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.

  • Article
  • Published:

ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation

Abstract

The murine neonatal heart can regenerate after injury through cardiomyocyte (CM) proliferation, although this capacity markedly diminishes after the first week of life. Neuregulin-1 (NRG1) administration has been proposed as a strategy to promote cardiac regeneration. Here, using loss- and gain-of-function genetic tools, we explore the role of the NRG1 co-receptor ERBB2 in cardiac regeneration. NRG1-induced CM proliferation diminished one week after birth owing to a reduction in ERBB2 expression. CM-specific Erbb2 knockout revealed that ERBB2 is required for CM proliferation at embryonic/neonatal stages. Induction of a constitutively active ERBB2 (caERBB2) in neonatal, juvenile and adult CMs resulted in cardiomegaly, characterized by extensive CM hypertrophy, dedifferentiation and proliferation, differentially mediated by ERK, AKT and GSK3β/β-catenin signalling pathways. Transient induction of caERBB2 following myocardial infarction triggered CM dedifferentiation and proliferation followed by redifferentiation and regeneration. Thus, ERBB2 is both necessary for CM proliferation and sufficient to reactivate postnatal CM proliferative and regenerative potentials.

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: NRG1-induced CM cell-cycle progression is diminished during the first week of postnatal life, in correlation with reduced ERBB2 levels.
Figure 2: ERBB2 is required for NRG1-induced CM proliferation.
Figure 3: Postnatal activation of ERBB2 signalling leads to cardiomegaly.
Figure 4: Postnatal activation of ERBB2 signalling promotes extensive CM proliferation and hypertrophy in vitro and in vivo.
Figure 5: Postnatal activation of ERBB2 signalling promotes CM dedifferentiation.
Figure 6: ERK, AKT and GSK3β/β-catenin differentially mediate ERBB2-induced CM proliferation, dedifferentiation and hypertrophy.
Figure 7: Transient ERBB2 induction triggers CM dedifferentiation, proliferation and cardiac regeneration following injury in juvenile and adult stages.
Figure 8: ERBB2 signalling triggers robust CM dedifferentiation and proliferation and reactivates a dormant postnatal regenerative window in mice.

Similar content being viewed by others

References

  1. Bergmann, O. et al. Evidence for cardiomyocyte renewal in humans. Science 324, 98–102 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Senyo, S. E. et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature 493, 433–436 (2013).

    CAS  PubMed  Google Scholar 

  3. Laflamme, M. A. & Murry, C. E. Regenerating the heart. Nat. Biotechnol. 23, 845–856 (2005).

    CAS  PubMed  Google Scholar 

  4. Poss, K. D. Getting to the heart of regeneration in zebrafish. Semin. Cell Dev. Biol. 18, 36–45 (2007).

    CAS  PubMed  Google Scholar 

  5. Ausoni, S. & Sartore, S. From fish to amphibians to mammals: in search of novel strategies to optimize cardiac regeneration. J. Cell Biol. 184, 357–364 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Mercola, M., Ruiz-Lozano, P. & Schneider, M. D. Cardiac muscle regeneration: lessons from development. Genes Dev. 25, 299–309 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Garbern, J. C. & Lee, R. T. Cardiac stem cell therapy and the promise of heart regeneration. Cell Stem Cell 12, 689–698 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Li, F., Wang, X., Capasso, J. M. & Gerdes, A. M. Rapid transition of cardiac myocytes from hyperplasia to hypertrophy during postnatal development. J. Mol. Cell. Cardiol. 28, 1737–1746 (1996).

    CAS  PubMed  Google Scholar 

  9. Soonpaa, M. H. & Field, L. J. Survey of studies examining mammalian cardiomyocyte DNA synthesis. Circ. Res. 83, 15–26 (1998).

