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:

The deployment of cell lineages that form the mammalian heart

Abstract

The function of the mammalian heart depends on the interplay between different cardiac cell types. The deployment of these cells, with precise spatiotemporal regulation, is also important during development to establish the heart structure. In this Review, we discuss the diverse origins of cardiac cell types and the lineage relationships between cells of a given type that contribute to different parts of the heart. The emerging lineage tree shows the progression of cell fate diversification, with patterning cues preceding cell type segregation, as well as points of convergence, with overlapping lineages contributing to a given tissue. Several cell lineage markers have been identified. However, caution is required with genetic-tracing experiments in comparison with clonal analyses. Genetic studies on cell populations provided insights into the mechanisms for lineage decisions. In the past 3 years, results of single-cell transcriptomics are beginning to reveal cell heterogeneity and early developmental trajectories. Equating this information with the in vivo location of cells and their lineage history is a current challenge. Characterization of the progenitor cells that form the heart and of the gene regulatory networks that control their deployment is of major importance for understanding the origin of congenital heart malformations and for producing cardiac tissue for use in regenerative medicine.

Key points

  • Clonal analysis shows that two myocardial cell lineages, which segregate early at gastrulation, form the heart, with cell sublineages contributing to the arterial and venous poles, with early left–right delineation.

  • The origin and cell fate choices of non-myocardial progenitors, such as those that give rise to cardiac interstitial fibroblasts or the coronary vasculature, are now clearer.

  • Overlapping cell sources provide potential for compensatory mechanisms, and thus developmental robustness, a process that is just beginning to be characterized.

  • As the heart begins to form, cardiac progenitors are located in the first and second heart fields, with characteristic and diverse gene expression patterns marking their cardiac contributions, which correspond to the first and second myocardial lineages.

  • Gene regulatory networks, governed by transcription factors, cofactors and chromatin modifications, in which non-coding RNAs also participate, control the deployment of cardiac progenitor cells during cardiogenesis.

  • Single-cell analyses have identified early cardiac progenitor cell types, providing new insights into cell heterogeneity and developmental trajectories of cardiac cells as the heart develops.

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: Cardiogenesis stages in the mouse.
Fig. 2: Cell lineage tree of cardiac and associated skeletal muscles.
Fig. 3: Location of cardiac progenitors and their derivatives.
Fig. 4: Gene regulatory networks in cardiac development.
Fig. 5: Sources of the different cardiac cell types.

Similar content being viewed by others

References

  1. Galli, D. et al. Atrial myocardium derives from the posterior region of the second heart field, which acquires left-right identity as Pitx2c is expressed. Development 135, 1157–1167 (2008).

    CAS  PubMed  Google Scholar 

  2. Kelly, R. G., Brown, N. A. & Buckingham, M. E. The arterial pole of the mouse heart forms from Fgf10-expressing cells in pharyngeal mesoderm. Dev. Cell 1, 435–440 (2001).

    CAS  PubMed  Google Scholar 

  3. Zaffran, S., Kelly, R. G., Meilhac, S. M., Buckingham, M. E. & Brown, N. A. Right ventricular myocardium derives from the anterior heart field. Circ. Res. 95, 261–268 (2004).

    CAS  PubMed  Google Scholar 

  4. Le Garrec, J. F. et al. A predictive model of asymmetric morphogenesis from 3D reconstructions of mouse heart looping dynamics. Elife 6, e28951 (2017).

    PubMed  PubMed Central  Google Scholar 

  5. Meilhac, S. M., Esner, M., Kerszberg, M., Moss, J. E. & Buckingham, M. E. Oriented clonal cell growth in the developing mouse myocardium underlies cardiac morphogenesis. J. Cell Biol. 164, 97–109 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Christoffels, V. M. et al. Chamber formation and morphogenesis in the developing mammalian heart. Dev. Biol. 223, 266–278 (2000).

    CAS  PubMed  Google Scholar 

  7. Hoffman, J. I. & Kaplan, S. The incidence of congenital heart disease. J. Am. Coll. Cardiol. 39, 1890–1900 (2002).

    PubMed  Google Scholar 

  8. de la Cruz, M., Sanchez-Gomez, C. & Palomino, M. The primitive cardiac regions in the straight heart tube (stage 9-) and their anatomical expression in the mature heart: an experimental study in the chick embryo. J. Anat. 165, 121–131 (1989).

    Google Scholar 

  9. Garcia-Martinez, V. & Schoenwolf, G. C. Primitive-streak origin of the cardiovascular system in avian embryos. Dev. Biol. 159, 706–719 (1993).

    CAS  PubMed  Google Scholar 

  10. Mikawa, T., Borisov, A., Brown, A. M. & Fischman, D. A. Clonal analysis of cardiac morphogenesis in the chicken embryo using a replication-defective retrovirus: I. Formation of the ventricular myocardium. Dev. Dyn. 193, 11–23 (1992).

    CAS  PubMed  Google Scholar 

  11. Rawles, M. E. The heart-forming areas of the early chick blastoderm. Physiol. Zool. 16, 22–42 (1943).

    Google Scholar 

  12. Redkar, A., Montgomery, M. & Litvin, J. Fate map of early avian cardiac progenitor cells. Development 128, 2269–2279 (2001).

    CAS  PubMed  Google Scholar 

  13. Stalsberg, H. The origin of heart asymmetry: right and left contributions to the early chick embryo heart. Dev. Biol. 19, 109–127 (1969).

