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:

Mesp1 controls the chromatin and enhancer landscapes essential for spatiotemporal patterning of early cardiovascular progenitors

Abstract

The mammalian heart arises from various populations of Mesp1-expressing cardiovascular progenitors (CPs) that are specified during the early stages of gastrulation. Mesp1 is a transcription factor that acts as a master regulator of CP specification and differentiation. However, how Mesp1 regulates the chromatin landscape of nascent mesodermal cells to define the temporal and spatial patterning of the distinct populations of CPs remains unknown. Here, by combining ChIP–seq, RNA-seq and ATAC-seq during mouse pluripotent stem cell differentiation, we defined the dynamic remodelling of the chromatin landscape mediated by Mesp1. We identified different enhancers that are temporally regulated to erase the pluripotent state and specify the pools of CPs that mediate heart development. We identified Zic2 and Zic3 as essential cofactors that act with Mesp1 to regulate its transcription-factor activity at key mesodermal enhancers, thereby regulating the chromatin remodelling and gene expression associated with the specification of the different populations of CPs in vivo. Our study identifies the dynamics of the chromatin landscape and enhancer remodelling associated with temporal patterning of early mesodermal cells into the distinct populations of CPs that mediate heart development.

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: Dynamics of gene expression regulated by Mesp1.
Fig. 2: Temporal dynamic of chromatin remodelling regulated by Mesp1.
Fig. 3: Characterization of Mesp1-bound enhancers and prediction of putative transcriptional cofactors.
Fig. 4: Validation of Mesp1 target genes and enhancer remodelling in the presence of endogenous Mesp1.
Fig. 5: Zic2 and Zic3 bind to a fraction of Mesp1-bound enhancers.
Fig. 6: Mesp1 and Zic3 co-regulate gene expression during mouse gastrulation.
Fig. 7: Zic3 and Zic2 are essential regulators of Mesp1 activity.

Similar content being viewed by others

Data availability

The next-generation-sequencing data (ChIP–seq, ATAC-seq and RNA-seq) generated during this study has been deposited in Gene Expression Omnibus (GEO) and is accessible through GEO Series accession number GSE165107. Previously published next-generation-sequencing data and microarray data that were re-analysed here are available under the accession codes GSE41361, GSE44288 and GSE59033. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding authors on reasonable request.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Harvey, R. P. Patterning the vertebrate heart. Nat. Rev. Genet. 3, 544–556 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Meilhac, S. M. & Buckingham, M. E. The deployment of cell lineages that form the mammalian heart. Nat. Rev. Cardiol. 15, 705–724 (2018).

    Article  PubMed  Google Scholar 

  4. Protze, S. I., Lee, J. H. & Keller, G. M. Human pluripotent stem cell-derived cardiovascular cells: from developmental biology to therapeutic applications. Cell Stem Cell 25, 311–327 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  6. Srivastava, D. Making or breaking the heart: from lineage determination to morphogenesis. Cell 126, 1037–1048 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Bruneau, B. G. The developmental genetics of congenital heart disease. Nature 451, 943–948 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. 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).

    Article  CAS  PubMed  Google Scholar 

  9. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. 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 

  15. 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 

  16. Chan, S. S. et al. Mesp1 patterns mesoderm into cardiac, hematopoietic, or skeletal myogenic progenitors in a context-dependent manner. Cell Stem Cell 12, 587–601 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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).

  18. Zhang, Q. et al. Unveiling complexity and multipotentiality of early heart fields. Circ. Res. 129, 474–487 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Ivanovitch, K. et al. Ventricular, atrial and outflow tract heart progenitors arise from spatially and molecularly distinct regions of the primitive streak. Preprint at bioRxiv https://doi.org/2020.2007.2012.198994 (2020).

