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:

A map of cis-regulatory elements and 3D genome structures in zebrafish

Abstract

The zebrafish (Danio rerio) has been widely used in the study of human disease and development, and about 70% of the protein-coding genes are conserved between the two species1. However, studies in zebrafish remain constrained by the sparse annotation of functional control elements in the zebrafish genome. Here we performed RNA sequencing, assay for transposase-accessible chromatin using sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing, whole-genome bisulfite sequencing, and chromosome conformation capture (Hi-C) experiments in up to eleven adult and two embryonic tissues to generate a comprehensive map of transcriptomes, cis-regulatory elements, heterochromatin, methylomes and 3D genome organization in the zebrafish Tübingen reference strain. A comparison of zebrafish, human and mouse regulatory elements enabled the identification of both evolutionarily conserved and species-specific regulatory sequences and networks. We observed enrichment of evolutionary breakpoints at topologically associating domain boundaries, which were correlated with strong histone H3 lysine 4 trimethylation (H3K4me3) and CCCTC-binding factor (CTCF) signals. We performed single-cell ATAC-seq in zebrafish brain, which delineated 25 different clusters of cell types. By combining long-read DNA sequencing and Hi-C, we assembled the sex-determining chromosome 4 de novo. Overall, our work provides an additional epigenomic anchor for the functional annotation of vertebrate genomes and the study of evolutionarily conserved elements of 3D genome organization.

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: Identification of cis-regulatory elements in the zebrafish genome.
Fig. 2: Characterization of tissue-specific cis-regulatory elements.
Fig. 3: Analysis of heterochromatin and repetitive elements and de novo assembly of zebrafish chromosome 4.
Fig. 4: Conservation of zebrafish cis-regulatory elements and transcriptional networks.
Fig. 5: Higher-order chromatin structure and zebrafish genome evolution.

Similar content being viewed by others

Data availability

All the sequencing data are deposited in the NCBI Gene Expression Omnibus under accession code GSE134055. All the genomic data generated in this study can be visualized in the WashU Epigenome Browser (https://epigenome.wustl.edu/zebrafishENCODE/). The human histone-modification ChIP-seq data were downloaded from the ROADMAP Project. The mouse histone modification ChIP-seq data were downloaded from the mouse ENCODE Consortium. The human tissue transcriptome data were downloaded from the GTEx Consortium. The public zebrafish ChIP-seq and ATAC-seq data used in this study are listed in Supplementary Table 6. The human h1-ESC Hi-C data were downloaded from GSE52457. GM12878 and K562 GRO-seq data were downloaded from GSE60456. GM12878 and K562 CTCF ChIP-seq were downloaded from GSE31477. GM12878 and K562 Pol2 ChIP-seq were downloaded from GSE91426 and GSE31477. Source data are provided with this paper.

References

  1. Howe, K. et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature 496, 498–503 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gerhard, G. S. et al. Life spans and senescent phenotypes in two strains of Zebrafish (Danio rerio). Exp. Gerontol. 37, 1055–1068 (2002).

    PubMed  Google Scholar 

  3. Lamason, R. L. et al. SLC24A5, a putative cation exchanger, affects pigmentation in zebrafish and humans. Science 310, 1782–1786 (2005).

    ADS  CAS  PubMed  Google Scholar 

  4. Vastenhouw, N. L. et al. Chromatin signature of embryonic pluripotency is established during genome activation. Nature 464, 922–926 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bogdanovic, O. et al. Dynamics of enhancer chromatin signatures mark the transition from pluripotency to cell specification during embryogenesis. Genome Res. 22, 2043–2053 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kaaij, L. J. et al. Enhancers reside in a unique epigenetic environment during early zebrafish development. Genome Biol. 17, 146 (2016).