    CAS  PubMed  Google Scholar 

  14. Waldo, K. L., Lo, C. W. & Kirby, M. L. Connexin 43 expression reflects neural crest patterns during cardiovascular development. Dev. Biol. 208, 307–323 (1999).

    CAS  PubMed  Google Scholar 

  15. Buckingham, M., Biben, C. & Lawson, K. in Genetic Control of Heart Development (eds Olson, E. N., Harvey, R. P., Schulz, R. A. & Altman, J. S.) 31–33 (Strasbourg : HFSP Pub, 1997).

    Google Scholar 

  16. Lawson, K. A. & Pedersen, R. A. Cell fate, morphogenetic movement and population kinetics of embryonic endoderm at the time of germ layer formation in the mouse. Development 101, 627–652 (1987).

    CAS  PubMed  Google Scholar 

  17. Tzouanacou, E., Wegener, A., Wymeersch, F. J., Wilson, V. & Nicolas, J. F. Redefining the progression of lineage segregations during mammalian embryogenesis by clonal analysis. Dev. Cell 17, 365–376 (2009).

    CAS  PubMed  Google Scholar 

  18. Kinder, S. J. et al. The orderly allocation of mesodermal cells to the extraembryonic structures and the anteroposterior axis during gastrulation of the mouse embryo. Development 126, 4691–4701 (1999).

    CAS  PubMed  Google Scholar 

  19. Ivanovitch, K., Temino, S. & Torres, M. Live imaging of heart tube development in mouse reveals alternating phases of cardiac differentiation and morphogenesis. Elife 6, e30668 (2017).

    PubMed  PubMed Central  Google Scholar 

  20. Kaufman, M. H. & Navaratnam, V. Early differentiation of the heart in mouse embryos. J. Anat. 133, 235–246 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Tam, P. P., Parameswaran, M., Kinder, S. J. & Weinberger, R. P. The allocation of epiblast cells to the embryonic heart and other mesodermal lineages: the role of ingression and tissue movement during gastrulation. Development 124, 1631–1642 (1997).

    CAS  PubMed  Google Scholar 

  22. Meilhac, S. M., Esner, M., Kelly, R. G., Nicolas, J. F. & Buckingham, M. E. The clonal origin of myocardial cells in different regions of the embryonic mouse heart. Dev. Cell 6, 685–698 (2004).

    CAS  PubMed  Google Scholar 

  23. Ragni, C. V. et al. Amotl1 mediates sequestration of the Hippo effector Yap1 downstream of Fat4 to restrict heart growth. Nat. Commun. 8, 14582 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Saga, Y. et al. MesP1 is expressed in the heart precursor cells and required for the formation of a single heart tube. Development 126, 3437–3447 (1999).

    CAS  PubMed  Google Scholar 

  25. Buckingham, M. E. & Meilhac, S. M. Tracing cells for tracking cell lineage and clonal behavior. Dev. Cell 21, 394–409 (2011).

    CAS  PubMed  Google Scholar 

  26. Lescroart, F. et al. Early lineage restriction in temporally distinct populations of Mesp1 progenitors during mammalian heart development. Nat. Cell Biol. 16, 829–840 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chabab, S. et al. Uncovering the number and clonal dynamics of Mesp1 progenitors during heart morphogenesis. Cell Rep. 14, 1–10 (2016).

    CAS  PubMed  Google Scholar 

  28. Zong, H., Espinosa, J. S., Su, H. H., Muzumdar, M. D. & Luo, L. Mosaic analysis with double markers in mice. Cell 121, 479–492 (2005).

    CAS  PubMed  Google Scholar 

  29. Devine, W. P., Wythe, J. D., George, M., Koshiba-Takeuchi, K. & Bruneau, B. G. Early patterning and specification of cardiac progenitors in gastrulating mesoderm. Elife 3, e03848 (2014).

    PubMed Central  Google Scholar 

  30. Franco, D. et al. Left and right ventricular contributions to the formation of the interventricular septum in the mouse heart. Dev. Biol. 294, 366–375 (2006).

    CAS  PubMed  Google Scholar 

  31. Buckingham, M. Gene regulatory networks and cell lineages that underlie the formation of skeletal muscle. Proc. Natl Acad. Sci. USA 114, 5830–5837 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lescroart, F. et al. Clonal analysis reveals common lineage relationships between head muscles and second heart field derivatives in the mouse embryo. Development 137, 3269–3279 (2010).

    CAS  PubMed  Google Scholar 

  33. Lescroart, F. et al. Clonal analysis reveals a common origin between nonsomite-derived neck muscles and heart myocardium. Proc. Natl Acad. Sci. USA 112, 1446–1451 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Lescroart, F., Mohun, T., Meilhac, S. M., Bennett, M. & Buckingham, M. Lineage tree for the venous pole of the heart: clonal analysis clarifies controversial genealogy based on genetic tracing. Circ. Res. 111, 1313–1322 (2012).

    CAS  PubMed  Google Scholar 

  35. Dominguez, J. N., Meilhac, S. M., Bland, Y. S., Buckingham, M. E. & Brown, N. A. Asymmetric fate of the posterior part of the second heart field results in unexpected left/right contributions to both poles of the heart. Circ. Res. 111, 1323–1335 (2012).

    CAS  PubMed  Google Scholar 

  36. Bajolle, F. et al. Conotruncal defects associated with anomalous pulmonary venous connections. Arch. Cardiovasc. Dis. 102, 105–110 (2009).