  20. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Shlyueva, D., Stampfel, G. & Stark, A. Transcriptional enhancers: from properties to genome-wide predictions. Nat. Rev. Genet. 15, 272–286 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wapinski, O. L. et al. Hierarchical mechanisms for direct reprogramming of fibroblasts to neurons. Cell 155, 621–635 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Soufi, A. et al. Pioneer transcription factors target partial DNA motifs on nucleosomes to initiate reprogramming. Cell 161, 555–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Paige, S. L. et al. A temporal chromatin signature in human embryonic stem cells identifies regulators of cardiac development. Cell 151, 221–232 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wamstad, J. A. et al. Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 151, 206–220 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Pikkarainen, S., Tokola, H., Kerkela, R. & Ruskoaho, H. GATA transcription factors in the developing and adult heart. Cardiovasc. Res. 63, 196–207 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. George, R. M. & Firulli, A. B. Hand factors in cardiac development. Anat. Rec. 302, 101–107 (2019).

    Article  Google Scholar 

  29. Greulich, F., Rudat, C. & Kispert, A. Mechanisms of T-box gene function in the developing heart. Cardiovasc. Res. 91, 212–222 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Kume, T. Novel insights into the differential functions of Notch ligands in vascular formation. J. Angiogenes. Res. 1, 8 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Oda, M. et al. DNA methylation restricts lineage-specific functions of transcription factor Gata4 during embryonic stem cell differentiation. PLoS Genet. 9, e1003574 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. Kattman, S. J. et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell 8, 228–240 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gebbia, M. et al. X-linked situs abnormalities result from mutations in ZIC3. Nat. Genet. 17, 305–308 (1997).

    Article  CAS  PubMed  Google Scholar 

  37. Ware, S. M. et al. Identification and functional analysis of ZIC3 mutations in heterotaxy and related congenital heart defects. Am. J. Hum. Genet. 74, 93–105 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Cowan, J., Tariq, M. & Ware, S. M. Genetic and functional analyses of ZIC3 variants in congenital heart disease. Hum. Mutat. 35, 66–75 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Purandare, S. M. et al. A complex syndrome of left-right axis, central nervous system and axial skeleton defects in Zic3 mutant mice. Development 129, 2293–2302 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Ware, S. M., Harutyunyan, K. G. & Belmont, J. W. Zic3 is critical for early embryonic patterning during gastrulation. Dev. Dyn. 235, 776–785 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Zhu, L. et al. Identification of a novel role of ZIC3 in regulating cardiac development. Hum. Mol. Genet. 16, 1649–1660 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Elms, P. et al. Overlapping and distinct expression domains of Zic2 and Zic3 during mouse gastrulation. Gene Expr. Patterns 4, 505–511 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Xu, J. et al. Genome-wide CRISPR screen identifies ZIC2 as an essential gene that controls the cell fate of early mesodermal precursors to human heart progenitors. Stem Cells 38, 741–755 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lim, L. S., Hong, F. H., Kunarso, G. & Stanton, L. W. The pluripotency regulator Zic3 is a direct activator of the Nanog promoter in ESCs. Stem Cells 28, 1961–1969 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Lim, L. S. et al. Zic3 is required for maintenance of pluripotency in embryonic stem cells. Mol. Biol. Cell 18, 1348–1358 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Perl, E. & Waxman, J. S. Retinoic acid signaling and heart development. Subcell. Biochem. 95, 119–149 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. 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 

  48. Ryckebusch, L. et al. Retinoic acid deficiency alters second heart field formation. Proc. Natl Acad. Sci. USA 105, 2913–2918 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Deshwar, A. R., Chng, S. C., Ho, L., Reversade, B. & Scott, I. C. The Apelin receptor enhances Nodal/TGFbeta signaling to ensure proper cardiac development. eLife 5, e13758 (2016).

  50. Scott, I. C. et al. The G protein-coupled receptor agtrl1b regulates early development of myocardial progenitors. Dev. Cell 12, 403–413 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Zeng, X. X., Wilm, T. P., Sepich, D. S. & Solnica-Krezel, L. Apelin and its receptor control heart field formation during zebrafish gastrulation. Dev. Cell 12, 391–402 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Klootwijk, R. et al. A deletion encompassing Zic3 in bent tail, a mouse model for X-linked neural tube defects. Hum. Mol. Genet. 9, 1615–1622 (2000).