    PubMed  PubMed Central  Google Scholar 

  7. Aday, A. W., Zhu, L. J., Lakshmanan, A., Wang, J. & Lawson, N. D. Identification of cis regulatory features in the embryonic zebrafish genome through large-scale profiling of H3K4me1 and H3K4me3 binding sites. Dev. Biol. 357, 450–462 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Vesterlund, L., Jiao, H., Unneberg, P., Hovatta, O. & Kere, J. The zebrafish transcriptome during early development. BMC Dev. Biol. 11, 30 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    ADS  Google Scholar 

  11. Yue, F. et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature 515, 355–364 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Anderson, J. L. et al. Multiple sex-associated regions and a putative sex chromosome in zebrafish revealed by RAD mapping and population genomics. PLoS ONE 7, e40701 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Klemm, S. L., Shipony, Z. & Greenleaf, W. J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20, 207–220 (2019).

    CAS  PubMed  Google Scholar 

  14. Meuleman, W. et al. Index and biological spectrum of human DNase I hypersensitive sites. Nature 584, 244–251 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Quillien, A. et al. Robust identification of developmentally active endothelial enhancers in zebrafish using FANS-assisted ATAC-seq. Cell Rep. 20, 709–720 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Letelier, J. et al. Evolutionary emergence of the rac3b/rfng/sgca regulatory cluster refined mechanisms for hindbrain boundaries formation. Proc. Natl Acad. Sci. USA 115, E3731–E3740 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Liu, G., Wang, W., Hu, S., Wang, X. & Zhang, Y. Inherited DNA methylation primes the establishment of accessible chromatin during genome activation. Genome Res. 28, 998–1007 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Marlétaz, F. et al. Amphioxus functional genomics and the origins of vertebrate gene regulation. Nature 564, 64–70 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  19. Meier, M. et al. Cohesin facilitates zygotic genome activation in zebrafish. Development 145, dev156521 (2018).

    PubMed  Google Scholar 

  20. Torbey, P. et al. Cooperation, cis-interactions, versatility and evolutionary plasticity of multiple cis-acting elements underlie krox20 hindbrain regulation. PLoS Genet. 14, e1007581 (2018).

    PubMed  PubMed Central  Google Scholar 

  21. Paik, E. J. et al. A Cdx4–Sall4 regulatory module controls the transition from mesoderm formation to embryonic hematopoiesis. Stem Cell Reports 1, 425–436 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kang, J. et al. Modulation of tissue repair by regeneration enhancer elements. Nature 532, 201–206 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kaufman, C. K. et al. A zebrafish melanoma model reveals emergence of neural crest identity during melanoma initiation. Science 351, aad2197 (2016).

    PubMed  PubMed Central  Google Scholar 

  24. Goldman, J. A. et al. Resolving heart regeneration by replacement histone profiling. Dev. Cell 40, 392–404 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Pérez-Rico, Y. A. et al. Comparative analyses of super-enhancers reveal conserved elements in vertebrate genomes. Genome Res. 27, 259–268 (2017).

    PubMed  PubMed Central  Google Scholar 

  26. Lister, R. et al. Global epigenomic reconfiguration during mammalian brain development. Science 341, 1237905 (2013).

    PubMed  PubMed Central  Google Scholar 

  27. Visel, A. et al. Ultraconservation identifies a small subset of extremely constrained developmental enhancers. Nat. Genet. 40, 158–160 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Dimitrieva, S. & Bucher, P. UCNEbase–a database of ultraconserved non-coding elements and genomic regulatory blocks. Nucleic Acids Res. 41, D101–D109 (2013).

    CAS  PubMed  Google Scholar 

  29. 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).

    CAS  PubMed  Google Scholar 

  30. Corces, M. R. et al. The chromatin accessibility landscape of primary human cancers. Science 362, eaav1898 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  31. Neph, S. et al. Circuitry and dynamics of human transcription factor regulatory networks. Cell 150, 1274–1286 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Yang, T. et al. HiCRep: assessing the reproducibility of Hi-C data using a stratum-adjusted correlation coefficient. Genome Res. 27, 1939–1949 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Krefting, J., Andrade-Navarro, M. A. & Ibn-Salem, J. Evolutionary stability of topologically associating domains is associated with conserved gene regulation. BMC Biol. 16, 87 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Lazar, N. H. et al. Epigenetic maintenance of topological domains in the highly rearranged gibbon genome. Genome Res. 28, 983–997 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Fishman, V. et al. 3D organization of chicken genome demonstrates evolutionary conservation of topologically associated domains and highlights unique architecture of erythrocytes’ chromatin. Nucleic Acids Res. 47, 648–665 (2019).