    PubMed  Google Scholar 

  37. Meilhac, S. M. et al. A retrospective clonal analysis of the myocardium reveals two phases of clonal growth in the developing mouse heart. Development 130, 3877–3889 (2003).

    CAS  PubMed  Google Scholar 

  38. Del Monte-Nieto, G. et al. Control of cardiac jelly dynamics by NOTCH1 and NRG1 defines the building plan for trabeculation. Nature 557, 439–445 (2018).

    PubMed  Google Scholar 

  39. Tian, X. et al. Identification of a hybrid myocardial zone in the mammalian heart after birth. Nat. Commun. 8, 87 (2017).

    PubMed  PubMed Central  Google Scholar 

  40. Miquerol, L. et al. Resolving cell lineage contributions to the ventricular conduction system with a Cx40-GFP allele: a dual contribution of the first and second heart fields. Dev. Dyn. 242, 665–677 (2013).

    CAS  PubMed  Google Scholar 

  41. Miquerol, L. et al. Biphasic development of the mammalian ventricular conduction system. Circ. Res. 107, 153–161 (2010).

    CAS  PubMed  Google Scholar 

  42. Bardot, E. et al. Foxa2 identifies a cardiac progenitor population with ventricular differentiation potential. Nat. Commun. 8, 14428 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ang, S. L. et al. The formation and maintenance of the definitive endoderm lineage in the mouse: involvement of HNF3/forkhead proteins. Development 119, 1301–1315 (1993).

    CAS  PubMed  Google Scholar 

  44. Kinder, S. J. et al. The organizer of the mouse gastrula is composed of a dynamic population of progenitor cells for the axial mesoderm. Development 128, 3623–3634 (2001).

    CAS  PubMed  Google Scholar 

  45. Park, E. J. et al. System for tamoxifen-inducible expression of cre-recombinase from the Foxa2 locus in mice. Dev. Dyn. 237, 447–453 (2008).

    CAS  PubMed  Google Scholar 

  46. Verzi, M. P., McCulley, D. J., De Val, S., Dodou, E. & Black, B. L. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev. Biol. 287, 134–145 (2005).

    CAS  PubMed  Google Scholar 

  47. Buckingham, M., Meilhac, S. & Zaffran, S. Building the mammalian heart from two sources of myocardial cells. Nat. Rev. Genet. 6, 826–835 (2005).

    CAS  PubMed  Google Scholar 

  48. Bruneau, B. G. et al. Chamber-specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome. Dev. Biol. 211, 100–108 (1999).

    CAS  PubMed  Google Scholar 

  49. Rana, M. S. et al. Tbx1 coordinates addition of posterior second heart field progenitor cells to the arterial and venous poles of the heart. Circ. Res. 115, 790–799 (2014).

    CAS  PubMed  Google Scholar 

  50. Vincentz, J. W., Toolan, K. P., Zhang, W. & Firulli, A. B. Hand factor ablation causes defective left ventricular chamber development and compromised adult cardiac function. PLOS Genet. 13, e1006922 (2017).

    PubMed  PubMed Central  Google Scholar 

  51. Barnes, R. M., Firulli, B. A., Conway, S. J., Vincentz, J. W. & Firulli, A. B. Analysis of the Hand1 cell lineage reveals novel contributions to cardiovascular, neural crest, extra-embryonic, and lateral mesoderm derivatives. Dev. Dyn. 239, 3086–3097 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Liang, X. et al. HCN4 dynamically marks the first heart field and conduction system precursors. Circ. Res. 113, 399–407 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Spater, D. et al. A HCN4+ cardiomyogenic progenitor derived from the first heart field and human pluripotent stem cells. Nat. Cell Biol. 15, 1098–1106 (2013).

    PubMed  Google Scholar 

  54. Fujii, M. et al. Sfrp5 identifies murine cardiac progenitors for all myocardial structures except for the right ventricle. Nat. Commun. 8, 14664 (2017).

    PubMed  PubMed Central  Google Scholar 

  55. Watanabe, Y. et al. Role of mesodermal FGF8 and FGF10 overlaps in the development of the arterial pole of the heart and pharyngeal arch arteries. Circ. Res. 106, 495–503 (2010).

    CAS  PubMed  Google Scholar 

  56. Cai, C. L. et al. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 5, 877–889 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Engleka, K. A. et al. Islet1 derivatives in the heart are of both neural crest and second heart field origin. Circ. Res. 110, 922–926 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ma, Q., Zhou, B. & Pu, W. T. Reassessment of Isl1 and Nkx2-5 cardiac fate maps using a Gata4-based reporter of Cre activity. Dev. Biol. 323, 98–104 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Sun, Y. et al. Islet 1 is expressed in distinct cardiovascular lineages, including pacemaker and coronary vascular cells. Dev. Biol. 304, 286–296 (2007).

    CAS  PubMed  Google Scholar 

  60. Prall, O. W. et al. An Nkx2-5/Bmp2/Smad1 negative feedback loop controls heart progenitor specification and proliferation. Cell 128, 947–959 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Stanley, E. G. et al. Efficient Cre-mediated deletion in cardiac progenitor cells conferred by a 3’UTR-ires-Cre allele of the homeobox gene Nkx2-5. Int. J. Dev. Biol. 46, 431–439 (2002).