    CAS  PubMed  Google Scholar 

  10. Naqvi, N. et al. A proliferative burst during preadolescence establishes the final cardiomyocyte number. Cell 157, 795–807 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Jopling, C. et al. Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation. Nature 464, 606–609 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Porrello, E. R. et al. Transient regenerative potential of the neonatal mouse heart. Science 331, 1078–1080 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. van Berlo, J. H. & Molkentin, J. D. An emerging consensus on cardiac regeneration. Nat. Med. 20, 1386–1393 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Xin, M., Olson, E. N. & Bassel-Duby, R. Mending broken hearts: cardiac development as a basis for adult heart regeneration and repair. Nat. Rev. Mol. Cell Biol. 14, 529–541 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gassmann, M. et al. Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor. Nature 378, 390–394 (1995).

    CAS  PubMed  Google Scholar 

  16. Lee, K. F. et al. Requirement for neuregulin receptor erbB2 in neural and cardiac development. Nature 378, 394–398 (1995).

    CAS  PubMed  Google Scholar 

  17. Meyer, D. & Birchmeier, C. Multiple essential functions of neuregulin in development. Nature 378, 386–390 (1995).

    CAS  PubMed  Google Scholar 

  18. Lai, D. et al. Neuregulin 1 sustains the gene regulatory network in both trabecular and nontrabecular myocardium. Circ. Res. 107, 715–727 (2010).

    CAS  PubMed  Google Scholar 

  19. Liu, J. et al. A dual role for ErbB2 signaling in cardiac trabeculation. Development 137, 3867–3875 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Staudt, D. W. et al. High-resolution imaging of cardiomyocyte behavior reveals two distinct steps in ventricular trabeculation. Development 141, 585–593 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhao, Y. Y. et al. Neuregulins promote survival and growth of cardiac myocytes. Persistence of ErbB2 and ErbB4 expression in neonatal and adult ventricular myocytes. J. Biol. Chem. 273, 10261–10269 (1998).

    CAS  PubMed  Google Scholar 

  22. Odiete, O., Hill, M. F. & Sawyer, D. B. Neuregulin in cardiovascular development and disease. Circ. Res. 111, 1376–1385 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Wadugu, B. & Kuhn, B. The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. Am. J. Physiol. Heart Circ. Physiol. 302, H2139–H2147 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Fukazawa, R. et al. Neuregulin-1 protects ventricular myocytes from anthracycline-induced apoptosis via erbB4-dependent activation of PI3-kinase/Akt. J. Mol. Cell. Cardiol. 35, 1473–1479 (2003).

    CAS  PubMed  Google Scholar 

  25. Liu, X. et al. Neuregulin-1/erbB-activation improves cardiac function and survival in models of ischemic, dilated, and viral cardiomyopathy. J. Am. Coll. Cardiol. 48, 1438–1447 (2006).

    CAS  PubMed  Google Scholar 

  26. Bersell, K., Arab, S., Haring, B. & Kuhn, B. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell 138, 257–270 (2009).

    CAS  PubMed  Google Scholar 

  27. Xu, Y., Li, X., Liu, X. & Zhou, M. Neuregulin-1/ErbB signaling and chronic heart failure. Adv. Pharmacol. 59, 31–51 (2010).

    CAS  PubMed  Google Scholar 

  28. Pentassuglia, L. & Sawyer, D. B. The role of Neuregulin-1β/ErbB signaling in the heart. Exp. Cell Res. 315, 627–637 (2009).

    CAS  PubMed  Google Scholar 

  29. Gao, R. et al. A Phase II, randomized, double-blind, multicenter, based on standard therapy, placebo-controlled study of the efficacy and safety of recombinant human neuregulin-1 in patients with chronic heart failure. J. Am. Coll. Cardiol. 55, 1907–1914 (2010).

    CAS  PubMed  Google Scholar 

  30. Jabbour, A. et al. Parenteral administration of recombinant human neuregulin-1 to patients with stable chronic heart failure produces favourable acute and chronic haemodynamic responses. Eur. J. Heart Fail. 13, 83–92 (2011).