    CAS  PubMed  Google Scholar 

  36. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Smagulova, F. et al. Genome-wide analysis reveals novel molecular features of mouse recombination hotspots. Nature 472, 375–378 (2011).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Canela, A. et al. Genome organization drives chromosome fragility. Cell 170, 507–521 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gothe, H. J. et al. Spatial chromosome folding and active transcription drive DNA fragility and formation of oncogenic MLL translocations. Mol. Cell 75, 267–283 (2019).

    CAS  PubMed  Google Scholar 

  40. Canela, A. et al. Topoisomerase II–induced chromosome breakage and translocation is determined by chromosome architecture and transcriptional activity. Mol. Cell 75, 252–266 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Postlethwait, J. H. et al. Vertebrate genome evolution and the zebrafish gene map. Nat. Genet. 18, 345–349 (1998).

    CAS  PubMed  Google Scholar 

  42. Pedroso, G. L. et al. Blood collection for biochemical analysis in adult zebrafish. J. Vis. Exp. 3865, e3865 (2012).

    Google Scholar 

  43. Rao, S. S. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Xie, W. et al. Epigenomic analysis of multilineage differentiation of human embryonic stem cells. Cell 153, 1134–1148 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  46. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang, L. et al. CPAT: Coding-Potential Assessment Tool using an alignment-free logistic regression model. Nucleic Acids Res. 41, e74 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  49. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Maertin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  53. Korhonen, J., Martinmäki, P., Pizzi, C., Rastas, P. & Ukkonen, E. MOODS: fast search for position weight matrix matches in DNA sequences. Bioinformatics 25, 3181–3182 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Kulakovskiy, I. V. et al. HOCOMOCO: towards a complete collection of transcription factor binding models for human and mouse via large-scale ChIP-seq analysis. Nucleic Acids Res. 46 (D1), D252–D259 (2018).

    CAS  PubMed  Google Scholar 

  55. Gerstein, M. B. et al. Integrative analysis of the Caenorhabditis elegans genome by the modENCODE project. Science 330, 1775–1787 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  57. Liu, T. Use model-based Analysis of ChIP-Seq (MACS) to analyze short reads generated by sequencing protein-DNA interactions in embryonic stem cells. Methods Mol. Biol. 1150, 81–95 (2014).

    CAS  PubMed  Google Scholar 

  58. Hiller, M. et al. Computational methods to detect conserved non-genic elements in phylogenetically isolated genomes: application to zebrafish. Nucleic Acids Res. 41, e151 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Lee, H. J. et al. Regenerating zebrafish fin epigenome is characterized by stable lineage-specific DNA methylation and dynamic chromatin accessibility. Genome Biol. 21, 52 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhou, X., Li, D., Lowdon, R. F., Costello, J. F. & Wang, T. methylC Track: visual integration of single-base resolution DNA methylation data on the WashU EpiGenome Browser. Bioinformatics 30, 2206–2207 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Burger, L., Gaidatzis, D., Schübeler, D. & Stadler, M. B. Identification of active regulatory regions from DNA methylation data. Nucleic Acids Res. 41, e155 (2013).

    PubMed  PubMed Central  Google Scholar 

  63. Wu, H. et al. Detection of differentially methylated regions from whole-genome bisulfite sequencing data without replicates. Nucleic Acids Res. 43, e141 (2015).

    PubMed  PubMed Central  Google Scholar 

  64. Hansen, K. D., Langmead, B. & Irizarry, R. A. BSmooth: from whole genome bisulfite sequencing reads to differentially methylated regions. Genome Biol. 13, R83 (2012).

    PubMed  PubMed Central  Google Scholar 

  65. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44 (W1), W160–W165 (2016).

    PubMed  PubMed Central  Google Scholar 

  66. Koren, S. et al. Canu: scalable and accurate long-read assembly via adaptive k-mer weighting and repeat separation. Genome Res. 27, 722–736 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Walker, B. J. et al. Pilon: an integrated tool for comprehensive microbial variant detection and genome assembly improvement. PLoS ONE 9, e112963 (2014).