    CAS  PubMed  Google Scholar 

  62. Seo, S. & Kume, T. Forkhead transcription factors, Foxc1 and Foxc2, are required for the morphogenesis of the cardiac outflow tract. Dev. Biol. 296, 421–436 (2006).

    CAS  PubMed  Google Scholar 

  63. Zhou, Z. et al. Temporally distinct Six2-positive second heart field progenitors regulate mammalian heart development and disease. Cell Rep. 18, 1019–1032 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Baldini, A., Fulcoli, F. G. & Illingworth, E. Tbx1: transcriptional and developmental functions. Curr. Top. Dev. Biol. 122, 223–243 (2017).

    CAS  PubMed  Google Scholar 

  65. Theveniau-Ruissy, M. et al. The del22q11.2 candidate gene Tbx1 controls regional outflow tract identity and coronary artery patterning. Circ. Res. 103, 142–148 (2008).

    CAS  PubMed  Google Scholar 

  66. Huynh, T., Chen, L., Terrell, P. & Baldini, A. A fate map of Tbx1 expressing cells reveals heterogeneity in the second cardiac field. Genesis 45, 470–475 (2007).

    CAS  PubMed  Google Scholar 

  67. Bajolle, F. et al. Myocardium at the base of the aorta and pulmonary trunk is prefigured in the outflow tract of the heart and in subdomains of the second heart field. Dev. Biol. 313, 25–34 (2008).

    CAS  PubMed  Google Scholar 

  68. Kelly, R. G. & Papaioannou, V. E. Visualization of outflow tract development in the absence of Tbx1 using an FgF10 enhancer trap transgene. Dev. Dyn. 236, 821–828 (2007).

    CAS  PubMed  Google Scholar 

  69. Cortes, C., Francou, A., De Bono, C. & Kelly, R. G. Epithelial properties of the second heart field. Circ. Res. 122, 142–154 (2018).

    CAS  PubMed  Google Scholar 

  70. Zhou, L. et al. Tbx5 and Osr1 interact to regulate posterior second heart field cell cycle progression for cardiac septation. J. Mol. Cell Cardiol. 85, 1–12 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Tian, Y. et al. Characterization and in vivo pharmacological rescue of a Wnt2-Gata6 pathway required for cardiac inflow tract development. Dev. Cell 18, 275–287 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Christoffels, V. M. et al. Formation of the venous pole of the heart from an Nkx2-5-negative precursor population requires Tbx18. Circ. Res. 98, 1555–1563 (2006).

    CAS  PubMed  Google Scholar 

  73. Mommersteeg, M. T. et al. The sinus venosus progenitors separate and diversify from the first and second heart fields early in development. Cardiovasc. Res. 87, 92–101 (2010).

    CAS  PubMed  Google Scholar 

  74. Bertrand, N. et al. Hox genes define distinct progenitor sub-domains within the second heart field. Dev. Biol. 353, 266–274 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Furtado, M. B., Biben, C., Shiratori, H., Hamada, H. & Harvey, R. P. Characterization of Pitx2c expression in the mouse heart using a reporter transgene. Dev. Dyn. 240, 195–203 (2011).

    CAS  PubMed  Google Scholar 

  76. Bajolle, F. et al. Rotation of the myocardial wall of the outflow tract is implicated in the normal positioning of the great arteries. Circ. Res. 98, 421–428 (2006).

    CAS  PubMed  Google Scholar 

  77. Liu, C. et al. Pitx2c patterns anterior myocardium and aortic arch vessels and is required for local cell movement into atrioventricular cushions. Development 129, 5081–5091 (2002).

    CAS  PubMed  Google Scholar 

  78. Dyer, L. A. & Kirby, M. L. The role of secondary heart field in cardiac development. Dev. Biol. 336, 137–144 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Kelly, R. G., Buckingham, M. E. & Moorman, A. F. Heart fields and cardiac morphogenesis. Cold Spring Harb. Perspect. Med. 4, a015750 (2014).

    PubMed  PubMed Central  Google Scholar 

  80. Vincent, S. D. & Buckingham, M. E. How to make a heart: the origin and regulation of cardiac progenitor cells. Curr. Top. Dev. Biol. 90, 1–41 (2010).

    PubMed  Google Scholar 

  81. Jain, R. et al. HEART DEVELOPMENT. Integration of Bmp and Wnt signaling by Hopx specifies commitment of cardiomyoblasts. Science 348, aaa6071(2015).

    PubMed  PubMed Central  Google Scholar 

  82. Kang, J., Nathan, E., Xu, S. M., Tzahor, E. & Black, B. L. Isl1 is a direct transcriptional target of Forkhead transcription factors in second-heart-field-derived mesoderm. Dev. Biol. 334, 513–522 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Kappen, C. & Salbaum, J. M. Identification of regulatory elements in the Isl1 gene locus. Int. J. Dev. Biol. 53, 935–946 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Dodou, E., Verzi, M. P., Anderson, J. P., Xu, S. M. & Black, B. L. Mef2c is a direct transcriptional target of ISL1 and GATA factors in the anterior heart field during mouse embryonic development. Development 131, 3931–3942 (2004).