    CAS  PubMed  Google Scholar 

  31. Sawyer, D. B. & Caggiano, A. Neuregulin-1beta for the treatment of systolic heart failure. J. Mol. Cell. Cardiol. 51, 501–505 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Reuter, S., Soonpaa, M. H., Firulli, A. B., Chang, A. N. & Field, L. J. Recombinant neuregulin 1 does not activate cardiomyocyte DNA synthesis in normal or infarcted adult mice. PLoS ONE 9, e115871 (2014).

    PubMed  PubMed Central  Google Scholar 

  33. Yarden, Y. & Sliwkowski, M. X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2, 127–137 (2001).

    CAS  PubMed  Google Scholar 

  34. Citri, A. & Yarden, Y. EGF-ERBB signalling: towards the systems level. Nat. Rev. Mol. Cell Biol. 7, 505–516 (2006).

    CAS  PubMed  Google Scholar 

  35. Pradeep, C. R. et al. Modeling invasive breast cancer: growth factors propel progression of HER2-positive premalignant lesions. Oncogene 31, 3569–3583 (2012).

    CAS  PubMed  Google Scholar 

  36. Tzahar, E. et al. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol. Cell Biol. 16, 5276–5287 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Graus-Porta, D., Beerli, R. R., Daly, J. M. & Hynes, N. E. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 16, 1647–1655 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Olayioye, M. A. et al. ErbB-1 and ErbB-2 acquire distinct signaling properties dependent upon their dimerization partner. Mol. Cell Biol. 18, 5042–5051 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Garrett, T. P. et al. The crystal structure of a truncated ErbB2 ectodomain reveals an active conformation, poised to interact with other ErbB receptors. Mol. Cell 11, 495–505 (2003).

    CAS  PubMed  Google Scholar 

  40. Agah, R. et al. Gene recombination in postmitotic cells. Targeted expression of Cre recombinase provokes cardiac-restricted, site-specific rearrangement in adult ventricular muscle in vivo. J. Clin. Invest. 100, 169–179 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ozcelik, C. et al. Conditional mutation of the ErbB2 (HER2) receptor in cardiomyocytes leads to dilated cardiomyopathy. Proc. Natl Acad. Sci. USA 99, 8880–8885 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Britsch, S. et al. The ErbB2 and ErbB3 receptors and their ligand, neuregulin-1, are essential for development of the sympathetic nervous system. Genes Dev. 12, 1825–1836 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bargmann, C. I., Hung, M. C. & Weinberg, R. A. Multiple independent activations of the neu oncogene by a point mutation altering the transmembrane domain of p185. Cell 45, 649–657 (1986).

    CAS  PubMed  Google Scholar 

  44. Penuel, E., Akita, R. W. & Sliwkowski, M. X. Identification of a region within the ErbB2/HER2 intracellular domain that is necessary for ligand-independent association. J. Biol. Chem. 277, 28468–28473 (2002).

    CAS  PubMed  Google Scholar 

  45. Ghashghaei, H. T. et al. Reinduction of ErbB2 in astrocytes promotes radial glial progenitor identity in adult cerebral cortex. Genes Dev. 21, 3258–3271 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Sysa-Shah, P. et al. Cardiac-specific over-expression of epidermal growth factor receptor 2 (ErbB2) induces pro-survival pathways and hypertrophic cardiomyopathy in mice. PLoS ONE 7, e42805 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Hattori, F. et al. Nongenetic method for purifying stem cell-derived cardiomyocytes. Nat. Methods 7, 61–66 (2010).

    CAS  PubMed  Google Scholar 

  48. Kubin, T. et al. Oncostatin M is a major mediator of cardiomyocyte dedifferentiation and remodeling. Cell Stem Cell 9, 420–432 (2011).

    CAS  PubMed  Google Scholar 

  49. Tseng, A. S., Engel, F. B. & Keating, M. T. The GSK-3 inhibitor BIO promotes proliferation in mammalian cardiomyocytes. Chem. Biol. 13, 957–963 (2006).

