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:

Human-gained heart enhancers are associated with species-specific cardiac attributes

An Author Correction to this article was published on 17 November 2022

This article has been updated

Abstract

Heart development is controlled by a relatively conserved network of transcriptional and chromatin regulators; how the human heart has evolved species-specific features to maintain adequate cardiac output and function remains to be defined. In this study, we performed a comparative epigenomic analysis of mouse and human cardiomyocytes at the earliest stages of cardiogenesis and identified enhancers and promoters that are specifically active in human cardiogenesis. These cis-regulatory elements (CREs) are associated with genes involved in heart development and function and are enriched in genetic variants associated with human cardiac phenotypic and disease traits, particularly those differing between humans and mice. Human-gained CREs are also gained within genomic loci of known transcriptional regulators, potentially expanding their role in human heart development. In particular, we found that a human-gained enhancer in the locus of the early developmental regulator ZIC3 regulates ZIC3 induction at the mesoderm stage as well as cardiomyocyte differentiation. Overall, our results illuminate how human-specific CREs can contribute to human-specific cardiac attributes and can expand the role of conserved transcriptional regulators in human cardiac 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: Dynamic epigenomic changes correlate with cardiac developmental gene expression programs.
Fig. 2: Comparative human and mouse epigenomic analysis reveals human-gained CREs that are associated with specific cardiac traits.
Fig. 3: Human-gained enhancers in the ZIC3 locus establish a potential ZIC3 mesoderm-related GRN that directs elevated human ZIC3 expression at the mesoderm stage.
Fig. 4: ZIC3 is required for human CM differentiation.
Fig. 5: A ZIC3 human-gained enhancer regulates mesoderm-specific ZIC3 expression and CM differentiation.

Similar content being viewed by others

Data availability

Sequencing data are available from the Gene Expression Omnibus database (snATAC: GSE192500; bulk sequencing data: GSE192365). A list of all the used sequencing datasets (new and public) and their accession numbers is available in Supplementary Table 13. Bulk sequencing data can be viewed using the UCSC Genome Browser (https://genome.ucsc.edu/s/Cbenner/Chi-211218-ReviewerTracks). All other data supporting the findings in this study are included in the main article and associated files.

Change history

References

  1. Long, H. K., Prescott, S. L. & Wysocka, J. Ever-changing landscapes: transcriptional enhancers in development and evolution. Cell 167, 1170–1187 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ren, B. & Yue, F. Transcriptional enhancers: bridging the genome and phenome. Cold Spring Harb. Symp. Quant. Biol. 80, 17–26 (2015).

    Article  PubMed  Google Scholar 

  3. Kathiriya, I. S., Nora, E. P. & Bruneau, B. G. Investigating the transcriptional control of cardiovascular development. Circ. Res. 116, 700–714 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Fahed, A. C., Gelb, B. D., Seidman, J. G. & Seidman, C. E. Genetics of congenital heart disease: the glass half empty. Circ. Res. 112, 707–720 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Homsy, J. et al. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science 350, 1262–1266 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Richter, F. et al. Genomic analyses implicate noncoding de novo variants in congenital heart disease. Nat. Genet. 52, 769–777 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Arvanitis, M. et al. Genome-wide association and multi-omic analyses reveal ACTN2 as a gene linked to heart failure. Nat. Commun. 11, 1122 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Hocker, J. D. et al. Cardiac cell type-specific gene regulatory programs and disease risk association. Sci. Adv. 7, eabf1444 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lahm, H. et al. Congenital heart disease risk loci identified by genome-wide association study in European patients. J. Clin. Invest. 131, e141837 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  11. Gilsbach, R. et al. Dynamic DNA methylation orchestrates cardiomyocyte development, maturation and disease. Nat. Commun. 5, 5288 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Gilsbach, R. et al. Distinct epigenetic programs regulate cardiac myocyte development and disease in the human heart in vivo. Nat. Commun. 9, 391 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Nothjunge, S. et al. DNA methylation signatures follow preformed chromatin compartments in cardiac myocytes. Nat. Commun. 8, 1667 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. 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 

  16. Zhao, M. T. et al. Cell type-specific chromatin signatures underline regulatory DNA elements in human induced pluripotent stem cells and somatic cells. Circ. Res. 121, 1237–1250 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Doevendans, P. A., Daemen, M. J., de Muinck, E. D. & Smits, J. F. Cardiovascular phenotyping in mice. Cardiovasc. Res. 39, 34–49 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Krishnan, A. et al. A detailed comparison of mouse and human cardiac development. Pediatr. Res. 76, 500–507 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Reilly, S. K. & Noonan, J. P. Evolution of gene regulation in humans. Annu. Rev. Genomics Hum.Genet. 17, 45–67 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Carroll, S. B. Evo-devo and an expanding evolutionary synthesis: a genetic theory of morphological evolution. Cell 134, 25–36 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Carroll, S. B. et al. Pattern formation and eyespot determination in butterfly wings. Science 265, 109–114 (1994).