    CAS  PubMed  Google Scholar 

  85. Watanabe, Y. et al. Fibroblast growth factor 10 gene regulation in the second heart field by Tbx1, Nkx2-5, and Islet1 reveals a genetic switch for down-regulation in the myocardium. Proc. Natl Acad. Sci. USA 109, 18273–18280 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Guo, C. et al. A Tbx1-Six1/Eya1-Fgf8 genetic pathway controls mammalian cardiovascular and craniofacial morphogenesis. J. Clin. Invest. 121, 1585–1595 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Yuan, X. et al. Disruption of spatiotemporal hypoxic signaling causes congenital heart disease in mice. J. Clin. Invest. 127, 2235–2248 (2017).

    PubMed  PubMed Central  Google Scholar 

  88. Liberatore, C. M., Searcy-Schrick, R. D., Vincent, E. B. & Yutzey, K. E. Nkx-2.5 gene induction in mice is mediated by a Smad consensus regulatory region. Dev. Biol. 244, 243–256 (2002).

    CAS  PubMed  Google Scholar 

  89. Lien, C. L., McAnally, J., Richardson, J. A. & Olson, E. N. Cardiac-specific activity of an Nkx2-5 enhancer requires an evolutionarily conserved Smad binding site. Dev. Biol. 244, 257–266 (2002).

    CAS  PubMed  Google Scholar 

  90. von Both, I. et al. Foxh1 is essential for development of the anterior heart field. Dev. Cell 7, 331–345 (2004).

    Google Scholar 

  91. Dorn, T. et al. Direct nkx2-5 transcriptional repression of isl1 controls cardiomyocyte subtype identity. Stem Cells 33, 1113–1129 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Xie, L. et al. Tbx5-hedgehog molecular networks are essential in the second heart field for atrial septation. Dev. Cell 23, 280–291 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Snarr, B. S., Wirrig, E. E., Phelps, A. L., Trusk, T. C. & Wessels, A. A spatiotemporal evaluation of the contribution of the dorsal mesenchymal protrusion to cardiac development. Dev. Dyn. 236, 1287–1294 (2007).

    PubMed  PubMed Central  Google Scholar 

  94. Snarr, B. S. et al. Isl1 expression at the venous pole identifies a novel role for the second heart field in cardiac development. Circ. Res. 101, 971–974 (2007).

    CAS  PubMed  Google Scholar 

  95. Goddeeris, M. M. et al. Intracardiac septation requires hedgehog-dependent cellular contributions from outside the heart. Development 135, 1887–1895 (2008).

    CAS  PubMed  Google Scholar 

  96. Hoffmann, A. D., Peterson, M. A., Friedland-Little, J. M., Anderson, S. A. & Moskowitz, I. P. Sonic hedgehog is required in pulmonary endoderm for atrial septation. Development 136, 1761–1770 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Hoffmann, A. D. et al. Foxf genes integrate tbx5 and hedgehog pathways in the second heart field for cardiac septation. PLOS Genet. 10, e1004604 (2014).

    PubMed  PubMed Central  Google Scholar 

  98. Zhou, L. et al. Gata4 potentiates second heart field proliferation and Hedgehog signaling for cardiac septation. Proc. Natl Acad. Sci. USA 114, E1422–E1431 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Durocher, D., Charron, F., Warren, R., Schwartz, R. J. & Nemer, M. The cardiac transcription factors Nkx2-5 and GATA-4 are mutual cofactors. EMBO J. 16, 5687–5696 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Garg, V. et al. GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature 424, 443–447 (2003).

    CAS  PubMed  Google Scholar 

  101. Maitra, M. et al. Interaction of Gata4 and Gata6 with Tbx5 is critical for normal cardiac development. Dev. Biol. 326, 368–377 (2009).

    CAS  PubMed  Google Scholar 

  102. Ang, Y. S. et al. Disease model of GATA4 mutation reveals transcription factor cooperativity in human cardiogenesis. Cell 167, 1734–1749.e22 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Luna-Zurita, L. et al. Complex interdependence regulates heterotypic transcription factor distribution and coordinates cardiogenesis. Cell 164, 999–1014 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Bouveret, R. et al. NKX2-5 mutations causative for congenital heart disease retain functionality and are directed to hundreds of targets. Elife 4, e06942 (2015).

    PubMed Central  Google Scholar 

  105. Witzel, H. R. et al. The LIM protein Ajuba restricts the second heart field progenitor pool by regulating Isl1 activity. Dev. Cell 23, 58–70 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Caputo, L. et al. The Isl1/Ldb1 complex orchestrates genome-wide chromatin organization to instruct differentiation of multipotent cardiac progenitors. Cell Stem Cell 17, 287–299 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Stoller, J. Z. et al. Ash2l interacts with Tbx1 and is required during early embryogenesis. Exp. Biol. Med. (Maywood) 235, 569–576 (2010).

    CAS  Google Scholar 

  108. Fulcoli, F. G. et al. Rebalancing gene haploinsufficiency in vivo by targeting chromatin. Nat. Commun. 7, 11688 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Chen, L. et al. Transcriptional control in cardiac progenitors: Tbx1 interacts with the BAF chromatin remodeling complex and regulates Wnt5a. PLOS Genet. 8, e1002571 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Takeuchi, J. K. & Bruneau, B. G. Directed transdifferentiation of mouse mesoderm to heart tissue by defined factors. Nature 459, 708–711 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Poleshko, A. et al. Genome-nuclear lamina interactions regulate cardiac stem cell lineage restriction. Cell 171, 573–587.e14 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Xue, Z. et al. A G-rich motif in the lncRNA braveheart interacts with a zinc-finger transcription factor to specify the cardiovascular lineage. Mol. Cell 64, 37–50 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Epstein, J. A. et al. Migration of cardiac neural crest cells in Splotch embryos. Development 127, 1869–1878 (2000).