    ADS  PubMed  PubMed Central  Google Scholar 

  68. Dudchenko, O. et al. De novo assembly of the Aedes aegypti genome using Hi-C yields chromosome-length scaffolds. Science 356, 92–95 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    PubMed  PubMed Central  Google Scholar 

  73. Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3, 95–98 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Abdennur, N. & Mirny, L. A. Cooler: scalable storage for Hi-C data and other genomically labeled arrays. Bioinformatics 36, 311–316 (2020).

    CAS  PubMed  Google Scholar 

  76. Crane, E. et al. Condensin-driven remodelling of X chromosome topology during dosage compensation. Nature 523, 240–244 (2015).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  77. Giorgetti, L. et al. Structural organization of the inactive X chromosome in the mouse. Nature 535, 575–579 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  78. Imakaev, M. et al. Iterative correction of Hi-C data reveals hallmarks of chromosome organization. Nat. Methods 9, 999–1003 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Darling, A. E., Mau, B. & Perna, N. T. progressiveMauve: multiple genome alignment with gene gain, loss and rearrangement. PLoS ONE 5, e11147 (2010).

    ADS  PubMed  PubMed Central  Google Scholar 

  80. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Johansen, N. & Quon, G. scAlign: a tool for alignment, integration, and rare cell identification from scRNA-seq data. Genome Biol. 20, 166 (2019).

    PubMed  PubMed Central  Google Scholar 

  82. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants R35GM124820, R01HG009906, R24DK106766 (R.C.H. and F.Y.) and R01DK107735 (G.S.G.). F.Y. is also supported by U01CA200060. T.W. is supported by NIH grants R01HG007175, R01HG007354, R01ES024992, U24ES026699 and U01HG009391. We thank J. A. Stamatoyannopoulos for discussion and suggestions; H. Lyu for proof reading and other Yue lab members for discussion; and E. DeForest, S. Stella, P. Hubley and Penn State Zebrafish Functional Genomics Core for fish husbandry and embryo collection.

Author information

Authors and Affiliations

Authors

Contributions

F.Y. conceived and supervised the project. H.Y. and T.L. collected tissue and conducted experiments. Y.L. led the data analysis. Y.L., H.Y., H.J.L., Y.W., X.W., B.Z., L.F. and J.W. conducted analyses. D.L. and T.W. provided the website for data presentation. K.C.A. and K.C.C. provided animal support. Q.J., X.X., J.X., F.S., I.S., C. K., T.S., M.N.K.C., J.T., K.W., G.S.G., R.C.H., T.W. and K.C.C. helped with data interpretation. H.Y., Y.L., T.L. and F.Y. prepared the manuscript with input from all authors.

Corresponding author

Correspondence to Feng Yue.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Michael Beer, Jesse Dixon and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Tissue-specific gene expression in zebrafish.

a, Clustering analysis of transcripts from RNA-seq data in embryonic and adult tissues (n = 31,842). b, c, Gene Ontology and KEGG pathway analysis for the tissue-specific genes in adult brain, heart and testis (the number of tissue-specific genes in these two figures are, Brain = 3,693, Heart = 392, Testis = 1,605). d, Distribution of H3K4me3 signals surrounding the known and predicted novel transcripts. e, Human orthologues of zebrafish tissue-specific genes were more tissue-specific compared to human orthologues of non-tissue-specific zebrafish genes (n = 14,764, 3,739, 6,043, Mann–Whitney U Test, two-sided, ***P < 2.2 × 10−16).

Source data

Extended Data Fig. 2 Comparative analysis of zebrafish cis-regulatory elements.

a, Comparison of the predicted regulatory elements identified with previous data. Enhancers were based on H3K27ac signals in the same four tissues (brain, heart, intestine, testis) from Perez-Rico et al. 2017. The data we generated are from Tübingen zebrafish strain and the published results were from the AB strain. b, Number of predicted cis-regulatory elements in each tissue. E-brain stands for 1 dpf embryonic neuron cells. E-trunk stands for 1 dpf zebrafish whole trunk region. c, An example showing genes with active promoters have higher expression level. Blue hollow bar indicates the known mrpl39 promoter. Orange hollow bar indicates the potential novel promoter. The mrpl39 promoter has H3K4me3 peaks in both muscle and brain, but only has strong H3K27ac signals in muscle and its expression is higher (4.43-fold). d, Gene Ontology results for the muscle-specific enhancers and skin-specific enhancers. We used the GREAT tool for this analysis (the numbers of tissue-specific enhancers used in this figure are muscle = 813, skin = 512).

Source data

Extended Data Fig. 3 Enhancer reporter assay for tissue-specific enhancers.

In total, 28 of 32 predicted tissue-specific enhancers showed consistent GFP signals in the corresponding tissues. For the eight brain enhancers tested, 63/95, 51/86, 85/119, 112/143, 27/45, 34/48, 27/41, 62/77, and 37/45 embryos, respectively, had green signals in the brain region. For the six tested heart enhancers, 64/94, 52/85, 79/121, 20/41, 51/95, 32/55 and 20/31 embryos, respectively, had green signals in the heart region. For the six tested muscle enhancers, 52/57, 26/30, 107/124, 53/63, 93/114, 61/67 and 66/78 embryos, respectively, had green signals in the trunk muscle. For the four selected kidney enhancers, 47/82, 35/67, 44/62, 15/42 and 56/110 embryos, respectively, had green signals in the kidney region.

Source data

Extended Data Fig. 4 Single-cell ATAC-seq in zebrafish brain.

a, Barcode selection of single cell ATAC-seq. The x-axis represents the log value of the number of unique molecular identifiers (UMI); the y axis represents the ratio of fragments in promoter regions; the red lines represent threshold, and the grey shadows represent that the barcode passed the filter. b, Genomic distribution of all differentially accessible (DA) peaks. c, Overlap of all differentially accessible peaks with enhancers predicted in bulk brain. d, Top, the cluster distribution in the tSNE projection. Bottom left, pileups of differentially accessible ATAC-seq signals for each cluster. Shown in the figure is the +/− 10kb flanking region surrounding peak centres. Bottom right, most significantly enriched transcription factor motif for each cluster. e, t-SNE projection of all scATAC-seq cells colored by Z-score of peak enrichment. f, Motif enrichment of known neuron-specific TFs in scATAC-seq predicted clusters (n = 19,955).

Source data

Extended Data Fig. 5 Heterochromatin annotation in adult tissues.

a, WashU Epigenome Browser screenshot of H3K9me3 and H3K9me2 histone ChIP-seq signals in 11 zebrafish adult tissues. The values on the y-axis were input-normalized. b, Distribution of H3K9me3 and H3K9me2 sites in the zebrafish genome. c, Venn diagram shows the overlap between H3K9me3 and H3K9me2 sites in zebrafish genome. d, Overlapping percentile of H3K9me3 and H3K9me2 peaks in adult tissues. e, H3K9me3 and H3K9me2 sites were depleted of ATAC-seq, H3K4me3 and H3K27ac ChIP-seq signals (n = 68,789 H3K9me3 sites and n = 73,777 H3K9me2 sites). f, Overlap of H3K9me3 sites, H3K9me2 sites, and ATAC-seq peaks with repetitive elements (The total number of each bar, from left to right, 68,789, 73,777 and 436,036). g, Examples of H3K9me3 sites in one tissue found to be active regions in other tissues. Horizontal scale 0-20 for H3K27ac and H3K4me3, 0-10 for RNA-seq, 0-5 for H3K9me3 and H3K9me2.