    Article  CAS  PubMed  Google Scholar 

  22. Cotney, J. et al. The evolution of lineage-specific regulatory activities in the human embryonic limb. Cell 154, 185–196 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Reilly, S. K. et al. Evolutionary genomics. Evolutionary changes in promoter and enhancer activity during human corticogenesis. Science 347, 1155–1159 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. de la Torre-Ubieta, L. et al. The dynamic landscape of open chromatin during human cortical neurogenesis. Cell 172, 289–304 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Van Vliet, P., Wu, S. M., Zaffran, S. & Pucéat, M. Early cardiac development: a view from stem cells to embryos. Cardiovasc. Res. 96, 352–362 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Veevers, J. et al. Cell-surface marker signature for enrichment of ventricular cardiomyocytes derived from human embryonic stem cells. Stem Cell Rep. 11, 828–841 (2018).

    Article  CAS  Google Scholar 

  27. Zhang, Y. et al. Transcriptionally active HERV-H retrotransposons demarcate topologically associating domains in human pluripotent stem cells. Nat. Genet. 51, 1380–1388 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhu, J. et al. Genome-wide chromatin state transitions associated with developmental and environmental cues. Cell 152, 642–654 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stergachis, A. B. et al. Developmental fate and cellular maturity encoded in human regulatory DNA landscapes. Cell 154, 888–903 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Heintzman, N. D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sahara, M. et al. Population and single-cell analysis of human cardiogenesis reveals unique LGR5 ventricular progenitors in embryonic outflow tract. Dev. Cell 48, 475–490 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Asp, M. et al. A spatiotemporal organ-wide gene expression and cell atlas of the developing human heart. Cell 179, 1647–1660 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Mononen, M. M., Leung, C. Y., Xu, J. & Chien, K. R. Trajectory mapping of human embryonic stem cell cardiogenesis reveals lineage branch points and an ISL1 progenitor-derived cardiac fibroblast lineage. Stem Cells 38, 1267–1278 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Grunert, M., Dorn, C. & Rickert-Sperling, S. Cardiac transcription factors and regulatory networks. in Congenital Heart Diseases: The Broken Heart (eds Rickert-Sperling, S., Kelly, R. G. & Driscoll, D. J.) 139–152 (Springer, 2016).

  35. Bertero, A. et al. Dynamics of genome reorganization during human cardiogenesis reveal an RBM20-dependent splicing factory. Nat. Commun. 10, 1538 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Liu, Q. et al. Genome-wide temporal profiling of transcriptome and open chromatin of early cardiomyocyte differentiation derived from hiPSCs and hESCs. Circ. Res. 121, 376–391 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. 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 

  39. Schug, J. et al. Promoter features related to tissue specificity as measured by Shannon entropy. Genome Biol. 6, R33 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Uebbing, S. et al. Massively parallel discovery of human-specific substitutions that alter enhancer activity. Proc. Natl Acad. Sci. USA 118, e2007049118 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Dowen, J. M. et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159, 374–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Visel, A., Minovitsky, S., Dubchak, I. & Pennacchio, L. A. VISTA Enhancer Browser—a database of tissue-specific human enhancers. Nucleic Acids Res. 35, D88–D92 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Aronsen, J. M., Swift, F. & Sejersted, O. M. Cardiac sodium transport and excitation-contraction coupling. J. Mol. Cell. Cardiol. 61, 11–19 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. George, A. L. Jr. et al. Assignment of the human heart tetrodotoxin-resistant voltage-gated Na+ channel alpha-subunit gene (SCN5A) to band 3p21. Cytogenet. Cell Genet. 68, 67–70 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Berthold, J., Schenkova, K. & Rivero, F. Rho GTPases of the RhoBTB subfamily and tumorigenesis. Acta Pharmacol. Sin. 29, 285–295 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. 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 

  47. Kerr, K. F. et al. Genome-wide association study of heart rate and its variability in Hispanic/Latino cohorts. Heart Rhythm 14, 1675–1684 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Creutz, C. E. et al. The copines, a novel class of C2 domain-containing, calcium-dependent, phospholipid-binding proteins conserved from Paramecium to humans. J. Biol. Chem. 273, 1393–1402 (1998).