    CAS  PubMed  Google Scholar 

  116. 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 

  117. Kimura, K., Ieda, M. & Fukuda, K. Development, maturation, and transdifferentiation of cardiac sympathetic nerves. Circ. Res. 110, 325–336 (2012).

    CAS  PubMed  Google Scholar 

  118. Saga, Y., Kitajima, S. & Miyagawa-Tomita, S. Mesp1 expression is the earliest sign of cardiovascular development. Trends Cardiovasc. Med. 10, 345–352 (2000).

    CAS  PubMed  Google Scholar 

  119. Kattman, S. J., Huber, T. L. & Keller, G. M. Multipotent flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. Dev. Cell 11, 723–732 (2006).

    CAS  PubMed  Google Scholar 

  120. Moretti, A. et al. Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 127, 1151–1165 (2006).

    CAS  PubMed  Google Scholar 

  121. Wu, S. M. et al. Developmental origin of a bipotential myocardial and smooth muscle cell precursor in the mammalian heart. Cell 127, 1137–1150 (2006).

    CAS  PubMed  Google Scholar 

  122. Nakano, A., Nakano, H., Smith, K. A. & Palpant, N. J. The developmental origins and lineage contributions of endocardial endothelium. Biochim. Biophys. Acta 1863, 1937–1947 (2016).

    CAS  PubMed  Google Scholar 

  123. Ema, M., Takahashi, S. & Rossant, J. Deletion of the selection cassette, but not cis-acting elements, in targeted Flk1-lacZ allele reveals Flk1 expression in multipotent mesodermal progenitors. Blood 107, 111–117 (2006).

    CAS  PubMed  Google Scholar 

  124. Motoike, T., Markham, D. W., Rossant, J. & Sato, T. N. Evidence for novel fate of Flk1+ progenitor: contribution to muscle lineage. Genesis 35, 153–159 (2003).

    PubMed  Google Scholar 

  125. de la Pompa, J. L. et al. Role of the NF-ATc transcription factor in morphogenesis of cardiac valves and septum. Nature 392, 182–186 (1998).

    PubMed  Google Scholar 

  126. Misfeldt, A. M. et al. Endocardial cells are a distinct endothelial lineage derived from Flk1+ multipotent cardiovascular progenitors. Dev. Biol. 333, 78–89 (2009).

    CAS  PubMed  Google Scholar 

  127. Wu, B. et al. Nfatc1 coordinates valve endocardial cell lineage development required for heart valve formation. Circ. Res. 109, 183–192 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Nakano, H. et al. Haemogenic endocardium contributes to transient definitive haematopoiesis. Nat. Commun. 4, 1564 (2013).

    PubMed  Google Scholar 

  129. Peterkin, T., Gibson, A. & Patient, R. Common genetic control of haemangioblast and cardiac development in zebrafish. Development 136, 1465–1474 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Schulte, I., Schlueter, J., Abu-Issa, R., Brand, T. & Manner, J. Morphological and molecular left-right asymmetries in the development of the proepicardium: a comparative analysis on mouse and chick embryos. Dev. Dyn. 236, 684–695 (2007).

    PubMed  Google Scholar 

  131. Wessels, A. et al. Epicardially derived fibroblasts preferentially contribute to the parietal leaflets of the atrioventricular valves in the murine heart. Dev. Biol. 366, 111–124 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhou, B., von Gise, A., Ma, Q., Rivera-Feliciano, J. & Pu, W. T. Nkx2-5- and Isl1-expressing cardiac progenitors contribute to proepicardium. Biochem. Biophys. Res. Commun. 375, 450–453 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Zhou, B. et al. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature 454, 109–113 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Christoffels, V. M. et al. Tbx18 and the fate of epicardial progenitors. Nature 458, E9–E10 (2009).

    Google Scholar 

  136. Rudat, C. & Kispert, A. Wt1 and epicardial fate mapping. Circ. Res. 111, 165–169 (2012).

    CAS  PubMed  Google Scholar 

  137. Martinez-Estrada, O. M. et al. Wt1 is required for cardiovascular progenitor cell formation through transcriptional control of Snail and E-cadherin. Nat. Genet. 42, 89–93 (2010).

    CAS  PubMed  Google Scholar 

  138. Villa Del Campo, C. et al. Myc overexpression enhances of epicardial contribution to the developing heart and promotes extensive expansion of the cardiomyocyte population. Sci. Rep. 6, 35366 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Ivey, M. J. & Tallquist, M. D. Defining the cardiac fibroblast. Circ. J. 80, 2269–2276 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Kolditz, D. P. et al. Epicardium-derived cells in development of annulus fibrosis and persistence of accessory pathways. Circulation 117, 1508–1517 (2008).

    PubMed  Google Scholar 

  141. Zhou, B., von Gise, A., Ma, Q., Hu, Y. W. & Pu, W. T. Genetic fate mapping demonstrates contribution of epicardium-derived cells to the annulus fibrosis of the mammalian heart. Dev. Biol. 338, 251–261 (2010).

    CAS  PubMed  Google Scholar 

  142. Ali, S. R. et al. Developmental heterogeneity of cardiac fibroblasts does not predict pathological proliferation and activation. Circ. Res. 115, 625–635 (2014).

