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:

Early lineage restriction in temporally distinct populations of Mesp1 progenitors during mammalian heart development

Abstract

Cardiac development arises from two sources of mesoderm progenitors, the first heart field (FHF) and the second (SHF). Mesp1 has been proposed to mark the most primitive multipotent cardiac progenitors common for both heart fields. Here, using clonal analysis of the earliest prospective cardiovascular progenitors in a temporally controlled manner during early gastrulation, we found that Mesp1 progenitors consist of two temporally distinct pools of progenitors restricted to either the FHF or the SHF. FHF progenitors were unipotent, whereas SHF progenitors were either unipotent or bipotent. Microarray and single-cell PCR with reverse transcription analysis of Mesp1 progenitors revealed the existence of molecularly distinct populations of Mesp1 progenitors, consistent with their lineage and regional contribution. Together, these results provide evidence that heart development arises from distinct populations of unipotent and bipotent cardiac progenitors that independently express Mesp1 at different time points during their specification, revealing that the regional segregation and lineage restriction of cardiac progenitors occur very early during gastrulation.

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: Mesp1–rtTA transgenic mice faithfully recapitulate Mesp1 endogenous expression.
Figure 2: Two temporally distinct populations of Mesp1 progenitors contribute to the development of the FHF and SHF.
Figure 3: Bio-statistical modelling of the the multicolour-labelled hearts.
Figure 4: Clonal analysis of lineage differentiation of Mesp1-derived progenitors in vivo.
Figure 5: Molecular signature of early and late Mesp1-expressing cells in vivo.
Figure 6: Different temporal expression of Mesp1 direct target genes.
Figure 7: Revised model of the early step of cardiovascular progenitor specification and lineage commitment during mouse development.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Garry, D. J. & Olson, E. N. A common progenitor at the heart of development. Cell 127, 1101–1104 (2006).

    Article  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).

    Article  CAS  PubMed  Google Scholar 

  3. 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).

    Article  CAS  PubMed  Google Scholar 

  4. 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 

  5. Bondue, A. & Blanpain, C. Mesp1: a key regulator of cardiovascular lineage commitment. Circ. Res. 107, 1414–1427 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Cohen-Gould, L. & Mikawa, T. The fate diversity of mesodermal cells within the heart field during chicken early embryogenesis. Dev. Biol. 177, 265–273 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Wei, Y. & Mikawa, T. Fate diversity of primitive streak cells during heart field formation in ovo. Dev. Dyn. 219, 505–513 (2000).

    Article  CAS  PubMed  Google Scholar 

  13. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. 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).

    Article  CAS  PubMed  Google Scholar 

  16. Haraguchi, S. et al. Transcriptional regulation of Mesp1 and Mesp2 genes: differential usage of enhancers during development. Mech. Dev. 108, 59–69 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Saga, Y. et al. MesP1: a novel basic helix-loop-helix protein expressed in the nascent mesodermal cells during mouse gastrulation. Development 122, 2769–2778 (1996).

    CAS  PubMed  Google Scholar 

  18. Kitajima, S., Miyagawa-Tomita, S., Inoue, T., Kanno, J. & Saga, Y. Mesp1-nonexpressing cells contribute to the ventricular cardiac conduction system. Dev. Dyn. 235, 395–402 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. 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).

    Article  CAS  PubMed  Google Scholar 

  22. Harel, I. et al. Distinct origins and genetic programs of head muscle satellite cells. Dev. Cell 16, 822–832 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sambasivan, R. et al. Distinct regulatory cascades govern extraocular and pharyngeal arch muscle progenitor cell fates. Dev. Cell 16, 810–821 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. 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).

    Article  CAS  PubMed  Google Scholar 

  25. Harris, I. S. & Black, B. L. Development of the endocardium. Pediatr. Cardiol. 31, 391–399 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  26. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  30. Milgrom-Hoffman, M. et al. The heart endocardium is derived from vascular endothelial progenitors. Development 138, 4777–4787 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Riley, P. R. & Smart, N. Vascularizing the heart. Cardiovasc. Res. 91, 260–268 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Van den Ameele, J. et al. Eomesodermin induces Mesp1 expression and cardiac differentiation from embryonic stem cells in the absence of Activin. EMBO Rep. 13, 355–362 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Costello, I. et al. The T-box transcription factor Eomesodermin acts upstream of Mesp1 to specify cardiac mesoderm during mouse gastrulation. Nat. Cell Biol. 13, 1084–1091 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Christiaen, L. et al. The transcription/migration interface in heart precursors of Ciona intestinalis. Science 320, 1349–1352 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Lossie, A. C., Nakamura, H., Thomas, S. E. & Justice, M. J. Mutation of l7Rn3 shows that Odz4 is required for mouse gastrulation. Genetics 169, 285–299 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nakamura, H., Cook, R. N. & Justice, M. J. Mouse Tenm4 is required for mesoderm induction. BMC Dev. Biol. 13, 9 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cruz, C. et al. Induction and patterning of trunk and tail neural ectoderm by the homeobox gene eve1 in zebrafish embryos. Proc. Natl Acad. Sci. USA 107, 3564–3569 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hart, A. H. et al. Mixl1 is required for axial mesendoderm morphogenesis and patterning in the murine embryo. Development 129, 3597–3608 (2002).