    CAS  PubMed  Google Scholar 

  50. Uosaki, H. et al. Identification of chemicals inducing cardiomyocyte proliferation in developmental stage-specific manner with pluripotent stem cells. Circ. Cardiovasc. Genet. 6, 624–633 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Schade, B. et al. beta-Catenin signaling is a critical event in ErbB2-mediated mammary tumor progression. Cancer Res. 73, 4474–4487 (2013).

    CAS  PubMed  Google Scholar 

  52. Reya, T. & Clevers, H. Wnt signalling in stem cells and cancer. Nature 434, 843–850 (2005).

    CAS  PubMed  Google Scholar 

  53. Plotnikov, A. et al. Oncogene-mediated inhibition of glycogen synthase kinase 3 beta impairs degradation of prolactin receptor. Cancer Res. 68, 1354–1361 (2008).

    CAS  PubMed  Google Scholar 

  54. Zaoui, K., Benseddik, K., Daou, P., Salaun, D. & Badache, A. ErbB2 receptor controls microtubule capture by recruiting ACF7 to the plasma membrane of migrating cells. Proc. Natl Acad. Sci. USA 107, 18517–18522 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Engel, F. B. et al. p38 MAP kinase inhibition enables proliferation of adult mammalian cardiomyocytes. Genes Dev. 19, 1175–1187 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Engel, F. B., Hsieh, P. C., Lee, R. T. & Keating, M. T. FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction. Proc. Natl Acad. Sci. USA 103, 15546–15551 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Kuhn, B. et al. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat. Med. 13, 962–969 (2007).

    PubMed  Google Scholar 

  58. Whelan, R. S., Kaplinskiy, V. & Kitsis, R. N. Cell death in the pathogenesis of heart disease: mechanisms and significance. Annu. Rev. Physiol. 72, 19–44 (2010).

    CAS  PubMed  Google Scholar 

  59. Krijnen, P. A. et al. Apoptosis in myocardial ischaemia and infarction. J. Clin. Pathol. 55, 801–811 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Mahmoud, A. I. et al. Meis1 regulates postnatal cardiomyocyte cell cycle arrest. Nature 497, 249–253 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Puente, B. N. et al. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell 157, 565–579 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Aurora, A. B. et al. Macrophages are required for neonatal heart regeneration. J. Clin. Invest. 124, 1382–1392 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Heallen, T. et al. Hippo signaling impedes adult heart regeneration. Development 140, 4683–4690 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    CAS  PubMed  Google Scholar 

  65. Xie, W., Chow, L. T., Paterson, A. J., Chin, E. & Kudlow, J. E. Conditional expression of the ErbB2 oncogene elicits reversible hyperplasia in stratified epithelia and up-regulation of TGFalpha expression in transgenic mice. Oncogene 18, 3593–3607 (1999).

    CAS  PubMed  Google Scholar 

  66. Yu, Z., Redfern, C. S. & Fishman, G. I. Conditional transgene expression in the heart. Circ. Res. 79, 691–697 (1996).

    CAS  PubMed  Google Scholar 

  67. Mahmoud, A. I., Porrello, E. R., Kimura, W., Olson, E. N. & Sadek, H. A. Surgical models for cardiac regeneration in neonatal mice. Nat. Protoc. 9, 305–311 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Chaudhry, H. W. et al. Cyclin A2 mediates cardiomyocyte mitosis in the postmitotic myocardium. J. Biol. Chem. 279, 35858–35866 (2004).