    Article  CAS  PubMed  Google Scholar 

  53. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Guibentif, C. et al. Diverse routes toward early somites in the mouse embryo. Dev. Cell 56, 141–153 e146 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zaret, K. S. Pioneer transcription factors initiating gene network changes. Annu. Rev. Genet. 54, 367–385 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zaret, K. S. Pioneering the chromatin landscape. Nat. Genet. 50, 167–169 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Zaret, K. S. & Carroll, J. S. Pioneer transcription factors: establishing competence for gene expression. Genes Dev. 25, 2227–2241 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Iwafuchi-Doi, M. & Zaret, K. S. Cell fate control by pioneer transcription factors. Development 143, 1833–1837 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Liu, Z. & Kraus, W. L. Catalytic-Independent functions of PARP-1 determine Sox2 pioneer activity at intractable genomic loci. Mol. Cell 65, 589–603 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Swinstead, E. E. et al. Steroid receptors reprogram FoxA1 occupancy through dynamic chromatin transitions. Cell 165, 593–605 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Donaghey, J. et al. Genetic determinants and epigenetic effects of pioneer-factor occupancy. Nat. Genet. 50, 250–258 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mayran, A. et al. Pioneer and nonpioneer factor cooperation drives lineage specific chromatin opening. Nat. Commun. 10, 3807 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Cernilogar, F. M. et al. Pre-marked chromatin and transcription factor co-binding shape the pioneering activity of Foxa2. Nucleic Acids Res. 47, 9069–9086 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sutherland, M. J., Wang, S., Quinn, M. E., Haaning, A. & Ware, S. M. Zic3 is required in the migrating primitive streak for node morphogenesis and left-right patterning. Hum. Mol. Genet. 22, 1913–1923 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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  PubMed  PubMed Central  Google Scholar 

  66. Tarbell, E. D. & Liu, T. HMMRATAC: a hidden Markov ModeleR for ATAC-seq. Nucleic Acids Res. 47, e91 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ran, F. A. et al. Double nicking by RNA-guided CRISPR-Cas9n for enhanced genome editing specificity. Cell 154, 1380–1389 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  70. Sambrook, J. & Russell, D. W. Identification of associated proteins by coimmunoprecipitation. CSH Protoc. 2006, pdb.prot3898 (2006).

  71. Roux, M., Laforest, B., Capecchi, M., Bertrand, N. & Zaffran, S. Hoxb1 regulates proliferation and differentiation of second heart field progenitors in pharyngeal mesoderm and genetically interacts with Hoxa1 during cardiac outflow tract development. Dev. Biol. 406, 247–258 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. de Soysa, T. Y. et al. Single-cell analysis of cardiogenesis reveals basis for organ-level developmental defects. Nature 572, 120–124 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  75. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Wickham, H. Ggplot2: Elegant Graphics for Data Analysis 2nd edn (Springer Publishing Company, Incorporated, 2009).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Shen, L., Shao, N., Liu, X. & Nestler, E. ngs.plot: Quick mining and visualization of next-generation sequencing data by integrating genomic databases. BMC Genomics 15, 284 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, Z. et al. Identification of transcription factor binding sites using ATAC-seq. Genome Biol. 20, 45 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the ULB animal facility and ULB genomic core facility (F. Libert and A. Lefort). We thank Y. Song for bioinformatic assistance. F.L., X.L. and B.S. were supported by an FNRS aspirant fellowship (B.S.), an EMBO long-term fellowship and the Leducq Foundation. S.G. was funded by a Royal Society Newton International Fellowship (grant no. NIF\R1\181950). C.G. is funded by the Swedish Research Council (grant no. 2017-06278). F.L. thanks the MMG imaging and the animal phenotyping core platforms. We thank R. Kelly for sharing antibodies. Work in the Göttgens laboratory is supported by grants from the Wellcome Trust, Blood Cancer UK, Cancer Research UK, NIDDK and core support grants from the Wellcome Trust to the Wellcome–MRC Cambridge Stem Cell Institute. Work in F.L.’s laboratory was supported by the INSERM ATIP-Avenir programme. C.B. is an investigator of WELBIO. Work in C.B.’s laboratory was supported by the FNRS, ULB foundation, European Research Council (ERC), foundation Bettencourt Schueller (C.B. and F.L.) and Leducq Foundation as part of the network ‘22q11.2 deletion syndrome: novel approaches to understand cardiopharyngeal pathogenesis’. C.B. and B.G. acknowledge support from the Fondation Philippe Wiener–Maurice Anspach.

