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.

  • Letter
  • Published:

A long noncoding RNA protects the heart from pathological hypertrophy

This article has been updated

Abstract

The role of long noncoding RNA (lncRNA) in adult hearts is unknown; also unclear is how lncRNA modulates nucleosome remodelling. An estimated 70% of mouse genes undergo antisense transcription1, including myosin heavy chain 7 (Myh7), which encodes molecular motor proteins for heart contraction2. Here we identify a cluster of lncRNA transcripts from Myh7 loci and demonstrate a new lncRNA–chromatin mechanism for heart failure. In mice, these transcripts, which we named myosin heavy-chain-associated RNA transcripts (Myheart, or Mhrt), are cardiac-specific and abundant in adult hearts. Pathological stress activates the Brg1–Hdac–Parp chromatin repressor complex3 to inhibit Mhrt transcription in the heart. Such stress-induced Mhrt repression is essential for cardiomyopathy to develop: restoring Mhrt to the pre-stress level protects the heart from hypertrophy and failure. Mhrt antagonizes the function of Brg1, a chromatin-remodelling factor that is activated by stress to trigger aberrant gene expression and cardiac myopathy3. Mhrt prevents Brg1 from recognizing its genomic DNA targets, thus inhibiting chromatin targeting and gene regulation by Brg1. It does so by binding to the helicase domain of Brg1, a domain that is crucial for tethering Brg1 to chromatinized DNA targets. Brg1 helicase has dual nucleic-acid-binding specificities: it is capable of binding lncRNA (Mhrt) and chromatinized—but not naked—DNA. This dual-binding feature of helicase enables a competitive inhibition mechanism by which Mhrt sequesters Brg1 from its genomic DNA targets to prevent chromatin remodelling. A Mhrt–Brg1 feedback circuit is thus crucial for heart function. Human MHRT also originates from MYH7 loci and is repressed in various types of myopathic hearts, suggesting a conserved lncRNA mechanism in human cardiomyopathy. Our studies identify a cardioprotective lncRNA, define a new targeting mechanism for ATP-dependent chromatin-remodelling factors, and establish a new paradigm for lncRNA–chromatin interaction.

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

Access options

Buy this article

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

Figure 1: Profile of the noncoding RNA Mhrt.
Figure 2: Mhrt inhibits cardiac hypertrophy and failure.
Figure 3: Mhrt complexes with Brg1 through the helicase domain.
Figure 4: Mhrt inhibits chromatin targeting and gene regulation by Brg1.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Data have been deposited in the Gene Expression Omnibus under accession number GSE49716.

Change history

  • 01 October 2014

    Minor changes were made to author names and affiliations.

References

  1. RIKEN Genome Exploration Research Group and Genome Science Group (Genome Network Project Core Group) and the FANTOM Consortium Antisense transcription in the mammalian transcriptome. Science 309, 1564–1566 (2005)

    Article  ADS  Google Scholar 

  2. Haddad, F., Bodell, P. W., Qin, A. X., Giger, J. M. & Baldwin, K. M. Role of antisense RNA in coordinating cardiac myosin heavy chain gene switching. J. Biol. Chem. 278, 37132–37138 (2003)

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hung, T. et al. Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nature Genet. 43, 621–629 (2011)

    Article  CAS  PubMed  Google Scholar 

  5. Lin, M. F., Jungreis, I. & Kellis, M. PhyloCSF: a comparative genomics method to distinguish protein coding and non-coding regions. Bioinformatics 27, i275–i282 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ingolia, N. T., Brar, G. A., Rouskin, S., McGeachy, A. M. & Weissman, J. S. The ribosome profiling strategy for monitoring translation in vivo by deep sequencing of ribosome-protected mRNA fragments. Nature Protocols 7, 1534–1550 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. van Rooij, E. et al. Control of stress-dependent cardiac growth and gene expression by a microRNA. Science 316, 575–579 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Miyata, S., Minobe, W., Bristow, M. R. & Leinwand, L. A. Myosin heavy chain isoform expression in the failing and nonfailing human heart. Circ. Res. 86, 386–390 (2000)