    CAS  PubMed  Google Scholar 

  42. Acampora, D. et al. OTX1 compensates for OTX2 requirement in regionalisation of anterior neuroectoderm. Gene. Expr. Patterns 3, 497–501 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Dush, M. K. & Martin, G. R. Analysis of mouse Evx genes: Evx-1 displays graded expression in the primitive streak. Dev. Biol. 151, 273–287 (1992).

    Article  CAS  PubMed  Google Scholar 

  44. Tsang, T. E. et al. Lim1 activity is required for intermediate mesoderm differentiation in the mouse embryo. Dev. Biol. 223, 77–90 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Niederreither, K. et al. Embryonic retinoic acid synthesis is essential for heart morphogenesis in the mouse. Development 128, 1019–1031 (2001).

    CAS  PubMed  Google Scholar 

  46. Sucov, H. M. et al. RXR alpha mutant mice establish a genetic basis for vitamin A signaling in heart morphogenesis. Genes Dev. 8, 1007–1018 (1994).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lickert, H. et al. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature 432, 107–112 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Kume, T., Jiang, H., Topczewska, J. M. & Hogan, B. L. The murine winged helix transcription factors, Foxc1 and Foxc2, are both required for cardiovascular development and somitogenesis. Genes Dev. 15, 2470–2482 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dunwoodie, S. L., Rodriguez, T. A. & Beddington, R. S. Msg1 and Mrg1, founding members of a gene family, show distinct patterns of gene expression during mouse embryogenesis. Mech. Dev. 72, 27–40 (1998).

    Article  CAS  PubMed  Google Scholar 

  52. Nomura-Kitabayashi, A. et al. Hypomorphic Mesp allele distinguishes establishment of rostrocaudal polarity and segment border formation in somitogenesis. Development 129, 2473–2481 (2002).

    CAS  PubMed  Google Scholar 

  53. 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).

    Article  CAS  PubMed  Google Scholar 

  54. Kubalak, S. W., Miller-Hance, W. C., O’Brien, T. X., Dyson, E. & Chien, K. R. Chamber specification of atrial myosin light chain-2 expression precedes septation during murine cardiogenesis. J. Biol. Chem. 269, 16961–16970 (1994).

    CAS  PubMed  Google Scholar 

  55. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kataoka, H. et al. Etv2/ER71 induces vascular mesoderm from Flk1+PDGFRα+ primitive mesoderm. Blood 118, 6975–6986 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Palencia-Desai, S. et al. Vascular endothelial and endocardial progenitors differentiate as cardiomyocytes in the absence of Etsrp/Etv2 function. Development 138, 4721–4732 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  60. Perl, A. K., Wert, S. E., Nagy, A., Lobe, C. G. & Whitsett, J. A. Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc. Natl Acad. Sci. USA 99, 10482–10487 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. 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).

    Article  CAS  PubMed  Google Scholar 

  62. Tumbar, T. et al. Defining the epithelial stem cell niche in skin. Science 303, 359–363 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Piette, D., Hendrickx, M., Willems, E., Kemp, C. R. & Leyns, L. An optimized procedure for whole-mount in situ hybridization on mouse embryos and embryoid bodies. Nat. Protoc. 3, 1194–1201 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Zinyk, D. L., Mercer, E. H., Harris, E., Anderson, D. J. & Joyner, A. L. Fate mapping of the mouse midbrain-hindbrain constriction using a site-specific recombination system. Curr. Biol. 8, 665–668 (1998).