    Article  CAS  PubMed  Google Scholar 

  49. DiFrancesco, D. HCN4, sinus bradycardia and atrial fibrillation. Arrhythm. Electrophysiol. Rev. 4, 9–13 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Hennis, K. et al. Speeding up the heart? Traditional and new perspectives on HCN4 function. Front. Physiol. 12, 669029 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Cibi, D. M. et al. Prdm16 deficiency leads to age-dependent cardiac hypertrophy, adverse remodeling, mitochondrial dysfunction, and heart failure. Cell Rep. 33, 108288 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Nam, J. M., Lim, J. E., Ha, T. W., Oh, B. & Kang, J. O. Cardiac-specific inactivation of Prdm16 effects cardiac conduction abnormalities and cardiomyopathy-associated phenotypes. Am. J. Physiol. Heart Circ. Physiol. 318, H764–h777 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Jiang, Z. et al. Zic3 is required in the extra-cardiac perinodal region of the lateral plate mesoderm for left–right patterning and heart development. Hum. Mol. Genet. 22, 879–889 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. 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 

  55. 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 

  56. 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 

  57. Preissl, S. et al. Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation. Nat. Neurosci. 21, 432–439 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kaestner, K. H. The FoxA factors in organogenesis and differentiation. Curr. Opin. Genet. Dev. 20, 527–532 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Tsankov, A. M. et al. Transcription factor binding dynamics during human ES cell differentiation. Nature 518, 344–349 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bedard, J. E., Haaning, A. M. & Ware, S. M. Identification of a novel ZIC3 isoform and mutation screening in patients with heterotaxy and congenital heart disease. PLoS ONE 6, e23755 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bellchambers, H. M. & Ware, S. M. Loss of Zic3 impairs planar cell polarity leading to abnormal left–right signaling, heart defects and neural tube defects. Hum. Mol. Genet. 30, 2402–2415 (2021).

  67. Cast, A. E., Gao, C., Amack, J. D. & Ware, S. M. An essential and highly conserved role for Zic3 in left–right patterning, gastrulation and convergent extension morphogenesis. Dev. Biol. 364, 22–31 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kitaguchi, T., Nagai, T., Nakata, K., Aruga, J. & Mikoshiba, K. Zic3 is involved in the left–right specification of the Xenopus embryo. Development 127, 4787–4795 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. 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  Google Scholar 

  70. Yang, R. et al. Amnion signals are essential for mesoderm formation in primates. Nat. Commun. 12, 5126 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  72. Belmont, J. W., Mohapatra, B., Towbin, J. A. & Ware, S. M. Molecular genetics of heterotaxy syndromes. Curr. Opin. Cardiol. 19, 216–220 (2004).

    Article  PubMed  Google Scholar 

  73. Chhin, B. et al. Elucidation of penetrance variability of a ZIC3 mutation in a family with complex heart defects and functional analysis of ZIC3 mutations in the first zinc finger domain. Hum. Mutat. 28, 563–570 (2007).

    Article  CAS  PubMed  Google Scholar 

  74. 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 

  75. Drakhlis, L. et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat. Biotechnol. 39, 737–746 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Hofbauer, P. et al. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 184, 3299–3317 (2021).

    Article  CAS  PubMed  Google Scholar 

  77. Lewis-Israeli, Y. R. et al. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat. Commun. 12, 5142 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Richards, D. J. et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat. Biomed. Eng. 4, 446–462 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Rossi, G. et al. Capturing cardiogenesis in gastruloids. Cell Stem Cell 28, 230–240 (2021).

    Article  CAS  PubMed  Google Scholar 

  80. Lian, X. et al. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat. Protoc. 8, 162–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Concordet, J. P. & Haeussler, M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 46, W242–W245 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. 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 

  85. Robinson, J. T. et al. Juicebox.js provides a cloud-based visualization system for Hi-C data. Cell Syst. 6, 256–258 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Deacon, D. C. et al. Combinatorial interactions of genetic variants in human cardiomyopathy. Nat. Biomed. Eng. 3, 147–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. 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 

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

    Article  CAS  PubMed  Google Scholar 

  89. 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 

  90. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Minn, K. T. et al. High-resolution transcriptional and morphogenetic profiling of cells from micropatterned human ESC gastruloid cultures. eLife 9, e59445 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Buniello, A. et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47, D1005–d1012 (2019).