    CAS  PubMed  Google Scholar 

  69. Jackson, T. et al. The c-myc proto-oncogene regulates cardiac development in transgenic mice. Mol. Cell Biol. 10, 3709–3716 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Vandoorne, K. et al. Multimodal imaging reveals a role for Akt1 in fetal cardiac development. Physiol. Rep. 1, e00143 (2013).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants to E.T. from the European Research Council, Israel Science Foundation, the Louis and Fannie Tolz Collaborative Research Project, Estate of Jack Gitlitz and to R.P.H. from the National Health and Medical Research Council (NHMRC) of Australia (573705; 573732) and the Australian Research Council Special Initiative in Stem Cell Science (Stem Cells Australia). We thank C. Birchmeier (MDC, Germany) for providing the Erbb2flox and Erbb2LacZ mice, I. Biton for helping with the MRI measurements, and D. Sawyer (Vanderbilt University, USA), R. Graham (Victor Chang Cardiac Research Institute, Australia), A. Aronheim (Technion, Israel) and K. Yaniv (Weizmann Institute of Science, Israel) for fruitful discussions. We also thank H. Sadek and E. Olson (UT Southwestern Medical Center, USA) for teaching us the neonatal cardiac regenerative model and L. Field (Indiana University School of Medicine, USA) for sharing unpublished data.

Author information

Authors and Affiliations

Authors

Contributions

G.D’U. and E.T. conceived and designed the experiments. G.D’U. with help from A.A. carried out most of the experiments and analysed the data. M. Lauriola and S.C. performed some western blots and helped with animal studies. D.K. performed myocardial infarction experiment in adult mice. Y.Y-R. performed time-lapse imaging. K.W. and T.K. assisted with myocardial infarction experiment in juvenile mice. E.B., D.R. and O.Y. assisted with gene expression analysis. M. Lysenko performed MRI analysis. O.B. helped with interpretation of histological slides. J.H. helped with the interpretation of echocardiographic analysis. R.S. contributed to the planning and progression of the project. J.L., Y.Y. and M.N. supervised the experiments done by their laboratory members, and E.T. supervised the entire project. R.P.H. provided conceptual inputs and G.D’U., R.P.H. and E.T. wrote the manuscript with editing contributions from all of the authors.

Corresponding author

Correspondence to Eldad Tzahor.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 4 Erbb2-cKO mice display enlarged heart with compensatory hypertrophy.

(a) Western Blot analysis of ERBB2 protein levels in P0 ctrl and Erbb2-cKO heart lysates; (b) Pictures of P0 ctrl and Erbb2-cKO hearts obtained by binocular microscope (LV, left ventricle; RV, right ventricle, LA, left atrium, RA, right atrium); scale bar, 1 mm; (c) Heart weight to body weight ratio in P0 ctrl and Erbb2-cKO mice (n = 22 mice); (d) Picture of P0 ctrl and Erbb2-cKO mice, displaying a pale skin colour before death; scale bar 1 cm; (e) Body weight of P0 ctrl and Erbb2-cKO mice (n = 51 mice); (f) CM size (area) quantification based on immunofluorescence analysis of cardiac Troponin T (cTnT) of P0 ctrl and Erbb2-cKO cells cultured in vitro for 5 days (n = 836 CMs pooled from the analysis of 6 mice); (gh) RT-PCR analysis of hypertrophic markers Nppa, Nppb, Acta1 and β/α-MHC in P0 ctrl and Erbb2-cKO heart lysates (n = 8 mice in g; n = 15 in h). In all panels numerical data are presented as mean + s.e.m.; statistical significance was calculated using two-tailed unpaired Student’s t-test in c and eh; results are marked with one asterisk () if P < 0.05, two () if P < 0.01, three () if P < 0.001 and four () if P < 0.0001; statistics source data can be found in Supplementary Table 1.

Supplementary Figure 5 Generation and analysis of CM restricted overexpression of a constitutively active Erbb2.