Extended Data Fig. 6 DNA methylation level and distribution in adult tissues.

a, Fraction of total CpGs with low (<25%), medium (≥25% and <75%), and high (≥75%) methylation levels and mean CpG methylation levels (mCG/CG) in zebrafish adult tissues (the mCG/CG ratio, from left to right, 0.788, 0.859, 0.790, 0.777, 0.791, 0.797, 0.781, 0.777, 0.804, 0.789, 0.781). b, Distribution of CpG methylation levels across zebrafish adult tissues. c, The distribution of non CpG methylation in 11 adult tissues. d, Mean methylation levels of the tissue-specific gene promoters. n represents the number of tissue-specific gene promoter. e, Mean methylation level of CpGs overlapping different genomic features or repetitive element classes. CDS, coding sequence. f, Number of UMRs and LMRs in zebrafish tissues and their overlap with enhancer and promoters (left panel) (number of UMR and LMR, from top to bottom, 14,990, 10,569, 14,569, 14,587, 14,831, 14,289, 13,842, 13,569, 14,424, 14,374, 13,908, 30,009, 7,916, 19,038, 21,411, 22,591, 16,796, 14,961, 16,268, 17,481, 15,932, 15,665) and ATAC-seq peaks (right panel)(numbers of UMR and LMR are the same with left panel). g, Clustering of tissue-specific hypoDMRs. Values in the heat map are mean methylation levels of hypoDMRs (n = 17,654, number of tissue-specific hypoDMRs).

Source data

Extended Data Fig. 7 De novo assembly of zebrafish chromosome 4 of the Tübingen strain.

a, WashU Epigenome Browser snapshot showing that heterochromatic marks H3K9me2 and H3K9me3 signals were enriched on chromosome 4 in zebrafish testis. The values on the y-axis were input-normalized. b, H3K9me2, H3K9me3, and DNA methylation level on chr4 long arm are significantly higher than other regions in all tissues (n = 11, two-sided, t-test). c, Overall strategy of de novo assembly of the Tübingen chr4 by integrating 10X, Nanopore, Bionano, and Hi-C data. d, Bionano long molecule sequencing data shows that there were many SVs on chr4 when mapped to the GRCz11 reference genome. e, SVs on chr4 detected by Bionano when the data were mapped to the de novo assembled chr4.

Source data

Extended Data Fig. 8 Conservation of cis-regulatory elements from zebrafish to other vertebrates.

a, Percentage of zebrafish enhancers whose sequences were conserved in human (the number of each bar, from left to right, 13,307, 7,018, 11,940, 7,499, 14,783, 14,272, 8,995, 13,777, 10,757, 15,505, 1,734, 4,011, 5,247). b, c, Similar to Fig. 4a. Percentage of zebrafish exons and cis-regulatory elements that have orthologous sequences in mouse and other fish species. Total number of each bar, from left to right: 1,000, 25,593, 58,065, 1,000. For exons and random, we randomly sample 1000 elements and computed their conservation percentage. The simulations were performed 20 times and the average percentage was presented. d, Another example of ultra-conserved noncoding element (UCNE). This element (FOXP1_Finn_1) is predicted to be a muscle enhancer in zebrafish, mouse, and human. Grey vertical bar marks the ultra-conserved region. Red vertical bar is the enhancer sequence in the human genome that was validated as a limb enhancer by transgenic mouse reporter assay in the VISTA Enhancer Browser (#hs956).

Source data

Extended Data Fig. 9 Distal ATAC-seq peak-to-gene pairs, enhancer-to-gene pairs, and transcriptional regulation network.

a, b, Distance distribution of cis-regulatory elements to their linked gene TSS. c, Correlation of ATAC-seq peak-to-gene pairs and Enhancer-to-gene pairs (n from left to right = 3,292, 3,827, 3,544, 3,281, 3,008, 2,795, 2,357, 2,001, 1,106). d, Validation of predicted enhancer-to-gene pairs by Hi-C interaction counts in muscle. e, mef2d is a regulator in both zebrafish muscle and heart, but it regulates different downstream targets by motif prediction analysis. f, The overall structure of the regulatory network is conserved between human and zebrafish. FFL connection analysis was performed, in this analysis, there are three types of nodes: A, driver node that regulates B and C; B, middle node, regulated by A but regulating node C; C, passenger node, regulated by both A and B.