    Article  CAS  PubMed  Google Scholar 

  93. Harley, J. B. et al. Transcription factors operate across disease loci, with EBNA2 implicated in autoimmunity. Nat. Genet. 50, 699–707 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. de Hoon, M. J., Imoto, S., Nolan, J. & Miyano, S. Open source clustering software. Bioinformatics 20, 1453–1454 (2004).

    Article  PubMed  Google Scholar 

  95. Saldanha, A. J. Java Treeview—extensible visualization of microarray data. Bioinformatics 20, 3246–3248 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Swim for help with analysis of flow cytometry data and E. Ines for technical assistance. We thank K. Jepsen and the UCSD Institute for Genomic Medicine for help with sequencing and the UCSD Human Embryonic Stem Cell Core Facility for help with cell sorting. This work was supported, in part, by grants from the National Institutes of Health (1UM1HL128773) to B.R., S.M.E. and N.C.C.; and the American Heart Association (18CDA34080195) to J.B.

Author information

Authors and Affiliations

Authors

Contributions

E.D., B.R. and N.C.C. conceived the project and the overall design of the experimental strategy. C.B. designed and performed bioinformatics analyses. E.D., F.Z., S.P., E.F., X.H., A.Y.L. and J.G. conducted experiments. Y.Z., O.B.P. and R.H. helped with bioinformatics analysis. B.R., S.M.E., J.B. and H.S.K. provided critical intellectual input and data interpretation. E.D. and N.C.C. prepared the manuscript, with input from all authors.

Corresponding authors

Correspondence to Chris Benner or Neil C. Chi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Mirana Ramialison 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.

Integrated supplementary information

Extended Data Fig. 1 Gene regulatory networks dynamically remodel during human cardiomyocyte differentiation.

a, Flow cytometry analysis shows the percentage of cell types generated at each cardiac developmental stage as assessed by the indicated markers. b, Heatmap shows cell-type cardiac enhancers through comparison with ENCODE-derived cell line enhancer data. c, CREs are enriched in intergenic and intronic regions during cardiomyocyte differentiation. d, Number of CREs for different epigenomic assays are shown across the six stages of human vCM differentiation. e, Bar plot shows the number of CREs that are gained or lost compared to the preceding stage during human ventricular cardiomyocyte differentiation. f, Analyzing the ratio of active enhancers (H3K27ac+/H3K4me1+) to all enhancers (H3K4me1+) reveals increased enhancer utilization during human vCM differentiation. g, Heatmap displays enhancer activity as assessed by H3K27ac signal during in vitro vCM differentiation and in purified human fetal CMs. PSC, human pluripotent stem cells; Mes, mesoderm; CMes, cardiac mesoderm; CP, cardiac progenitor; CM, cardiomyocyte; vCM, ventricular cardiomyocyte; fvCM, fetal ventricular cardiomyocyte; CRE, cis-regulatory region; pos., positive; snRNA, small nuclear RNA; snoRNA, small nucleolar RNA; UTR, untranslated region; pseudo, pseudogene; TTS, transcription termination site; ncRNA; non-coding RNA; miRNA, microRNA; HUVEC, human umbilical vein endothelial cells; NHEK, normal human epidermal keratinocytes; fvCM, fetal ventricular cardiomyocyte.

Extended Data Fig. 2 Integrative transcriptional and chromatin accessibility analysis identifies putative key cardiac regulators.

a, Dot plot shows expression (circle color) and transcription factor motif enrichment (circle size) of known and potential novel regulators of human ventricle cardiomyocyte differentiation. b, Heatmap shows differentially regulated super-enhancers during cardiomyocyte differentiation. c, Distribution of H3K27ac signal for each of the indicated stages shows super-enhancer loci with associated genes of interest based on proximity. PSC, pluripotent stem cells; Mes, mesoderm; Cmes, cardiac mesoderm; CP, cardiac progenitor; CM, cardiomyocyte; vCM, ventricular cardiomyocyte; FPKM, fragments per kilobase of transcript per million mapped reads.

