, J. E., Bennett, C. F. & Cooper, T. A. RNase H-mediated degradation of toxic RNA in myotonic dystrophy type 1. Proc. Natl Acad. Sci. USA 109, 4221–4226 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Liu, J. Y. et al. Pathogenic role of lncRNA-MALAT1 in endothelial cell dysfunction in diabetes mellitus. Cell Death Dis. 5, e1506 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Wu, G. et al. LincRNA-p21 regulates neointima formation, vascular smooth muscle cell proliferation, apoptosis, and atherosclerosis by enhancing p53 activity. Circulation 130, 1452–1465 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kumarswamy, R. et al. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ. Res. 114, 1569–1575 (2014).

    Article  CAS  PubMed  Google Scholar 

  80. Kirchhof, P. et al. The continuum of personalized cardiovascular medicine: a position paper of the European Society of Cardiology. Eur. Heart J. 35, 3250–3257 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Elashoff, M. R. et al. Development of a blood-based gene expression algorithm for assessment of obstructive coronary artery disease in non-diabetic patients. BMC Med. Genomics 4, 26 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Li, D. et al. Transcriptome analysis reveals distinct patterns of long noncoding RNAs in heart and plasma of mice with heart failure. PLoS ONE 8, e77938 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Huang, X. et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics 14, 319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lorenzen, J. M. et al. Circulating long noncoding RNA TapSAKI is a predictor of mortality in critically ill patients with acute kidney injury. Clin. Chem. 61, 191–201 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Podlowski, S., Bramlage, P., Baumann, G., Morano, I. & Luther, H. P. Cardiac troponin I sense-antisense RNA duplexes in the myocardium. J. Cell. Biochem. 85, 198–207 (2002).

    Article  CAS  PubMed  Google Scholar 

  86. Zolk, O., Solbach, T. F., Eschenhagen, T., Weidemann, A. & Fromm, M. F. Activation of negative regulators of the hypoxia-inducible factor (HIF) pathway in human end-stage heart failure. Biochem. Biophys. Res. Commun. 376, 315–320 (2008).

    Article  CAS  PubMed  Google Scholar 

  87. Carrion, K. et al. The long non-coding HOTAIR is modulated by cyclic stretch and WNT/β-CATENIN in human aortic valve cells and is a novel repressor of calcification genes. PLoS ONE 9, e96577 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Bokil, N. J., Baisden, J. M., Radford, D. J. & Summers, K. M. Molecular genetics of long QT syndrome. Mol. Genet. Metab. 101, 1–8 (2010).

    Article  CAS  PubMed  Google Scholar 

  89. Tsuiji, H. et al. Competition between a noncoding exon and introns: Gomafu contains tandem UACUAAC repeats and associates with splicing factor-1. Genes Cells 16, 479–490 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ritter, O., Haase, H., Schulte, H. D., Lange, P. E. & Morano, I. Remodeling of the hypertrophied human myocardium by cardiac bHLH transcription factors. J. Cell. Biochem. 74, 551–561 (1999).

    Article  CAS  PubMed  Google Scholar 

  91. Zhu, J. G. et al. Long noncoding RNAs expression profile of the developing mouse heart. J. Cell. Biochem. 115, 910–918 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are writing on behalf of the Cardiolinc network (http://www.cardiolinc.org/). Y.D. is supported by the Ministry of Higher Education and Research and the National Research Fund of Luxembourg. J.Z. has received a fellowship from the National Research Fund of Luxembourg (grant PhD-AFR 3972501). C.-P.C. is supported by the NIH (HL118087, HL121197), the AHA (Established Investigator Award 12EIA8960018), March of Dimes Foundation (#6-FY11-260), Indiana University (IU) School of Medicine—IU Health Strategic Research Initiative, and the IU Physician–Scientist Initiative, endowed by Lilly Endowment. G.W.D. is supported by a grant from the NIH (R01 HL108943). S.H. is supported by the European Union Commission's Seventh Framework programme under grant agreement N°305507 (HOMAGE), N°602904 (FIBROTARGETS), N°261409 (MEDIA), N°278249 (EU MASCARA), N°602156 (HECATOS), and the Marie-Curie Industry Academy Pathways and Partnerships (CARDIOMIR) N°285991. We acknowledge the Netherlands Heart Foundation, the Netherlands Organization for Scientific Research (KNAW), and the Royal Dutch Academy of Sciences (KNAW) for funding the concerted research activity of CVON Arena.

Author information

Authors and Affiliations

Consortia

Contributions

All the authors researched data for the article, discussed its contents, and wrote the manuscript. Y.D. and S.H. reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Yvan Devaux.

Ethics declarations

Competing interests

The author declare no competing financial interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

The Cardiolinc network. Long noncoding RNAs in cardiac development and ageing. Nat Rev Cardiol 12, 415–425 (2015). https://doi.org/10.1038/nrcardio.2015.55

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrcardio.2015.55

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing