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:

Epicardial cells derived from human embryonic stem cells augment cardiomyocyte-driven heart regeneration

Abstract

The epicardium and its derivatives provide trophic and structural support for the developing and adult heart. Here we tested the ability of human embryonic stem cell (hESC)-derived epicardium to augment the structure and function of engineered heart tissue in vitro and to improve efficacy of hESC-cardiomyocyte grafts in infarcted athymic rat hearts. Epicardial cells markedly enhanced the contractility, myofibril structure and calcium handling of human engineered heart tissues, while reducing passive stiffness compared with mesenchymal stromal cells. Transplanted epicardial cells formed persistent fibroblast grafts in infarcted hearts. Cotransplantation of hESC-derived epicardial cells and cardiomyocytes doubled graft cardiomyocyte proliferation rates in vivo, resulting in 2.6-fold greater cardiac graft size and simultaneously augmenting graft and host vascularization. Notably, cotransplantation improved systolic function compared with hearts receiving either cardiomyocytes alone, epicardial cells alone or vehicle. The ability of epicardial cells to enhance cardiac graft size and function makes them a promising adjuvant therapeutic for cardiac repair.

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

Fig. 1: Generation and maturation of 3D-EHTs using hESC-EPIs and CMs.
Fig. 2: hESC-EPIs promote contractility and Ca2+ handling of 3D-EHTs.
Fig. 3: Cotransplantation of hESC-EPIs with CMs promotes microvascular density.
Fig. 4: hESC-EPIs potentiate cardiac regeneration.
Fig. 5: Cotransplantation of epicardial cells and CMs promotes functional recovery.
Fig. 6: Epicardial secretome.

Similar content being viewed by others

Data availability

The raw data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Braunwald, E. Shattuck lecture—cardiovascular medicine at the turn of the millennium: triumphs, concerns, and opportunities. N. Engl. J. Med. 337, 1360–1369 (1997).

    Article  CAS  Google Scholar 

  2. Mozaffarian, D. et al. Heart disease and stroke statistics-2016 update: a report from the American Heart Association. Circulation 133, e38–e360 (2016).

    PubMed  Google Scholar 

  3. McMurray, J. J., Petrie, M. C., Murdoch, D. R. & Davie, A. P. Clinical epidemiology of heart failure: public and private health burden. Eur. Heart J. 19, P9–P16 (1998).

    Article  Google Scholar 

  4. Bertero, A. & Murry, C. E. Hallmarks of cardiac regeneration. Nat. Rev. Cardiol. 15, 579–580 (2018).

    Article  CAS  Google Scholar 

  5. Weinberger, F., Mannhardt, I. & Eschenhagen, T. Engineering cardiac muscle tissue: a maturating field of research. Circ. Res. 120, 1487–1500 (2017).

    Article  CAS  Google Scholar 

  6. Burridge, P. W. et al. Chemically defined generation of human cardiomyocytes. Nat. Meth. 11, 855–860 (2014).

    Article  CAS  Google Scholar 

  7. Lian, X. et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl Acad. Sci. USA 109, E1848–E1857 (2012).

    Article  CAS  Google Scholar 

  8. Laflamme, M. A. et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 25, 1015–1024 (2007).

    Article  CAS  Google Scholar 

  9. Patsch, C. et al. Generation of vascular endothelial and smooth muscle cells from human pluripotent stem cells. Nat. Cell Biol. 17, 994–1003 (2015).

    Article  CAS  Google Scholar 

  10. Orlova, V. V. et al. Generation, expansion and functional analysis of endothelial cells and pericytes derived from human pluripotent stem cells. Nat. Protoc. 9, 1514–1531 (2014).

    Article  CAS  Google Scholar 

  11. Cheung, C., Bernardo, A. S., Trotter, M. W., Pedersen, R. A. & Sinha, S. Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility. Nat. Biotechnol. 30, 165–173 (2012).

    Article  CAS  Google Scholar 

  12. Iyer, D. et al. Robust derivation of epicardium and its differentiated smooth muscle cell progeny from human pluripotent stem cells. Development 142, 1528–1541 (2015).

    Article  CAS  Google Scholar 

  13. Witty, A. D. et al. Generation of the epicardial lineage from human pluripotent stem cells. Nat. Biotechnol. 32, 1026–1035 (2014).

    Article  CAS  Google Scholar 

  14. Palpant, N. J. et al. Inhibition of beta-catenin signaling respecifies anterior-like endothelium into beating human cardiomyocytes. Development 142, 3198–3209 (2015).

    Article  CAS  Google Scholar 

  15. Palpant, N. J. et al. Generating high-purity cardiac and endothelial derivatives from patterned mesoderm using human pluripotent stem cells. Nat. Protoc. 12, 15–31 (2017).

    Article  CAS  Google Scholar 

  16. Shiba, Y. et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature 489, 322–325 (2012).

    Article  CAS  Google Scholar 

  17. Weinberger, F. et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci. Transl. Med. 8, 363ra148 (2016).

    Article  Google Scholar 

  18. Chong, J. J. et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510, 273–277 (2014).

    Article  CAS  Google Scholar 

  19. Shiba, Y. et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 538, 388–391 (2016).

    Article  CAS  Google Scholar 

  20. Liu, Y. W. et al. Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat. Biotechnol. 36, 597–605 (2018).

    Article  CAS  Google Scholar 

  21. van den Berg, C. W. et al. Transcriptome of human foetal heart compared with cardiomyocytes from pluripotent stem cells. Development 142, 3231–3238 (2015).

    Article  Google Scholar 

  22. Guadix, J. A., Carmona, R., Munoz-Chapuli, R. & Perez-Pomares, J. M. In vivo and in vitro analysis of the vasculogenic potential of avian proepicardial and epicardial cells. Dev. Dyn. 235, 1014–1026 (2006).

    Article  CAS  Google Scholar 

  23. Gittenberger-de Groot, A. C., Vrancken Peeters, M. P., Mentink, M. M., Gourdie, R. G. & Poelmann, R. E. Epicardium-derived cells contribute a novel population to the myocardial wall and the atrioventricular cushions. Circ. Res. 82, 1043–1052 (1998).

    Article  CAS  Google Scholar 

  24. Dettman, R. W., Denetclaw, W. Jr., Ordahl, C. P. & Bristow, J. Common epicardial origin of coronary vascular smooth muscle, perivascular fibroblasts, and intermyocardial fibroblasts in the avian heart. Dev. Biol. 193, 169–181 (1998).

    Article  CAS  Google Scholar 

  25. Manner, J. Does the subepicardial mesenchyme contribute myocardioblasts to the myocardium of the chick embryo heart? A quail-chick chimera study tracing the fate of the epicardial primordium. Anat. Rec. 255, 212–226 (1999).

    Article  CAS  Google Scholar 

  26. Ieda, M. et al. Cardiac fibroblasts regulate myocardial proliferation through beta1 integrin signaling. Dev. Cell 16, 233–244 (2009).

    Article  CAS  Google Scholar 

  27. Eid, H. et al. Role of epicardial mesothelial cells in the modification of phenotype and function of adult rat ventricular myocytes in primary coculture. Circ. Res. 71, 40–50 (1992).

    Article  CAS  Google Scholar 

  28. Stuckmann, I., Evans, S. & Lassar, A. B. Erythropoietin and retinoic acid, secreted from the epicardium, are required for cardiac myocyte proliferation. Dev. Biol. 255, 334–349 (2003).

    Article  CAS  Google Scholar 

  29. Weeke-Klimp, A. et al. Epicardium-derived cells enhance proliferation, cellular maturation and alignment of cardiomyocytes. J. Mol. Cell. Cardiol. 49, 606–616 (2010).

    Article  CAS  Google Scholar 

  30. Braitsch, C. M., Kanisicak, O., van Berlo, J. H., Molkentin, J. D. & Yutzey, K. E. Differential expression of embryonic epicardial progenitor markers and localization of cardiac fibrosis in adult ischemic injury and hypertensive heart disease. J. Mol. Cell. Cardiol. 65, 108–19 (2013).

    Article  CAS  Google Scholar 

  31. Ruan, J. L. et al. Mechanical stress conditioning and electrical stimulation promote contractility and force maturation of induced pluripotent stem cell-derived human cardiac tissue. Circulation 134, 1557–1567 (2016).

    Article  CAS  Google Scholar 

  32. Dubois, N. C. et al. SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells. Nat. Biotechnol. 29, 1011–1018 (2011).

    Article  CAS  Google Scholar 

  33. Winter, E. M. et al. A new direction for cardiac regeneration therapy: application of synergistically acting epicardium-derived cells and cardiomyocyte progenitor cells. Circ. Heart Fail. 2, 643–653 (2009).

    Article  Google Scholar 

  34. Gerbin, K. A., Yang, X., Murry, C. E. & Coulombe, K. L. Enhanced electrical integration of engineered human myocardium via intramyocardial versus epicardial delivery in infarcted rat hearts. PloS ONE 10, e0131446 (2015).

    Article  Google Scholar 

  35. van Tuyn, J. et al. Epicardial cells of human adults can undergo an epithelial-to-mesenchymal transition and obtain characteristics of smooth muscle cells in vitro. Stem Cells 25, 271–278 (2007).

    Article  CAS  Google Scholar 

  36. Bax, N. A. et al. Epithelial-to-mesenchymal transformation alters electrical conductivity of human epicardial cells. J. Cell. Mol. Med. 15, 2675–2683 (2011).

    Article  CAS  Google Scholar 

  37. Kirby, M. L., Gale, T. F. & Stewart, D. E. Neural crest cells contribute to normal aorticopulmonary septation. Science 220, 1059–1061 (1983).

    Article  CAS  Google Scholar 

  38. Porras, D. & Brown, C. B. Temporal-spatial ablation of neural crest in the mouse results in cardiovascular defects. Dev. Dyn. 237, 153–162 (2008).

    Article  CAS  Google Scholar 

  39. Jiang, X., Rowitch, D. H., Soriano, P., McMahon, A. P. & Sucov, H. M. Fate of the mammalian cardiac neural crest. Development 127, 1607–1616 (2000).

    CAS  PubMed  Google Scholar 

  40. Cai, C. L. et al. A myocardial lineage derives from Tbx18 epicardial cells. Nature 454, 104–108 (2008).

    Article  CAS  Google Scholar 

  41. Gittenberger-de Groot, A. C., Vrancken Peeters, M. P., Bergwerff, M., Mentink, M. M. & Poelmann, R. E. Epicardial outgrowth inhibition leads to compensatory mesothelial outflow tract collar and abnormal cardiac septation and coronary formation. Circ. Res. 87, 969–971 (2000).

    Article  CAS  Google Scholar 

  42. Ogle, B. M. et al. Distilling complexity to advance cardiac tissue engineering. Sci. Transl. Med. 8, 342ps13 (2016).

    Article  Google Scholar 

  43. Lepilina, A. et al. A dynamic epicardial injury response supports progenitor cell activity during zebrafish heart regeneration. Cell 127, 607–619 (2006).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  45. Wang, J., Karra, R., Dickson, A. L. & Poss, K. D. Fibronectin is deposited by injury-activated epicardial cells and is necessary for zebrafish heart regeneration. Dev. Biol. 382, 427–435 (2013).

    Article  CAS  Google Scholar 

  46. Winter, E. M. et al. Preservation of left ventricular function and attenuation of remodeling after transplantation of human epicardium-derived cells into the infarcted mouse heart. Circulation 116, 917–927 (2007).

    Article  CAS  Google Scholar 

  47. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).

    Article  CAS  Google Scholar 

  48. Bargehr, J. et al. Embryological origin of human smooth muscle cells influences their ability to support endothelial network formation. Stem Cells Transl. Med. 5, 946–959 (2016).

    Article  Google Scholar 

  49. Hofsteen, P., Robitaille, A. M., Chapman, D. P., Moon, R. T. & Murry, C. E. Quantitative proteomics identify DAB2 as a cardiac developmental regulator that inhibits WNT/beta-catenin signaling. Proc. Natl Acad. Sci. USA 113, 1002–1007 (2016).

    Article  CAS  Google Scholar 

  50. Palpant, N. J., Hofsteen, P., Pabon, L., Reinecke, H. & Murry, C. E. Cardiac development in zebrafish and human embryonic stem cells is inhibited by exposure to tobacco cigarettes and e-cigarettes. PloS ONE 10, e0126259 (2015).

    Article  Google Scholar 

  51. Young, J. L. & Engler, A. J. Hydrogels with time-dependent material properties enhance cardiomyocyte differentiation in vitro. Biomaterials 32, 1002–1009 (2011).

    Article  CAS  Google Scholar 

  52. Ruan, J. L. et al. Mechanical stress promotes maturation of human myocardium from pluripotent stem cell-derived progenitors. Stem Cells 33, 2148–2157 (2015).

    Article  CAS  Google Scholar 

  53. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  Google Scholar 

  54. Barnett, D. W., Garrison, E. K., Quinlan, A. R., Stromberg, M. P. & Marth, G. T. BamTools: a C++ API and toolkit for analyzing and managing BAM files. Bioinformatics 27, 1691–1692 (2011).

    Article  CAS  Google Scholar 

  55. Edgar, R., Domrachev, M. & Lash, A. E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  Google Scholar 

  56. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

  57. Binns, D. et al. QuickGO: a web-based tool for Gene Ontology searching. Bioinformatics 25, 3045–3046 (2009).

    Article  CAS  Google Scholar 

  58. Eisen, M. B., Spellman, P. T., Brown, P. O. & Botstein, D. Cluster analysis and display of genome-wide expression patterns. Proc. Natl Acad. Sci. USA 95, 14863–14868 (1998).

    Article  CAS  Google Scholar 

  59. Wang, J., Vasaikar, S., Shi, Z., Greer, M. & Zhang, B. WebGestalt 2017: a more comprehensive, powerful, flexible and interactive gene set enrichment analysis toolkit. Nucleic Acids Res. 45, W130–W137 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the British Heart Foundation (BHF; grants nos. NH/11/1/28922, G1000847, FS/13/29/30024 and FS/18/46/33663), Oxford-Cambridge Centre for Regenerative Medicine (grant no. RM/13/3/30159), the UK Medical Research Council (MRC) and the Cambridge Hospitals National Institute for Health Research Biomedical Research Centre funding (S.S.), as well as National Institutes of Health grant nos. P01HL094374, P01GM081619 and R01HL128362 and a grant from the Fondation Leducq Transatlantic Network of Excellence (C.E.M.). J.B. was supported by a Cambridge National Institute for Health Research Biomedical Research Centre Cardiovascular Clinical Research Fellowship and, subsequently, by a BHF Studentship (grant no. FS/13/65/30441). D.I. received a University of Cambridge Commonwealth Scholarship. L.G. is supported by BHF Award RM/l3/3/30159 and L.P.O. is funded by a Wellcome Trust Fellowship (grant no. 203568/Z/16/Z). N.F. was supported by BHF grant no. RG/13/14/30314. N.L.N. was supported by the Biotechnology and Biological Sciences Research Council (Institute Strategic Programmes BBS/E/B/000C0419 and BBS/E/B/000C0434). S.S. and M.R.B. were supported by the BHF Centre for Cardiovascular Research Excellence. Core support was provided by the Wellcome-MRC Cambridge Stem Cell Institute (grant no. 203151/Z/16/Z). The authors thank Osiris for providing the primary mesenchymal stem cells47.

Author information

Authors and Affiliations

Authors

Contributions

J.B. was the principal experimentalist and was responsible for study design and conceptualization, data acquisition and interpretation, production of figures and manuscript writing. L.P.O. performed tissue culture, 3D-EHT generation, force measurement and assisted during surgery. M.C. performed tissue culture and 3D-EHT generation. H.D. performed force measurement and RT–qPCR. P.H. was responsible for preparation of cell suspensions on the day of transplantation, necropsy, assisting during surgery and postoperative animal care. S.B. performed the casting of 3D-EHT and force measurement. L.G. performed tissue histology, immunofluorescence and sample preparation for RNAseq. N.L.N. performed the bioinformatics analysis. D.I. contributed conceptual ideas and critically revised the manuscript for important intellectual content. F.S. critically revised the manuscript for important intellectual content. F.W. performed the functional analysis of echocardiographs. A.B. performed the gene expression analysis. A.L. was responsible for experimental guidance and force measurement data analysis. W.G.B. was responsible for data interpretation and logistics. A.M. was responsible for animal surgery and logistics. N.F. was responsible for processing of histologic tissue and preparation of slides. M.R. was responsible for force measurement equipment. M.R.B. critically revised the manuscript for important intellectual content. C.E.M. was responsible for study design and conception, obtaining research funding, study supervision, and editing and final approval of the manuscript. S.S. was responsible for study design and conception, obtaining research funding, study supervision, interpretation of data, and editing and final approval of the manuscript.

Corresponding authors

Correspondence to Charles E. Murry or Sanjay Sinha.

Ethics declarations

Competing interests

A patent has been filed on the cardiac application of epicardial cells, on which C.E.M., S.S. and J.B. are coinventors (WO2018170280A1). C.E.M. is a scientific founder and equity holder in Cytocardia.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figs. 1–20 and Supplementary Tables. 1–5

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bargehr, J., Ong, L.P., Colzani, M. et al. Epicardial cells derived from human embryonic stem cells augment cardiomyocyte-driven heart regeneration. Nat Biotechnol 37, 895–906 (2019). https://doi.org/10.1038/s41587-019-0197-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0197-9

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research