Extended Data Fig. 3 Human gained enhancers consist of traditional and super-enhancers during cardiomyocyte differentiation.

a, HGEs are enriched in intergenic and intronic regions during cardiomyocyte differentiation. b, HGEs consist of both traditional (TE) and super-enhancers (SE). c, Violin plots reveal comparable activity of HGEs compared to all other mouse stages combined. Dashed line in violin plots indicates the mean and dotted lines indicate quartiles. d, Shannon entropy calculations show stage-specific expression of genes near gained or stable enhancers. Lower entropy indicates more stage-specific gene expression. Horizontal line in boxplots represents the median, the box indicates the interquartile range and the dots represent outliers. Data was compared using Wilcoxon rank sum test (****p value < 0.0001). PSC-Stable, n = 7339; PSC-Gained, n = 1240; Mes-Stable, n = 6436; Mes-Gained, n = 1924; CP-Stable, n = 7525; CP-Stable, n = 1531; CM-Gained, n = 7531; CM-Gained, n = 1653. e, Stable and gained super-enhancer domains are primarily associated with transcription factors based on GO-term analysis. f, Representative super-enhancer TADs with HGEs (blue boxes) are shown at various cardiomyocyte developmental stages. HGE, human-gained enhancer; PSC, pluripotent stem cells; Mes, mesoderm; CP, cardiac progenitor; CM, cardiomyocyte; UTR, untranslated region; pseudo, pseudogene; TTS, transcription termination site; enh., enhancers; TAD, topologically associated domain; hs, human; ms, mouse.

Extended Data Fig. 4 HGEs are associated with cardiac developmental specific activity and function.

a, Bar graph shows the percentage and number of VISTA validated enhancers driving cardiac LacZ reporter activity in vivo that overlap with stable and gained enhancers at different cardiomyocyte developmental stages. b, GREAT analysis reveals biological processes associated with HGEs at the examined cardiac developmental stages. c-f, ATAC-seq/ChIP-seq profiles show that SNPs associated with (c) atrial fibrillation and (e) QRS duration overlap with HGEs, and luciferase reporter assays (d, f) confirm that the SNPs affect enhancer activity. Sequence comparison surrounding the SNP is shown on the bottom left of (c) and (e) with nucleotides that are not conserved between mouse and human indicated in red. Gene expression in human (grey) and mouse (black) cardiomyocytes is shown on the bottom right of (c) and (e). n = 9 for the luciferase reporter assays. Data was compared using a two-tailed Student’s t-test. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. Data represented as the mean ± SEM with individual data points. g, Bar graph shows the percentage and absolute number of noncoding mutations found in CHD patients that overlap with stable and gained enhancers at different cardiomyocyte developmental stages. HGE, human-gained enhancer; PSC, pluripotent stem cells; Mes, mesoderm; CMes, cardiac mesoderm; CP, cardiac progenitor; CM, cardiomyocyte; vCM, ventricular cardiomyocyte; snRNA, small nuclear RNA; mitoch., mitochondrial; transl., translation; carb., carbohydrate; catab., catabolism; cyto., cytoplasmic; FDR, false discovery rate; Hs, human; Ms, mouse; expr, expression; FKPM, fragments per kilobase per million reads; EV, empty vector; WT, wild-type; SNP; single nucleotide polymorphism; CHD, congenital heart disease.

Extended Data Fig. 5 Gene expression and transcription factor motif analyses reveal distinct differences between human and mouse mesoderm.

a, Clustered heatmap of human and mouse mesoderm gene expression profiles shows both conserved and divergent gene expression clusters. b, Table shows top transcription factor motifs for human and mouse mesoderm active enhancers. Mes, mesoderm; val., value. Motif enrichment p-values are derived from a one-sided binomial test.

Extended Data Fig. 6 CREs within mesoderm cell subpopulations are associated with distinct transcription factor binding activity.

a, Clustered heatmap shows differentially (FDR < 0.05) accessible CREs between identified snATAC mesoderm cell subpopulations. b, Heatmap of transcription factor binding motif analysis of cluster-specific CREs from human mesodermal snATAC-seq data reveals that ZIC3 along with other cardiac developmental transcription factor motifs are enriched in the pre-cardiac mesoderm subpopulation. c, ZIC3 motif enrichment is projected onto the UMAP of human PSC-derived mesoderm snATAC-seq data. d, (Left) Mouse scRNA-seq data from E7.25-E8.5 Mesp1-Cre; Rosa26-tdTomato embryos are displayed by tSNE (t-distributed stochastic neighbor embedding) plots for all developmental stages combined. (Right) Zic3 expression is projected onto the tSNE plot. PSC, pluripotent stem cell; N, neural; Epi, epiblast; ExE, extra-embryonic; APS/ME, anterior primitive streak/mesendoderm; LPM, lateral-plate mesoderm; PCM, pre-cardiac mesoderm; PM; paraxial mesoderm, PSM, pre-somitic mesoderm; CRE, cis-regulatory region; adj., adjusted.