    Article  CAS  PubMed  Google Scholar 

  9. Lompre, A. M. et al. Myosin isoenzyme redistribution in chronic heart overload. Nature 282, 105–107 (1979)

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Schultz, J. J. et al. TGF-β1 mediates the hypertrophic cardiomyocyte growth induced by angiotensin II. J. Clin. Invest. 109, 787–796 (2002)

    Article  CAS  PubMed Central  Google Scholar 

  11. Molkentin, J. D. & Dorn, G. W., II Cytoplasmic signaling pathways that regulate cardiac hypertrophy. Annu. Rev. Physiol. 63, 391–426 (2001)

    Article  CAS  PubMed  Google Scholar 

  12. López, B. et al. Osteopontin-mediated myocardial fibrosis in heart failure: a role for lysyl oxidase? Cardiovasc. Res. 99, 111–120 (2013)

    Article  PubMed  CAS  Google Scholar 

  13. Frey, N. & Olson, E. N. Cardiac hypertrophy: the good, the bad, and the ugly. Annu. Rev. Physiol. 65, 45–79 (2003)

    Article  CAS  PubMed  Google Scholar 

  14. Guttman, M. et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458, 223–227 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rando, O. J. & Chang, H. Y. Genome-wide views of chromatin structure. Annu. Rev. Biochem. 78, 245–271 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hahn, M. A., Wu, X., Li, A. X., Hahn, T. & Pfeifer, G. P. Relationship between gene body DNA methylation and intragenic H3K9me3 and H3K36me3 chromatin marks. PLoS ONE 6, e18844 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Musselman, C. A. et al. Molecular basis for H3K36me3 recognition by the Tudor domain of PHF1. Nature Struct. Mol. Biol. 19, 1266–1272 (2012)

    Article  CAS  Google Scholar 

  19. Liu, R. et al. Regulation of CSF1 promoter by the SWI/SNF-like BAF complex. Cell 106, 309–318 (2001)

    Article  CAS  PubMed  Google Scholar 

  20. Muchardt, C. & Yaniv, M. A human homologue of Saccharomyces cerevisiae SNF2/SWI2 and Drosophila brm genes potentiates transcriptional activation by the glucocorticoid receptor. EMBO J. 12, 4279–4290 (1993)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Szabó, G. et al. Poly(ADP-ribose) polymerase inhibition reduces reperfusion injury after heart transplantation. Circ. Res. 90, 100–106 (2002)

    Article  PubMed  Google Scholar 

  22. Hesselberth, J. R. et al. Global mapping of protein–DNA interactions in vivo by digital genomic footprinting. Nature Methods 6, 283–289 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gupta, M. P. Factors controlling cardiac myosin-isoform shift during hypertrophy and heart failure. J. Mol. Cell. Cardiol. 43, 388–403 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Clapier, C. R. & Cairns, B. R. The biology of chromatin remodeling complexes. Annu. Rev. Biochem. 78, 273–304 (2009)

    Article  CAS  PubMed  Google Scholar 

  25. Jankowsky, E. & Fairman, M. E. RNA helicases—one fold for many functions. Curr. Opin. Struct. Biol. 17, 316–324 (2007)

    Article  CAS  PubMed  Google Scholar 

  26. Mallam, A. L., Del Campo, M., Gilman, B., Sidote, D. J. & Lambowitz, A. M. Structural basis for RNA-duplex recognition and unwinding by the DEAD-box helicase Mss116p. Nature 490, 121–125 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Dürr, H., Korner, C., Muller, M., Hickmann, V. & Hopfner, K. P. X-ray structures of the Sulfolobus solfataricus SWI2/SNF2 ATPase core and its complex with DNA. Cell 121, 363–373 (2005)

    Article  PubMed  CAS  Google Scholar 

  28. Feng, Y. et al. Histone H4 acetylation differentially modulates arginine methylation by an in cis mechanism. J. Biol. Chem. 286, 20323–20334 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zuker, M. On finding all suboptimal foldings of an RNA molecule. Science 244, 48–52 (1989)

    Article  ADS  MathSciNet  CAS  PubMed  MATH  Google Scholar 

  30. Wu, B. et al. Inducible cardiomyocyte-specific gene disruption directed by the rat Tnnt2 promoter in the mouse. Genesis 48, 63–72 (2010)