Author information

Authors and Affiliations

Authors

Contributions

X.L., B.S., F.L. and C.B. designed the experiments, performed data analyses and wrote the manuscript. X.L. and B.S. performed most of the biological experiments. B.S. performed bioinformatic analyses of all sequencing data. C.D. performed FACS. E.P. and F.L. performed the immunofluorescence and RNAscope experiments on mouse embryos. Y.A. generated the Zic3-KO mouse lines. B.S., S.Z., E.B. and F.L. described and analysed the Zic3-mutant phenotypes. Chimeric embryos were generated by C.G., M.-L.N.T. and W.M., and processed and analysed by F.L. and B.G. Knockout cell lines for the chimeric embryos were generated by B.S. and C.G. C.P. provided technical support. E.C. and F.F. provided help with some of the next-generation sequencing. S.G. and J.C.M. performed the scRNA-seq analyses for the initial submission. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Fabienne Lescroart or Cédric Blanpain.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks José Luis de la Pompa and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Temporal regulation of gene expression mediated by homogeneous Mesp1 induction within embryoid bodies.

a, RNA-FISH on sections of EBs from Mesp1 Dox-inducible PSC lines in control conditions (NO DOX) or 24 h after doxycycline induction (+DOX) showing Mesp1 expression in red. (representative image of 6 independent embryonic bodies). b, Percentage of cells that are positive for Mesp1 using RNA in situ hybridization, in control (NO DOX) or upon Mesp1 overexpression (+DOX) (n = 6 for NO DOX and n = 7 for +DOX independent embryoid bodies). Error bars indicate SEM. Statistical analyses were performed by two-tail unpaired student t tests. p = 2.53×10−6 c, Level of Mesp1 expression in control (NO DOX) or doxycycline condition (+DOX) as measured by the signal intensity from the smRNA-FISH. Error bars indicate SEM. Statistical analyses were performed by two-tail unpaired student t tests. n = 30 representative Mesp1+ cells per condition. p = 6.87×10−10 d, Principal component analysis of RNA-seq samples performed at day 2.5 (0 h), day 3 (12 h following Mesp1 overexpression) and day 3.5 (24 h following Mesp1 overexpression) in control (no dox) and Mesp1 overexpression (dox) conditions during PSC differentiation. Note the excellent concordance between biological duplicates. e, Representative examples of genes that undergo early, constant or late downregulation mediated by Mesp1. Examples were chosen to represent the diversity of kinetics we could find in genes repressed by Mesp1. f, Plots representing all genes classified as early, constant or late downregulated, as well as their respective average profile (thick lines).

Source data

Extended Data Fig. 2 Temporal analysis of Mesp1 TF activity.

a, Pie chart representing the distribution of the position of Mesp1 ChIP-seq peaks relative to protein-coding genes (data shown represent two biologically independent replicates. b-c, Graph representing the dynamics of ATAC-seq (b) and H3K27Ac ChIP-seq (c) signal within early, constant and late Mesp1 ChIP-seq peaks. Average profiles are shown by the think red (dox) and blue (no dox) lines. d, Dot plot illustrating the correlation between temporality of Mesp1 binding and surrounding H3K27Ac deposition (left), or Mesp1 binding and ATAC-seq opening of the chromatin (right). For each individual Mesp1 ChIP-seq peak, the average signal was measured for all three types of experiments at 12 h and 24 h dox, in order to extract a measure of fold-change between 12 and 24 h. The log2 value of this fold-change for each peak was compared between each type of experiment. Data shown represent two biologically independent replicates; p-values were calculated through a two-tailored t-test.