    CAS  PubMed  Google Scholar 

  143. Moore-Morris, T. et al. Resident fibroblast lineages mediate pressure overload-induced cardiac fibrosis. J. Clin. Invest. 124, 2921–2934 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Acharya, A. et al. The bHLH transcription factor Tcf21 is required for lineage-specific EMT of cardiac fibroblast progenitors. Development 139, 2139–2149 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Kanisicak, O. et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat. Commun. 7, 12260 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Lie-Venema, H. et al. Origin, fate, and function of epicardium-derived cells (EPDCs) in normal and abnormal cardiac development. ScientificWorldJournal 7, 1777–1798 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Chen, Q. et al. Endothelial cells are progenitors of cardiac pericytes and vascular smooth muscle cells. Nat. Commun. 7, 12422 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Mikawa, T. & Fischman, D. A. Retroviral analysis of cardiac morphogenesis: discontinuous formation of coronary vessels. Proc. Natl Acad. Sci. USA 89, 9504–9508 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Sharma, B., Chang, A. & Red-Horse, K. Coronary artery development: progenitor cells and differentiation pathways. Annu. Rev. Physiol. 79, 1–19 (2017).

    CAS  PubMed  Google Scholar 

  150. Tian, X., Pu, W. T. & Zhou, B. Cellular origin and developmental program of coronary angiogenesis. Circ. Res. 116, 515–530 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Red-Horse, K., Ueno, H., Weissman, I. L. & Krasnow, M. A. Coronary arteries form by developmental reprogramming of venous cells. Nature 464, 549–553 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Chen, H. I. et al. The sinus venosus contributes to coronary vasculature through VEGFC-stimulated angiogenesis. Development 141, 4500–4512 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Katz, T. C. et al. Distinct compartments of the proepicardial organ give rise to coronary vascular endothelial cells. Dev. Cell 22, 639–650 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Lavine, K. J. et al. Fibroblast growth factor signals regulate a wave of Hedgehog activation that is essential for coronary vascular development. Genes Dev. 20, 1651–1666 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Tian, X. et al. Subepicardial endothelial cells invade the embryonic ventricle wall to form coronary arteries. Cell Res. 23, 1075–1090 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Sharma, B. et al. Alternative progenitor cells compensate to rebuild the coronary vasculature in Elabela- and Apj-deficient hearts. Dev. Cell 42, 655–666.e3 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Wu, B. et al. Endocardial cells form the coronary arteries by angiogenesis through myocardial-endocardial VEGF signaling. Cell 151, 1083–1096 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Zhang, Z. & Zhou, B. Accelerated coronary angiogenesis by vegfr1-knockout endocardial cells. PLOS One 8, e70570 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Zhang, H. et al. Endocardium minimally contributes to coronary endothelium in the embryonic ventricular free walls. Circ. Res. 118, 1880–1893 (2016).

    CAS  PubMed  Google Scholar 

  160. Cano, E. et al. Extracardiac septum transversum/proepicardial endothelial cells pattern embryonic coronary arterio-venous connections. Proc. Natl Acad. Sci. USA 113, 656–661 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Klotz, L. et al. Cardiac lymphatics are heterogeneous in origin and respond to injury. Nature 522, 62–67 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Ulvmar, M. H., Martinez-Corral, I., Stanczuk, L. & Makinen, T. Pdgfrb-Cre targets lymphatic endothelial cells of both venous and non-venous origins. Genesis 54, 350–358 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Bernanke, D. H. & Markwald, R. R. Migratory behavior of cardiac cushion tissue cells in a collagen-lattice culture system. Dev. Biol. 91, 235–245 (1982).

    CAS  PubMed  Google Scholar 

  164. Markwald, R. R., Fitzharris, T. P. & Manasek, F. J. Structural development of endocardial cushions. Am. J. Anat. 148, 85–119 (1977).

    CAS  PubMed  Google Scholar 

  165. Mjaatvedt, C. H., Lepera, R. C. & Markwald, R. R. Myocardial specificity for initiating endothelial-mesenchymal cell transition in embryonic chick heart correlates with a particulate distribution of fibronectin. Dev. Biol. 119, 59–67 (1987).

    CAS  PubMed  Google Scholar 

  166. Ma, L., Lu, M. F., Schwartz, R. J. & Martin, J. F. Bmp2 is essential for cardiac cushion epithelial-mesenchymal transition and myocardial patterning. Development 132, 5601–5611 (2005).

    CAS  PubMed  Google Scholar 

  167. de Lange, F. J. et al. Lineage and morphogenetic analysis of the cardiac valves. Circ. Res. 95, 645–654 (2004).

    PubMed  Google Scholar 

  168. Waldo, K. et al. A novel role for cardiac neural crest in heart development. J. Clin. Invest. 103, 1499–1507 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Odelin, G. et al. Krox20 defines a subpopulation of cardiac neural crest cells contributing to arterial valves and bicuspid aortic valve. Development 145, https://doi.org/10.1242/dev.151944 (2018).

    Google Scholar 

  170. Lincoln, J., Alfieri, C. M. & Yutzey, K. E. Development of heart valve leaflets and supporting apparatus in chicken and mouse embryos. Dev. Dyn. 230, 239–250 (2004).

    CAS  PubMed  Google Scholar 

  171. Eley, L. et al. A novel source of arterial valve cells linked to bicuspid aortic valve without raphe in mice. Elife 7, e34110 (2018).