    PubMed  PubMed Central  Google Scholar 

  31. Wei, K., Kuhnert, F. & Kuo, C. J. Recombinant adenovirus as a methodology for exploration of physiologic functions of growth factor pathways. J. Mol. Med. (Berl.) 86, 161–169 (2008)

    Article  CAS  Google Scholar 

  32. Kuhnert, F. et al. Essential regulation of CNS angiogenesis by the orphan G protein-coupled receptor GPR124. Science 330, 985–989 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xiong, Y. et al. Brg1 governs a positive feedback circuit in the hair follicle for tissue regeneration and repair. Dev. Cell 25, 169–181 (2013)

    Article  CAS  PubMed  Google Scholar 

  34. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nature Methods 9, 357–359 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nature Protocols 7, 562–578 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Stankunas, K. et al. Endocardial Brg1 represses ADAMTS1 to maintain the microenvironment for myocardial morphogenesis. Dev. Cell 14, 298–311 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chang, C. P. et al. A field of myocardial-endocardial NFAT signaling underlies heart valve morphogenesis. Cell 118, 649–663 (2004)

    Article  CAS  PubMed  Google Scholar 

  40. Khavari, P. A., Peterson, C. L., Tamkun, J. W., Mendel, D. B. & Crabtree, G. R. BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature 366, 170–174 (1993)

    Article  ADS  CAS  PubMed  Google Scholar 

  41. 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 

  42. 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 

  43. van der Vlag, J., den Blaauwen, J. L., Sewalt, R. G., van Driel, R. & Otte, A. P. Transcriptional repression mediated by polycomb group proteins and other chromatin-associated repressors is selectively blocked by insulators. J. Biol. Chem. 275, 697–704 (2000)

    Article  CAS  PubMed  Google Scholar 

  44. Sengoku, T., Nureki, O., Nakamura, A., Kobayashi, S. & Yokoyama, S. Structural basis for RNA unwinding by the DEAD-box protein Drosophila Vasa. Cell 125, 287–300 (2006)

    Article  CAS  PubMed  Google Scholar 

  45. Thomä, N. H. et al. Structure of the SWI2/SNF2 chromatin-remodeling domain of eukaryotic Rad54. Nature Struct. Mol. Biol. 12, 350–356 (2005)

    Article  CAS  Google Scholar 

  46. Hauk, G., McKnight, J. N., Nodelman, I. M. & Bowman, G. D. The chromodomains of the Chd1 chromatin remodeler regulate DNA access to the ATPase motor. Mol. Cell 39, 711–723 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zuker, M. & Stiegler, P. Optimal computer folding of large RNA sequences using thermodynamics and auxiliary information. Nucleic Acids Res. 9, 133–148 (1981)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gruber, A. R., Lorenz, R., Bernhart, S. H., Neubock, R. & Hofacker, I. L. The Vienna RNA websuite. Nucleic Acids Res. 36, W70–W74 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wan, Y., Kertesz, M., Spitale, R. C., Segal, E. & Chang, H. Y. Understanding the transcriptome through RNA structure. Nature Rev. Genet. 12, 641–655 (2011)

    Article  CAS  PubMed  Google Scholar 

  50. Fu, X. M., Yao, Y. J., Yang, Z., Xiang, L. & Gao, J. [Alteration and its significance to expression of aquaporin-4 in cultured neonatal rat astrocytes in the model of hypoxic damage.] Sichuan Da Xue Xue Bao Yi Xue Ban. 36, 641–644 (2005)