Extended Data Fig. 3 Motif discovery of enhancers activated by Mesp1.

a, Motif discovery performed separately in Mesp1 ChIP-seq peaks classified as early, constant or late peaks. p-values are calculated through a binomial test. b, Same pipeline as panel a but separating peaks as de novo versus primed Mesp1 peaks. Stars represent the enrichment of the CAAATGG motif in pioneer peaks in comparison to non-pioneer peaks through a two-tailored Z-test. Data shown represent two biologically independent replicates. c, Quantification of the occurrence of all forms of bHLH motifs in Mesp1 ChIP-seq peaks, showing the prevalence of CAAATG motif, most often with an extra G. *** The top 2 bHLH motifs were notably over-represented (p < 0.00001) through a two-tailored Z-test, with z = 17.5 for CAAATG and z = 5.9 for CAGATG. d, Representative genomic locus where ATAC-seq and H3K27Ac ChIP-seq peaks are upregulated 24 h following Mesp1 expression but which are not directly bound by Mesp1 (red boxes), suggesting that their regulation is mediated by other TFs, whose expression is induced directly or indirectly by Mesp1. Gata4 ChIP-seq data31 (green) shows a strong overlap between these de novo opened peaks not bound but induced by Mesp1. e, Motif discovery searching for known TF binding sites within ATAC-seq peaks that get opened by Mesp1 but are not directly bound by Mesp1. p-values are calculated through a binomial test. f, Quantification of peaks containing a Gata4 motif, both in Mesp1 ChIP-seq peaks and ATAC-seq peaks UP without Mesp1 binding, separated as peaks bound by Gata4 or not bound by Gata4 using a published ChIP-seq dataset31. *** Gata4 motifs are statistically notably enriched in Gata4-binding peaks in Mesp1 ChIP-seq (z = 8.34, p < 0.00001) and in ATAC-seq peaks UP (z = 13.6, p < 0.00001). Data shown represent two biologically independent replicates; n = 1 for previously published Gata4 ChIP-seq31. These values were calculated through a two-tailored Z-test.

Extended Data Fig. 4 Repression of the core pluripotency network by Mesp1.

a, Representative genomic locus (Cdh1) where ATAC-seq and H3K27Ac ChIP peaks found in control (no dox) conditions are absent or smaller in dox conditions, without presenting any Mesp1 binding (blue boxes), suggesting indirect repression of chromatin opening by other factors. b, Motif discovery within peaks that are closed in dox conditions, including enrichment of a compound OCT-SOX-TCF-NANOG motif. p-values are calculated through a binomial test. c, Heatmap showing signal of ATAC-seq, H3K27Ac ChIP-seq and published Nanog, Oct4 and Sox2 ChIP-seq data within peaks that were closed after dox-induced Mesp1 overexpression32.

Extended Data Fig. 5 Context-dependency of Mesp1 activator potential.