(a) Schematic diagram of the cross-breeding to generate CM restricted overexpression of a constitutively active Erbb2 (caErbb2); (b) Kinetics of ERBB2 protein levels in ctrl and neonatal caErbb2 heart lysates following DOX withdrawal according to scheme in Fig. 3a; (ce) Pictures of hearts isolated from ctrl and caErbb2 mice; images were obtained by binocular microscope; Scale bar 1 mm; (fh) Echocardiographic measurements of cardiac ejection fraction (EF) and fractional shortening (FS) in ctrl and caErbb2 mice (n = 14 mice in f; n = 14 mice in g; n = 20 mice in h). In all panels neonatal, juvenile and adult caErbb2 mice were generated according to the schema in Fig. 3a, b. Numerical data are presented as mean + s.e.m.; statistical significance was calculated using two-tailed unpaired Student’s t-test in fh; results are marked with one asterisk () if P < 0.05, two () if P < 0.01, three () if P < 0.001 and four () if P < 0.0001; statistics source data can be found in Supplementary Table 1.

Supplementary Figure 6 in vivo induction of ERBB2 signalling promotes CM dedifferentiation, proliferation and hypertrophy.

(a) CM cross-sectional area evaluation by WGA immunofluorescence analysis of P56 ctrl and adult caErbb2 left ventricular (LV) heart sections (n = 306 CMs pooled from the analysis of 6 mice); representative pictures are provided; Scale bar 10 μm; (b) Calculation of CM number in P56 ctrl and adult caErbb2 hearts (n = 6 mice); (cd) Hematoxilin and Eosin (H&E) histological analysis of LV heart sections in in ctrl and caErbb2 mice; Scale bar 25 μm; (ef) in vivo CM sarcomeric status evaluation by immunofluorescence analysis of cardiac Troponin T (cTnT) in ctrl and caErbb2 LV heart sections; Images were obtained using a spinning-disk confocal microscope; Scale bar 5 μm 0(g) WB analysis of cKIT protein levels in P7 ctrl and neonatal caErbb2 heart lysates. In all panels neonatal, juvenile and adult caErbb2 mice were generated according to the schema in Fig. 3a, b. Numerical data are presented as mean + s.e.m.; statistical significance was calculated using two-tailed unpaired Student’s t-test in a,b; results are marked with one asterisk () if P < 0.05, two () if P < 0.01, three () if P < 0.001 and four () if P < 0.0001; statistics source data can be found in Supplementary Table 1.

Supplementary Figure 7 Analysis of mediators and modulators of caERBB2-induced CM dedifferentiation, proliferation and hypertrophy.

(a) CM sarcomeric status evaluation based on cardiac Troponin T (cTnT) immunofluorescence analysis in P7 ctrl and neonatal caErbb2 heart cells treated in vitro for 2 days with pharmacological inhibitors of ERK and AKT (ERK-i and AKT-i); sarcomeric status was scored according to the criteria presented in Fig. 5c (n = 466 CMs pooled from the analysis of 24 samples); (b) in vivo immunofluorescence analysis of β-Catenin and sarcomeric Troponin T (cTnT) in LV heart sections of P7 ctrl and neonatal caErbb2 mice; images were obtained using a spinning-disk confocal microscope; Scale bar 10 μm (c) in vitro immunofluorescence analysis of β-Catenin and Troponin T (cTnT) in P7 neonatal caErbb2 heart cells treated with pharmacological inhibitors of GSK3β or β-Catenin (GSK3β-i and β-Catenin-i); Scale bar 60 μm; (df) P7 ctrl and neonatal caErbb2 heart cells were treated with p38 inhibitor (p38-i), FGF1 or Periostin in vitro for 2 days; CM were identified by Troponin T (cTnT) staining and analysed for (d) cell-cycle re-entry (Ki67), (e) dedifferentiation marker RUNX1 and (f) size by immunofluorescence analysis (n = 6013 CMs pooled from the analysis of 48 samples in d; n = 4217 CMs pooled from the analysis of 43 samples in e; n = 484 CMs pooled from the analysis of 17 samples in f). In all panels numerical data are presented as mean + s.e.m.; statistical significance was calculated using one-way ANOVA followed by Tukey’s test in a and df; results are marked with one asterisk () if P < 0.05, two () if P < 0.01, three () if P < 0.001 and four () if P < 0.0001; statistics source data can be found in Supplementary Table 1.