    CAS  PubMed  Google Scholar 

  51. Yang, J. et al. C-reactive protein augments hypoxia-induced apoptosis through mitochondrion-dependent pathway in cardiac myocytes. Mol. Cell. Biochem. 310, 215–226 (2008)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank C.-H. Chen for assisting with echocardiography; L. Chen, A. Kuo and G. Crabtree for transgene injection and northern blot; M. Ecarkt and E. Zuo for ribosome analysis. C.-P.C. was supported by the American Heart Association (AHA; Established Investigator Award 12EIA8960018), National Institutes of Health (NIH; HL118087, HL121197), March of Dimes Foundation (#6-FY11-260), California Institute of Regenerative Medicine (CIRM; RN2-00909), Oak Foundation, Baxter Foundation, Stanford Heart Center Research Program, Indiana University (IU) School of Medicine—IU Health Strategic Research Initiative, and the IU Physician-Scientist Initiative, endowed by Lilly Endowment. W.L. and Y.X. were supported by the Oak Foundation; Y.X. by the AHA and Lucile Packard Children’s Foundation; C.S. by an NIH fellowship; T.Q. by NIH (HL109512); H.-S.V.C. by CIRM (RB2-01512, RB4-06276) and NIH (HL105194); P.-S.C. by NIH (HL78931, HL71140); B.Z. by NIH (HL116997, HL111770).

Author information

Authors and Affiliations

Authors

Contributions

C.-P.C. and P.H. were responsible for the original concepts, design and manuscript preparation. W.L. and C.-H.L. contributed equally to the work. P.H. conducted most experiments; W.L. and J.Y. assisted with TAC, echo and reporter analyses; C.-H.L. assisted with protein purification; S.T.N. assisted with ribosome data analysis; K.K.J. assisted with protein sequence and motif analysis; C.S. assisted with western blot studies; W.X. assisted with CSF scoring; Y.X. assisted with RNA/protein staining; C.-J.L. and C.-Y.L. assisted with Brg1-null tissue preparation and H-10 antibody-ChIP optimization; H.-C.C. assisted with cloning; H.-S.V.C. assisted with tissue collection; E.A. assisted with tissue collection/rat tissue supply; B.Z. assisted with driver line generation; D.B., P.-S.C. and T.Q. assisted with data analysis.

Corresponding author

Correspondence to Ching-Pin Chang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Mhrt RNAs have no coding potential.

a, RNA in situ analysis of Mhrt (blue) in a mouse E12 heart. The RNA probe targets all Mhrt species. Red: nuclear fast red. Black arrowheads indicate nuclei of endothelial, endocardial or epicardial cells. Inset shows magnified region from the boxed area. endo, endocardium; epi, epicardium; IVS, interventricular septum; LV, left ventricle; RA and RV, right atrium and ventricle, respectively. Scale bars = 100 μm. b, Codon substitution frequency (CSF) scores of TfIIb and Hprt1 mRNA, as well as full-length Mhrt species. c, In vitro translation of control Mhrt species (709, 779, 826, 828, 857, 1147) and luciferase (Luc). Arrow points to the protein product of luciferase. d, Biotin-labelling of Mhrt species (709, 779, 826, 828, 857, 1147) and luciferase in the in vitro translation reactions. Arrow points to the RNA product of luciferase. e, Ribosome profiling relative to whole transcriptome RNA sequencing. x-axis: genomic position at the human GAPDH and the murine Myh7 loci. y-axis: mapped reads. f, Scatter plot of RNA in fragments per kilobase per million reads (FPKM). Noncoding RNAs (purple) cluster towards the x-axis; coding RNAs (orange) towards the y-axis. Mhrt779 is found below both the identity line (dashed, slope = 1, intercept = 0) and the smooth-fit regression line (in blue). RNA examples are endogenous except that HOTAIR was co-transfected with Mhrt779.

Extended Data Figure 2 Quantification of Myh6/Myh7, northern blot, and Mhrt779 characterization.

a, Quantification of cardiac Myh6/Myh7 ratio 2–42 days after sham or TAC operation. b, Northern blot analysis of Mhrt, Myh6 and Myh7. Negative: control RNA from 293T cells. Size control: 826 is recombinant Mhrt826; 643 (not a distinct Mhrt species) contains the 5′ common region of Mhrt. Heart: adult heart ventricles. c, Un-cropped northern blots of Mhrt, Myh6 and Myh7. d, RNA in situ hybridization of Mhrt779 of adult heart ventricles. White arrowheads indicate nuclei of myocardial cells. Black arrowheads indicate nuclei of endothelial, endocardial or epicardial cells. Blue: Mhrt779; Red: nuclear fast red. Epi, epicardium. The dashed line separates the epicardium from myocardium. Scale bars = 50 μm. e, Quantification of TfIIb, Hprt1, 28S rRNA, Neat1 and Mhrt779 in the nuclear and cytoplasmic fraction of adult heart ventricle extracts. The nuclear/cytoplasmic ratio of TfIIb is set as 1. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 3 Wheat germ agglutinin staining, time course and molecular marker studies of the stressed Tg779 mice.