Source data

Extended Data Fig. 10 Compartment and TADs in zebrafish.

a, Heat map of genome-wide Hi-C interaction matrices in zebrafish brain (blue) and muscle (red). b, Active marks (H3K4me3, H3K27ac, and ATAC-seq) were enriched in compartment A and depleted in compartment B. Repressive marks (H3K9me2 and H3K9me3) were enriched in compartment B. Error bands represent standard error of the mean. c, Genome browser snapshot of A/B compartment in brain and muscle. The blue vertical shaded area marks a region that is located in compartment B in brain but in compartment A in muscle. As expected, A compartment which is associated with more ATAC-seq peaks, H3K27ac and RNA-seq signals. d, Examples of shared TADs between zebrafish brain and muscle. e, Average DI scores surrounding TAD boundaries identified in brain (upper panel) and muscle (lower panel). f, ChIP-seq data shows that CTCF binding sites were enriched at TAD boundaries. g, Footprint analysis of ATAC-seq peaks in the TAD boundaries shows enrichment of CTCF binding motif (number of each bar, from left to right, 0.213, 0.24, 0.22, 0.237, 0.251, 0.232, 0.24, 0.262, 0.271, 0.281, 0.37, 0.27, 0.253, 0.25, 0.252, 0.253, 0.26, 0.23, 0.238, 0.24, 0.22). h, Repetitive elements enriched at TAD boundaries (left panel) and loop anchors (right panel).

Source data

Extended Data Fig. 11 Comparing zebrafish evolutionary breakpoints with TAD annotation.

a. Similar to Fig. 5d. Enrichment of evolutionary breakpoints at TAD boundaries. Relative positions of evolutionary breakpoints to TADs in 15 vertebrates. In all cases, we found that the evolutionary breakpoints were enriched at zebrafish TAD boundaries and depleted from the centre of TADs. Grey vertical bar labels the TAD body area. b, By comparing zebrafish with 17 vertebrates, H3K4me3 signals were found to be more enriched at TAD boundaries with breakpoints than those without breakpoints. Orange vertical bar labels the TAD boundaries. c, Higher H3K4me3 levels at breakpoint-containing TAD boundaries when using TADs annotation from zebrafish muscle were found as well, similar to Fig. 5g. d, H3K4me3 enrichment in human ESCs (H1) TAD boundaries with or without zebrafish-to-human breakpoints. e, H3K4me3 enrichment in mouse ESCs TAD boundaries with or without zebrafish-to-mouse breakpoints. f, H3K4me3 enrichment in human ESCs (H1) TAD boundaries with or without mouse-to-human breakpoints.

Extended Data Fig. 12 TADs with and without breakpoints.

a, H3K27ac and ATAC-seq signals do not show differences at TAD boundaries with breakpoints compared to those without breakpoints. Orange vertical bar labels the TAD boundaries. b, Sizes of TADs with and without evolutionary breakpoints were similar (n = 573, 777, two-sided, t-test). c, Enrichment of transcription at breakpoints (BP) that overlap with CTCF TAD boundaries in K562 cells (the number of breakpoints in blue line is 639, red line is 625). d, In 17 vertebrates, TADs without evolutionary breakpoints (bottom panel) have stronger interaction frequencies in the middle than TADs with evolutionary breakpoints (upper panel). Breakpoints in these 17 vertebrates were defined by comparing their genomes to the zebrafish genome. e, Distribution of correlations between the expression pattern of each pair of paralogs across 11 adult zebrafish tissues. f, Correlations between pairs of paralogs located on the same chromosome. Among them, 17 pairs were located within the same TAD, and the rest of the 65 pairs were located in different TADs. As a control, we randomly sampled 100 genes. Number of each bar, from left to right, 17, 65, 100.

Source data

Supplementary information

Supplementary Information

This file contains a guide to the Supplementary Tables 1-19.

Reporting Summary

Supplementary Tables

This zip file contains Supplementary Tables 1-19.

Supplementary Data

This zip file contains Supplementary Dataset 1.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, H., Luan, Y., Liu, T. et al. A map of cis-regulatory elements and 3D genome structures in zebrafish. Nature 588, 337–343 (2020). https://doi.org/10.1038/s41586-020-2962-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2962-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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