Supplementary Figure 8 Transient caErbb2 induces transient CM dedifferentiation and neovascularization following heart injury.

(a) Hematoxylin and eosin (H&E) histological analysis of remote zone of LV heart sections of ctrl and adult transient caErbb2 mice 1-month-post-MI (P70) and 2-months-post-MI (P98, one-month after caErbb2 signal termination) following myocardial infarction at P42 according to the schema in Fig. 7b; Scale bar 50 μm; (b) Angiogenesis evaluation by immunofluorescence analysis of CD34 in heart sections of 3-weeks-post-MI (P63) ctrl and adult caErbb2 following myocardial infarction at P42 according to the scheme in Fig. 7b; heart sections were co-stained with antibodies to cardiac Troponin T (cTnT) for identification of CMs (n = 50 fields pooled form the analysis of 6 mice); numerical data are presented as mean + s.e.m.; statistical significance was calculated using two-tailed unpaired Student’s t-test; results are marked with one asterisk () if P < 0.05; Representative pictures are provided; Scale bar 30 μm. statistics source data can be found in Supplementary Table 1.

Supplementary Figure 9 Uncropped images of immunoblots displayed in Figs 1g, 6a, 6e

Supplementary Table 1 Statistic data source.
Supplementary Table 2 List of primers used for mouse genotyping.
Supplementary Table 3 List of primers used for real-time PCR experiments.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1045 kb)

Cardiac MRI of a control mouse.

4 chamber cardiac MRI of P22 ctrl mouse. (MOV 2077 kb)

Cardiac MRI of caErbb2 mouse.

4 chamber cardiac MRI of P22 juvenile caErbb2 mouse generated according to the schema in Fig. 3b. (MOV 2917 kb)

Time-lapse movie of karyokinesis plus cytokinesis (cell division) in mono-nucleated caErbb2 CMs.

This is a representative time-lapse video of P7 caErbb2 mono-nucleated CMs performing karyokinesis plus cytokinesis (cell division). Heart cells were isolated from P7 caErbb2mice, cultured in vitro for 48 h, then labelled with TMRE to identify CMs (see ‘Methods’ section for further details) and imaged for 12 h at 10 min intervals; Scale bar 30 μm. (AVI 401 kb)

Time-lapse movie of karyokinesis with no cytokinesis (bi-nucleation) in mono-nucleated caErbb2 mice CMs.

This is a representative time-lapse video of P7 caErbb2 mono-nucleated CMs performing karyokinesis but not cytokinesis (bi-nucleation). Heart cells were isolated from P7 neonatal caErbb2m mice, cultured in vitro for 48 h, then labelled with TMRE to identify CMs (see ‘Methods’ section for further details) and imaged for 12 h at 10 min intervals; Scale bar 30 μm. (AVI 566 kb)

Time-lapse movie of cytokinesis in bi-nucleated caErbb2 mice CMs.

This is a representative time-lapse video of P7 caErbb2 bi-nucleated CMs performing cytokinesis. Heart cells were isolated from P7 neonatal caErbb2mice, cultured in vitro for 48 h, then labelled with TMRE to identify CMs (see ‘Methods’ section for further details) and imaged for 12 h at 10 min intervals; Scale bar 30 μm. (AVI 556 kb)

Time-lapse movie of CMs isolated from P7 ctrl mice.

This is a representative time-lapse video of P7 ctrl CMs, showing no events of karyokinesis and cytokinesis. Heart cells were isolated from P7 ctrl mice, cultured in vitro for 48 h, then labelled with TMRE to identify CMs (see ‘Methods’ section for further details) and imaged for 12 h at 10 min intervals; Scale bar 30 μm. (AVI 265 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

D’Uva, G., Aharonov, A., Lauriola, M. et al. ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat Cell Biol 17, 627–638 (2015). https://doi.org/10.1038/ncb3149

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3149

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