a, Heatmap of the expression values of Mesp1 upregulated genes in undifferentiated PSCs (2i conditions), with or without Mesp1 induction (dox). RNA-seq samples in 2i were performed twice. b, Overlap between genes directly and indirectly activated by Mesp1 during PSC differentiation or in 2i, illustrating the paucity of Mesp1-mediated gene activation in pluripotency. c, Heatmap illustrating Mesp1 binding affinity to its enhancers in 2i by Mesp1 ChIP-seq and the subsequent lack of chromatin opening by ATAC-seq in 2i conditions. Each row represents a Mesp1 binding site detected during differentiation at 24 h dox. Peaks were ordered by unsupervised k-means clustering. 1, de novo peaks where Mesp1 binding and subsequent chromatin opening is lost in 2i; 2 and 2’, primed peaks with conserved Mesp1 binding and chromatin opening in 2i; 3, de novo ATAC-seq peaks where Mesp1 binding and chromatin opening is conserved in 2i. All samples collected in 2i were performed twice. d, Representative examples of Mesp1 binding loci in 2i conditions. e-f, Classification of Mesp1 ChIP-seq peaks found in 2i into previously detailed chromatin opening (e) or kinetic (f) groups. *** Late peaks were notably depleted in 2i conditions (z = -7.86, p < 0.00001). Data shown represent two biologically independent replicates. These values were calculated through a two-tailored Z-test. g, Quantification of the correlation between Mesp1 binding strength measured by Mesp1 ChIP-seq and chromatin opening in ATAC-seq in 2i (24 h dox), demonstrating a linear correlation between these two variables. Data shown represent two biologically independent replicates.

Extended Data Fig. 6 Zic2 and Zic3 cooperate with Mesp1 and potentially with other mesoderm-inducing TFs.

a, Western blot illustrating the expression of Zic2 and Zic3 with and without Mesp1 induction, at day 3.5 (24 h) of PSC differentiation, as well as the lack of Zic2 and Zic3 protein expression in Zic2/3 double KO cell lines. (Data shown represent 2 independent experiments) b, Illustration of the two most enriched motifs in all Zic2 and Zic3 ChIP-seq detected peaks. p-values are calculated through a binomial test. c, Venn diagram illustrating the number of overlapping peaks between Mesp1, Zic2 and Zic3 ChIP-seq datasets. P-value was calculated using a hypergeometric test, using bedtools fisher. d, Illustration of the temporality of Zic3 binding within primed and de novo Mesp1-bound peaks, with associated ATAC-seq signal. 1, peaks already bound by Zic3 at day 2.5 (0 h); 2, peaks bound by Zic3 at 24 h no dox; 3, peaks bound by Zic3 at 24 h dox.

Source data

Extended Data Fig. 7 Zic2 and Zic3 regulate Mesp1-induced CP specification and differentiation.

a, mRNA expression of the core pluripotency associated TFs in Mesp1-inducible WT, Zic2 and Zic3 KO PSCs in Lif/2i pluripotency conditions, as measured by RT-qPCR. (n = 4 biologically independent replicates covering two independent KO clones with each assessed by two independent experiments. Error bars indicate mean + /-SEM, statical analysis was performed by 2-way ANOVA. b, Representative immunofluorescence for Troponin T in Mesp1-inducible WT, Zic2 and Zic3 KO cell lines at day 10 of differentiation, illustrating the ability of Mesp1 overexpression to overcome cardiac differentiation defects in Zic2 and Zic3 KO cell lines. (Data shown represent 6 independent experiments. Scale bars=100 µm. c, Illustrative examples of genes that are notably upregulated in Zic3 KO cells in comparison to WT cells, including known important factors of pluripotency and endoderm differentiation.

Source data

Extended Data Fig. 8 Heart defects observed in the newly generated Zic3-KO line.

a, Zygote injection strategy used to generate Zic3 KO mice. b, Pictures of Wild type (WT) and homozygous null (Zic3KO) E14.5 embryos, showing severe neural tube closure defects and exencephaly found in a subset of Zic3 KO embryos (n = 10/32). Scale bars= 1 mm. c, Range of cardiac morphological abnormalities found in Zic3 KO embryos at E14.5. We observe outflow tract defects with persistent truncus arteriosus (PTA bottom left panel – n = 1/32), hypoplasia of the right ventricle (RV) (upper right panel – n = 5/32) and mutants with a situs inversus phenotype (bottom right panel – n = 2/32). Scale bars= 1 mm. d, ratio of the surface area of the right ventricle compared to the surface area of the left ventricle in wild type (black – n = 26) and Zic3 KO (red – n = 44). Error bars indicate mean + /-SEM. Unpaired, two-tailed t-test showed a p-value=0.0004. e, Mean thickness of the compact myocardial (CM) layer of the ventricles in wild type (black – n = 6) and Zic3 KO embryos (red – n = 12). Error bars indicate mean + /-SEM. Unpaired, two-tailed t-test showed a p-value=0.0074. f-f’, Immunofluorescence on E14.5 wild type (f) and Zic3 KO (f’) hearts using an anti-cardiac Troponin T (cTnT) antibody to label the cardiomyocytes and isolectinB4 to label the endocardium (representative pictures from 4 independent hearts of each genotype). No endocardial defect was observed in Zic3 KO embryos while the cTnT+ layer was thinner in f’. Scale bars= 200 µm. RV, right ventricle; LV, left ventricle; RA, right atrium; LA, left atrium; pt, pulmonary trunk; ao, aorta.