    Article  CAS  PubMed  Google Scholar 

  65. Gonzalez-Roca, E. et al. Accurate expression profiling of very small cell populations. PLoS ONE 5, e14418 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Sasagawa, Y. et al. Quartz-Seq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity. Genome Biol. 14, R31 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Jensen, K. B. & Watt, F. M. Single-cell expression profiling of human epidermal stem and transit-amplifying cells: Lrig1 is a regulator of stem cell quiescence. Proc. Natl Acad. Sci. USA 103, 11958–11963 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Tan, D. W. et al. Single-cell gene expression profiling reveals functional heterogeneity of undifferentiated human epidermal cells. Development 140, 1433–1444 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank F. Bollet-Quivogne and J-M. Vanderwinden for their help with confocal imaging and M. Tarabichi for his help with gene set enrichment analysis. We thank M. Buckingham and A. Joyner for kindly providing the probes for in situ hybridization. We also thank the Genomic core facility of the EMBL, Heidelberg for their help with the ChIP-Seq. F.L. has been sequentially supported by the FNRS and the EMBO long-term fellowship. S.C. is supported by a fellowship of the FRS/FRIA. X.L. is supported by the FNRS. A.B. is supported by the FRS/FNRS. S.R. and B.D.S. are supported by the Wellcome Trust (grant number 098357/Z/12/Z). C.B. is an investigator of WELBIO. This work was supported by the FNRS, the ULB foundation, the Fondation contre le Cancer, the European Research Council (ERC), and the foundation Bettencourt Schueller (C.B. and F.L.).

Author information

Authors and Affiliations

Authors

Contributions

C.B., F.L., S.C. and X.L. designed the experiments and performed data analysis. F.L. and S.C. performed most of the experiments. X.L. performed the single-cell PCR analysis. Y.A. generated the Mesp1–rtTA transgenic mice. A.R. and H. A. performed microarrays. C.P. provided technical assistance. C.D. helped with FACS isolation of Mesp1-expressing cells. A.B. helped in the design and initial characterization of the Mesp1–rtTA transgene. B.D.S. and S.R. performed the bio-statistical analysis of the clonal fate data. C.B. and F.L. wrote the manuscript.

Corresponding author

Correspondence to Cédric Blanpain.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Mesp1-rtTA/tetO-Cre transgenic mice induced Cre expression similarly to Mesp1-Cre Knock-in mice.

(a,b) Sections of E12.5 Mesp1-Cre/Rosa-tdTomato (a) and Mesp1-rtTA/TetO-Cre/Rosa-tdTomato hearts (induced by Dox administration between E6.25 and E7.5) (b) and co-stained with DAPI. Both transgenic hearts have a similar expression of the tdTomato with a negative region in the OFT that derive from Mesp1 negative neural crest cells (asterisks). ce Doxycycline injection has no effect on Mesp1 expression during early mouse embryonic development. (cd) In situ hybridization for Mesp1 expression in early embryo at E6.5. The detection of Mesp1 mRNA in the primitive streak (PS) and the nascent lateral mesoderm is similar in embryo that did not receive DOX (c) and in embryos injected with DOX (+ DOX) (d). A, anterior; P, posterior. e Expression of Mesp1 analysed by RT-qPCR in early embryos (E6.75) without (n = 9) or after doxycycline injections (n = 6). These data show no difference in Mesp1 expression after DOX injection. (fg) In situ hybridization for Cre expression in early embryos at E6.75. The detection of Cre mRNA in the primitive streak (PS) and the nascent lateral mesoderm is similar in Mesp1-Cre knock-in (f) and in Mesp1-rtTA/TetO-Cre transgenic embryos injected with DOX at E6.25 (+ DOX) (g). Cre expression is similar to the endogenous Mesp1 expression in wild type embryos (h). A, anterior; P, posterior. Scale bar, 500 μm.

Supplementary Figure 2 Temporal Dox administration in Mes1-rtTA/TetO-Cre/Rosa-Confetti embryos.

(a) While clonal dose of DOX (0.575 μg g−1) induces labelling in Mesp1-rtTA/TetO-Cre/Rosa-Confetti embryos at E6.25 (n = 53), at E6.75 (n = 118) or at E7.25 (n = 65), this dose was not sufficient to induce labelling at E5.75 (n = 13). A much higher dose of Dox (25 μg g−1) was required to produce labelling at a clonal density at E5.75 (n = 90). This 40 fold increase of DOX is likely to persist at a concentration sufficient to activate the Cre at the time of endogenous Mesp1 expression. This high dose of DOX never labelled any heart after administration at E8.5 or E9.5 (n = 24) supporting the absence of transgene expression after the end of endogenous Mesp1 expression. (b,c) Examples of Mesp1-rtTA/TetO-Cre/Rosa-Confetti unicolour labelled hearts at E12.5 induced at E5.75 after administration of high dose of Doxycycline (25 μg g−1). Note that each cluster is localized within the LV, FHF derivative and no labelling was detected other compartments. OFT, outflow tract; RV, right ventricle; LV, left ventricle; RA, right atrium; LA, left atrium; IFT, inflow tract. Scale bars, 200 μm. The number on the upper right in each panel refers to the ID of the labelled heart. (d) Quantification of the regional (FHF and SHF) contribution of patches of Mesp1 labelled cells in unicolour hearts induced at E5.75 with the high dose of Doxycycline (25 μg g−1), shows the exclusive labelling of the FHF (red) similarly to was found at E6.25 (Fig. 2m).