Extended Data Fig. 7 Generation of clonal ZIC3 HGE deletion lines.

Human PSC ZIC3 HGE1-3 individual knockout (KO) and triple KO (TKO) lines were generated through CRISPR/Cas9-mediated deletion of ZIC3 HGE1-3 enhancer regions (9.2 kb, 1.8 kb, and 1.5 kb, respectively). PCR genotyping using external (ext., that is, outside deleted regions) and internal (int., that is, inside deleted regions) primers confirmed the individual human PSC ZIC3 HGE KOs (left) and the human PSC ZIC3 HGE TKO (right). HGE TKO was confirmed by individual HGE1-3 genotyping. Wild-type (WT) cells were used as control. Note that the WT HGE1 band is 9.2 kb and thus difficult to PCR amplify. M, marker, ext., external; int., internal.

Extended Data Fig. 8 HGE3 KO and HGE TKO human pluripotent stem cell knockout lines display reduced cardiomyocyte differentiation and mesoderm-specific ZIC3 expression defects.

All individual PCR data for WT and ZIC3 HGE knockout (KO) and HGE triple knockout (TKO) lines are shown with statistical significance indicated (ZIC3, n= 9 for all lines; HOPX, n = 9 for WT/HGE1-3 KO lines, n = 12 for WT/HGE TKO lines; APLNR, n = 6 for WT/HGE1 and HGE2 KO lines, n = 9 for WT/HGE3 KO lines, n = 12 for WT/HGE TKO lines; NODAL, n = 6 for WT/HGE1 and HGE2 KO lines, n = 12 for WT/HGE3 KO and HGE TKO lines; LEFTY2, n = 6 for WT/HGE1 and HGE2 KO lines, n = 9 for WT/HGE3 KO lines, n = 12 for WT/HGE TKO lines. TBP gene expression was used for gene expression normalization. b, qPCR analysis of ZIC3 HGE3 KO and HGE TKO human PSC lines reveals no significant changes in ZIC3 expression at the PSC stage compared to WT cells (n = 9). c, qPCR analysis reveals significant changes in the expression of ZIC3 but not neighboring genes, RBMX and FGF13, in HGE TKO human PSC lines when compared to WT human PSC lines at the mesoderm stage (n = 6). d, Representative examples of flow cytometry analyses for TNNT2 staining (x-axis) show that ZIC3 HGE3 and TKO lines (but not HGE1 nor HGE2) display reduced cardiac differentiation. Data is represented as the mean ± SEM with individual data points. Data was analyzed using a two-tailed Student’s t-test. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. HGE, human-gained enhancer; expr., expression; PSC, pluripotent stem cells; Mes, mesoderm.

Supplementary information

Supplementary Fig. 1

Reporting Summary

Supplementary Table 1 HGEs for each cardiac stage

Supplementary Table 2 Genes in the vicinity of HGEs

Supplementary Table 3 MGEs for each cardiac stage

Supplementary Table 4 Validated VISTA enhancers overlapping HGEs

Supplementary Table 5 Genes differentially expressed between human and mouse mesoderm

Supplementary Table 6 snATAC-seq statistics

Supplementary Table 7 snATAC-seq-associated genes with cluster-specific gene scores

Supplementary Table 8 Annotated snATAC-seq peaks for mesoderm subclusters

Supplementary Table 9 Cluster-specific snATAC-seq peaks

Supplementary Table 10 TF binding motifs enriched in snATAC-seq peaks

Supplementary Table 11 ZIC3-regulated mesoderm genes

Supplementary Table 12 Sequences of oligonucleotides used in the study

Supplementary Table 13 Raw sequencing datasets used in the study

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Destici, E., Zhu, F., Tran, S. et al. Human-gained heart enhancers are associated with species-specific cardiac attributes. Nat Cardiovasc Res 1, 830–843 (2022). https://doi.org/10.1038/s44161-022-00124-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-022-00124-7

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