Source data

Extended Data Fig. 9 Zic2 and Zic3 redundantly regulate Mesp1 activity.

a, Expression of three core pluripotency genes in Mesp1 WT and two independent Zic2/3 KO PSC cell lines cultured in Lif/2i medium. Data from two independent experiments, b, FACS profiles of EBs at day 4 of differentiation from Mesp1 WT and Zic2/3 dKO cell lines, illustrating the decrease in Flk1 and PDGFRa expression in Zic2/3 dKO cell lines both in no dox and dox conditions. c, Table shows the distribution of genes that were downregulated in Zic3 KO and Zic2/3 dKO cells within the temporal categories of Mesp1 direct upregulated target genes. There was no particular enrichment for early, constant and late genes within the Zic2/3-dependent fraction of Mesp1 target. d, Barplot illustrating the proportion of early, constant and late Mesp1 binding sites within Mesp1 ChIP-seq peaks conserved in Zic2/3 dKO cell lines. *** for late genes, z = -5.845, p < 0.00001. These values were calculated through a two-tailored Z-test. Data shown represent two biologically independent replicates. e, Representation of the proportion of Mesp1 ChIP-seq peaks as well as ATAC-seq peaks that are opened (UP) or closed (DOWN- upon Mesp1 induction in WT cells which are preferentially closed in Zic2/3 dKO cells after Mesp1 induction. n = 2 independent experiments for Mesp1 ChIP-seq and ATAC-seq in WT cells; n = 3 independent experiments for ATAC-seq in Zic2/3 dKO cell lines. *** all three comparisons were significant with p < 0.00001 and respectively z = 34.9 (Mesp1 ChIP-seq), z = 54.2 (ATAC-seq UP) and z = -20.1 (ATAC-seq DOWN). These values were calculated through a two-tailored Z-test. f, Motif enrichment analysis of ATAC-seq peaks that were preferentially closed in Zic2/3 dKO cells in comparison to WT cells. p-values are calculated through a binomial test.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Supplementary Table 1. Guide RNA sequences used for CRISPR–Cas9n gene and enhancer KO as well as enhancer-motif editing. Supplementary Table 2. PCR primers used for screening of the KO cell lines. Supplementary Table 3. Sequences of qPCR primers used. Supplementary Table 4. List of primary antibodies used. Supplementary Table 5. Probes used for smRNA-FISH.

Source data

Source Data Fig. 3

Statistical source data for Fig. 3b,e,g.

Source Data Fig. 5

Unprocessed western blots for Fig. 5f.

Source Data Fig. 5

Statistical source data for Fig. 5a,b.

Source Data Fig. 6

Statistical source data for Fig. 6a,b.

Source Data Fig. 7

Statistical source data for Fig. 7a,b,k.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1b,c.

Source Data Extended Data Fig. 6

Unprocessed western blots for Extended Data Fig. 6a.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7a.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8d,e.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9a.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, X., Swedlund, B., Ton, ML.N. et al. Mesp1 controls the chromatin and enhancer landscapes essential for spatiotemporal patterning of early cardiovascular progenitors. Nat Cell Biol 24, 1114–1128 (2022). https://doi.org/10.1038/s41556-022-00947-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00947-3

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