Supplementary Figure 3 Biostatistical modeling of the clonal fate data.

(a) The likelihood function F gives the probability of the experimental data for different values of the induction frequency pN and the fragmentation rate f. The numeric values have been rescaled such that the maximum of the likelihood function corresponds to 1. Colour denotes the value of F, such that red signifies a large value and blue signifies small value. Lines of equal values are indicated on the bottom of the figure. One sees that the maximum value of F is relatively featureless along a curve in the pN-f-plane. To infer the values of pN and f we must therefore refer to an independent measurement of one of the two parameters. (b) The multicolour labelling strategy allows us to independently infer the induction frequency pN = 1.3 by evaluating the abundances of hearts with a given number of colours. With this, we are left with a slice through the pN-f-plane and the fragmentation rate can be determined with a higher accuracy. (c) Monoclonal datasets (n = 89) identify two subpopulations in Mesp1 expressing cells: FHF progenitors, which contribute to the LV and SHF progenitors, which contribute to OFT and IFT. The plot shows the probabilities of monoclonal fragments in the different heart compartments. (d) Values for the induction frequency, pN, and the fragmentation rate, f, for the two FHF (n = 188) and SHF (n = 102) precursors. While the overall induction frequency is higher for FHF precursors, which we attribute to highest expression of Mesp1 at the early time points, the fragmentation rate is higher for SHF precursors. (e) We may use the distribution of monoclonal fragments (c) to predict the distribution of fragments in all hearts (n = 263). We find an excellent agreement with the notable exception of the RV, which might suggest the existence of an independent pool of progenitors contributing to RV morphogenesis. Error bars indicate one sigma (c and e) or 95% (d) confidence intervals.

Supplementary Figure 4 Late Mesp1 progenitors also contribute to the head.

(aa’) Example of a Mesp1-rtTA/TetO-Cre/Rosa-Confetti embryo with co-labelled head (a) and heart (a’) at E12.5. Scale bars, 200 μm. (b–b’) Sections of a Mesp1-rtTA/TetO-Cre/Rosa-Confetti labelled embryo showing labelling in the head in an extraocular muscle (EOM) (b) as well as in the right ventricle RV (b’). Scale bars, 200 μm. (c) Temporal appearance of head muscle labelling inferred from all datasets (n = 105 independent embryos translating to n = 181 embryos by colour). Plotted is the fraction of head muscle labelling induced at each induction time for a given colour. Head muscles are preferentially labelled at the late time points. (d) Regional contribution of head progenitors in monoclonal datasets (n = 5), showing the co-labelling of the head with the heart and preferentially SHF derivatives or the right ventricle (RV). OFT, outflow tract; RV, right ventricle; LV, left ventricle; RA, right atrium; LA, left atrium. Errors bars indicate one sigma confidence intervals.

Supplementary Figure 5 Cohesive versus dispersive mode of growth of the myocardium and the endocardium.

(ab) Sections of E12.5 Mesp1-rtTA/tetO-Cre/Rosa-Confetti unicolour hearts. (a) Example of a compact myocardial YFP-labelled clone showing cohesive growth of the myocardium. (b) Example of a dispersed endocardial RFP-unicolour clone showing dispersive mode of growth of the endocardium. Scale bars, 200 μm. The number on the upper right in each panel refers to the ID of the labelled heart.

Supplementary Figure 6 Semi-quantification of single cell RT-PCR analysis.

Examples showing strong, weak and no gene expression in single cells.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1990 kb)

Supplementary table 1

Supplementary Information (XLSX 64 kb)

Supplementary table 2

Supplementary Information (XLSX 51 kb)

Supplementary table 3

Supplementary Information (XLSX 14 kb)

Supplementary table 4

Supplementary Information (XLSX 40 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lescroart, F., Chabab, S., Lin, X. et al. Early lineage restriction in temporally distinct populations of Mesp1 progenitors during mammalian heart development. Nat Cell Biol 16, 829–840 (2014). https://doi.org/10.1038/ncb3024

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3024

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