    PubMed  PubMed Central  Google Scholar 

  172. Mifflin, J. J., Dupuis, L. E., Alcala, N. E., Russell, L. G. & Kern, C. B. Intercalated cushion cells within the cardiac outflow tract are derived from the myocardial troponin T type 2 (Tnnt2) Cre lineage. Dev. Dyn. 247, 1005–1017 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Ferdous, A. et al. Nkx2-5 transactivates the Ets-related protein 71 gene and specifies an endothelial/endocardial fate in the developing embryo. Proc. Natl Acad. Sci. USA 106, 814–819 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Rasmussen, T. L. et al. ER71 directs mesodermal fate decisions during embryogenesis. Development 138, 4801–4812 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. He, Z., Grunewald, M., Dor, Y. & Keshet, E. VEGF regulates relative allocation of Isl1(+) cardiac progenitors to myocardial and endocardial lineages. Mech. Dev. 142, 40–49 (2016).

    CAS  PubMed  Google Scholar 

  176. Rentschler, S. et al. Myocardial Notch signaling reprograms cardiomyocytes to a conduction-like phenotype. Circulation 126, 1058–1066 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Mellgren, A. M. et al. Platelet-derived growth factor receptor beta signaling is required for efficient epicardial cell migration and development of two distinct coronary vascular smooth muscle cell populations. Circ. Res. 103, 1393–1401 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Smith, C. L., Baek, S. T., Sung, C. Y. & Tallquist, M. D. Epicardial-derived cell epithelial-to-mesenchymal transition and fate specification require PDGF receptor signaling. Circ. Res. 108, e15–e26 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Braitsch, C. M., Combs, M. D., Quaggin, S. E. & Yutzey, K. E. Pod1/Tcf21 is regulated by retinoic acid signaling and inhibits differentiation of epicardium-derived cells into smooth muscle in the developing heart. Dev. Biol. 368, 345–357 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. DeLaughter, D. M. et al. Single-cell resolution of temporal gene expression during heart development. Dev. Cell 39, 480–490 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Li, G. et al. Transcriptomic profiling maps anatomically patterned subpopulations among single embryonic cardiac cells. Dev. Cell 39, 491–507 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Jia, G. et al. Single-cell transcriptional regulations and accessible chromatin landscape of cell fate decisions in early heart development. bioRxiv https://doi.org/10.1101/210930 (2017).

    Article  Google Scholar 

  183. Lescroart, F. et al. Defining the earliest step of cardiovascular lineage segregation by single-cell RNA-seq. Science 359, 1177–1181 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Kitajima, S., Takagi, A., Inoue, T. & Saga, Y. MesP1 and MesP2 are essential for the development of cardiac mesoderm. Development 127, 3215–3226 (2000).

    CAS  PubMed  Google Scholar 

  185. Chiapparo, G. et al. Mesp1 controls the speed, polarity, and directionality of cardiovascular progenitor migration. J. Cell Biol. 213, 463–477 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Lindsley, R. C. et al. Mesp1 coordinately regulates cardiovascular fate restriction and epithelial-mesenchymal transition in differentiating ESCs. Cell Stem Cell 3, 55–68 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Bondue, A. et al. Mesp1 acts as a master regulator of multipotent cardiovascular progenitor specification. Cell Stem Cell 3, 69–84 (2008).

    CAS  PubMed  Google Scholar 

  188. Bondue, A. et al. Defining the earliest step of cardiovascular progenitor specification during embryonic stem cell differentiation. J. Cell Biol. 192, 751–765 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. David, R. et al. MesP1 drives vertebrate cardiovascular differentiation through Dkk-1-mediated blockade of Wnt-signalling. Nat. Cell Biol. 10, 338–345 (2008).

    CAS  PubMed  Google Scholar 

  190. Downs, K. M. & Davies, T. Staging of gastrulating mouse embryos by morphological landmarks in the dissecting microscope. Development 118, 1255–1266 (1993).

    CAS  PubMed  Google Scholar 

  191. Spanjaard, B. & Junker, J. P. Methods for lineage tracing on the organism-wide level. Curr. Opin. Cell Biol. 49, 16–21 (2017).

    CAS  PubMed  Google Scholar 

  192. Meilhac, S. M., Lescroart, F., Blanpain, C. & Buckingham, M. E. Cardiac cell lineages that form the heart. Cold Spring Harb Perspect Med. 4, a013888 (2014).

    PubMed  PubMed Central  Google Scholar 

  193. Moore-Morris, T., Cattaneo, P., Puceat, M. & Evans, S. M. Origins of cardiac fibroblasts. J. Mol. Cell. Cardiol. 91, 1–5 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work in the group of S.M.M. is supported by the Institut Imagine, the Institut Pasteur, INSERM and the Université Paris Descartes. M.E.B. acknowledges support from the Institut Pasteur and the Centre National de la Recherche Scientifique (CNRS; UMR 3738). The authors thank R. Kelly (Institut de Biologie du Développement de Marseille, France) and S. Zaffran and F. Lescroart (Marseille Medical Genetics, France) for helpful comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Both authors researched data for the article, discussed its content, wrote the manuscript and reviewed and edited it before submission.

Corresponding authors

Correspondence to Sigolène M. Meilhac or Margaret E. Buckingham.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meilhac, S.M., Buckingham, M.E. The deployment of cell lineages that form the mammalian heart. Nat Rev Cardiol 15, 705–724 (2018). https://doi.org/10.1038/s41569-018-0086-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41569-018-0086-9

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