a, Wheat germ agglutinin (WGA) immunostaining 6 weeks after the sham or TAC operation. Green: WGA stain, outlining cell borders of cardiomyocytes. Blue: 4′,6-diamidino-2-phenylindole (DAPI). Ctrl, control mice. Scale bars = 50 μm. b, Time course of fractional shortening (FS) in control and Tg779 mice. c, Quantification of Anf, Bnp, Serca2 and Tgfb1 in control and Tg779 mice 2 weeks after sham or TAC operation. d, Experimental design for treatment study and time course of left ventricular fractional shortening changes. e, Fractional shortening of the left ventricle (LV) 8 weeks after the operation. f, Ventricular weight/body weight ratio of hearts harvested 8 weeks after sham or TAC operation. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 4 Regulation of the Mhrt promoter.

a, Sequence alignment of Mhrt promoter loci from mouse, human and rat. Peak heights indicate degree of sequence homology. Black boxes (a1–a4) are sequences of high homology, which were used for further ChIP analysis. Green box region between Myh6 and Mhrt is the putative Mhrt promoter. Red, promoter regions; salmon, introns; yellow, untranslated regions. bd, ChIP–qPCR analysis of Mhrt promoter using antibodies against Pol II (b), H3K4me3 (c), and H3K36me3 (d) in tissues of adult mice. e, RT–qPCR quantification of Mhrt in control and Brg1-null hearts after 7 days of TAC. Ctrl, control. Brg1-null, Tnnt2-rtTA;Tre-Cre;Brg1fl/fl. f, Luciferase reporter assay of Mhrt promoter in SW13 cells. Ctrl: dimethylsulphoxide (DMSO). PJ-34, PARP inhibitor; TSA, trichostatin (HDAC inhibitor). g, ChIP analysis of BRG1, HDAC2, HDAC9 and PARP1 in SW13 cells. The cells were transfected with episomal Mhrt promoter cloned in pREP4. h, Deletional analyses of the Mhrt promoter in luciferase reporter assays in SW13 cells. Luciferase activity of full-length Mhrt promoter was set up as 1. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 5 Mhrt does not affect Myh expression by direct RNA sequence interference.

a, qPCR analysis of Mhrt779, Myh6 and Myh7 in mice without TAC operation. Expression levels were normalized to TfIIb, and the control is set as 1. Ctrl, control mice. b, c, RNA quantification of Mhrt (b) and HOTAIR (c) in SW13 cells transfected with Vector (pAdd2), HOTAIR (pAdd2-HOTAIR) or Mhrt (pAdd2-Mhrt779). Expression in vector-transfected cells is set as 1. Constructs containing Myh6 or Myh7 were co-transfected into SW13 cells used for Fig. 2b–i. d, e, RNA quantification of Myh6 (d) and Myh7 (e) in SW13 cells relative to GAPDH. f, g, Western blot analysis of Myh6 (f) and Myh7 (g) in SW13 cells. Constructs containing Myh6- and Myh7-coding sequences were tagged with Flag and co-transfected with vector, HOTAIR or Mhrt779. GAPDH was used as the loading control. Flag–D1 was used as a positive control for the Flag antibody. h, i, Protein quantification of Myh6 (h) and Myh7 (i) in control and transfected SW13 cells relative to GAPDH. Signals of Myh6 and Myh7 from major bands or the entire lanes were quantified.WB, western blot. j, Luciferase reporter assay of Mhy6 and Myh7 promoters in SW13 cells transfected with vector (pAdd2) or Mhrt (pAdd2-Mhrt779). P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 6 RNA-IP controls; Opn is another target gene of Brg1 in stressed hearts.

a, Immunostaining of Brg1 in P1 heart. Red: Brg1. Green: WGA. Blue: DAPI. Ctrl, control. Scale bar = 50 μm. b, RNA-IP of Mhrt in P1 hearts using antibodies against Ezh2 and Suz12. Right panels show immunostaining of Ezh2 and Suz12 in P1 hearts. PRC2, polycomb repressor complex 2. Red: Ezh2 or Suz12. Green: WGA. Blue: DAPI. Scale bars = 50 μm. c, Quantification of Opn mRNA in control and Brg1-null (Tnnt2-rtTA;Tre-Cre;Brg1fl/fl) mice after sham or TAC operation. d, ChIP of Brg1 on Opn proximal promoter in control and transgenic (Tg779) mice after sham or TAC operation. e, Quantification of Opn in control and transgenic (Tg779) mice after sham or TAC operation. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 7 Induction of Mhrt779 is insufficient to change Brg1 mRNA or protein level.

a, qPCR analysis of Brg1 expression in hearts without TAC operation. Ctrl: control mice. be, Immunostaining of Brg1 (red) in adult heart ventricles 2 weeks after sham or TAC operation. Green: WGA. Blue: DAPI. Scale bars = 50 μm. f, Western blot analysis of Brg1 and Coomassie staining of total proteins in control or Tg779 hearts after 2 weeks of sham or TAC operation. g, Quantification of Myh6 and Myh7 in control (Ctrl) and Tg779 hearts after 2 weeks of sham or TAC operation. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 8 Brg1 sequence alignment and motif analysis.

Schematics of the architecture of mouse Brg1 and the sequence alignment of Brg1, Vasa (fruit fly), Rad54 (zebrafish, Sulfolobus solfataricus) and Chd1 (yeast). The motifs were outlined by blue boxes (D1 domain) and purple boxes (D2 domain).

Extended Data Figure 9 Purification of Brg1 helicase core domains, EMSA of naked Myh6 promoter, ChIP and reporter studies in SW13 cells.

a, Coomassie blue staining of purified MBP-tagged Brg1 helicase domains. Bovine serum albumin (BSA) was loaded as a control. b, EMSA assay of naked Myh6 promoter (−426 to +170) with helicase domains of Brg1. Probe: biotin-labelled Myh6 promoter. 50 μM of MBP, MBP–D1, MBP–D2 and MBP–D1D2 proteins were used for EMSA. c, d, ChIP (c) and luciferase reporter (d) analysis of Brg1 on chromatinized (episomal) and naked Myh6 promoter in SW13 cells. GFP, green fluorescent protein control. e, The luciferase reporter of helicase-deficient Brg1 on chromatinized (episomal) Myh6 promoter in SW13 cells. ΔD1: Brg1 lacking amino acids 774–913. ΔD2: Brg1 lacking amino acids 1086–1246. ChIP: H-10 antibody recognizing N terminus, non-disrupted region of Brg1. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 10 Brg1 outruns Mhrt to bind to its target Mhrt promoter.

a, Assembly of nucleosomes on the Mhrt promoter (a3/4). b, Amylose pull-down assay: amylose was used to pull down the chromatinized Mhrt promoter that was incubated with various doses of MBP and MBP–Brg1 D1D2. DNA precipitated by amylose was further quantified by qPCR. P values: Student’s t-test. Error bars show s.e.m.

Extended Data Figure 11 Sequence alignment and secondary structure prediction of human and mouse MHRT, and demography of heart transplantation donors.

a, Sequence alignment of human MHRT and mouse Mhrt779. b, Predicted secondary structure of mouse Mhrt779 and human MHRT, using minimal free energy (MFE) calculation of RNAfold WebServer. c, Demography of human subjects whose tissues were used for RT–qPCR analysis (Fig. 4l). ICM, ischaemic cardiomyopathy; IDCM, idiopathic cardiomyopathy; LVH, left ventricular hypertrophy.

Supplementary information

Supplementary Information

This file contains Supplementary Notes, Sequences of alternative spliced Mhrts, Sequences of human MHRT and additional references. (PDF 225 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Han, P., Li, W., Lin, CH. et al. A long noncoding RNA protects the heart from pathological hypertrophy. Nature 514, 102–106 (2014). https://doi.org/10.1038/nature13596

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13596

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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