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:

Enhancer selection dictates gene expression responses in remote organs during tissue regeneration

Abstract

Acute trauma stimulates local repair mechanisms but can also impact structures distant from the injury, for example through the activity of circulating factors. To study the responses of remote tissues during tissue regeneration, we profiled transcriptomes of zebrafish brains after experimental cardiac damage. We found that the transcription factor gene cebpd was upregulated remotely in brain ependymal cells as well as kidney tubular cells, in addition to its local induction in epicardial cells. cebpd mutations altered both local and distant cardiac injury responses, altering the cycling of epicardial cells as well as exchange between distant fluid compartments. Genome-wide profiling and transgenesis identified a hormone-responsive enhancer near cebpd that exists in a permissive state, enabling rapid gene expression in heart, brain and kidney after cardiac injury. Deletion of this sequence selectively abolished cebpd induction in remote tissues and disrupted fluid regulation after injury, without affecting its local cardiac expression response. Our findings suggest a model to broaden gene function during regeneration in which enhancer regulatory elements define short- and long-range expression responses to injury.

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: cebpd is induced in local and remote tissues during heart regeneration.
Fig. 2: cebpd expression features with different stimuli and in various tissues.
Fig. 3: Local and remote Cebpd requirements during heart regeneration.
Fig. 4: Profiling and transgenesis identify an enhancer downstream of cebpd that responds to injury.
Fig. 5: Corticosteroid signals regulate distant cebpd expression during heart regeneration.
Fig. 6: CEN is required for distant cebpd activation and fluid regulation after cardiac injury.

Similar content being viewed by others

Data availability

RNA-seq and ATAC-seq data are deposited in the NCBI GEO database under accession nos. GSE158079 and GSE193630. The brain, kidney, heart, muscle and kidney H3K27ac, H3K4me3, WGBS and the deep sequencing of brain Hi-C data are downloaded from GEO: GSE13405534. Other heart-related ChIP-seq data were downloaded from GSE818624, GSE7589431 and GSE9692840. Unique reagents generated in this study, and all data supporting the findings of this study, are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. King, R. S. & Newmark, P. A. The cell biology of regeneration. J. Cell Biol. 196, 553–562 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Lai, S.-L., Marín-Juez, R. & Stainier, D. Y. R. Immune responses in cardiac repair and regeneration: a comparative point of view. Cell. Mol. Life Sci. 76, 1365–1380 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. González-Rosa, J. M., Burns, C. E. & Burns, C. G. Zebrafish heart regeneration: 15 years of discoveries. Regeneration 4, 105–123 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Poss, K. D., Wilson, L. G. & Keating, M. T. Heart regeneration in zebrafish. Science 298, 2188–2190 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Schefold, J. C., Filippatos, G., Hasenfuss, G., Anker, S. D. & Von Haehling, S. Heart failure and kidney dysfunction: epidemiology, mechanisms and management. Nat. Rev. Nephrol. 12, 610–623 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Legrand, M. & Rossignol, P. Cardiovascular consequences of acute kidney injury. N. Engl. J. Med. 382, 2238–2247 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Van Biesen, W. & Vanholder, R. in Cardiovascular Complications of Acute Kidney Injury 221–227 (Springer, 2010).

  9. Rodgers, J. T. et al. mTORC1 controls the adaptive transition of quiescent stem cells from G0 to GAlert. Nature 510, 393–396 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rodgers, J. T., Schroeder, M. D., Ma, C. & Rando, T. A. HGFA is an injury-regulated systemic factor that induces the transition of stem cells into GAlert. Cell Rep. 19, 479–486 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Halme, A., Cheng, M. & Hariharan, I. K. Retinoids regulate a developmental checkpoint for tissue regeneration in Drosophila. Curr. Biol. 20, 458–463 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hirose, K. et al. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 364, 184–188 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nachtrab, G., Czerwinski, M. & Kenneth Sexually dimorphic fin regeneration in zebrafish controlled by androgen/GSK3 signaling. Curr. Biol. 21, 1912–1917 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kang, J., Nachtrab, G. & Kenneth Local Dkk1 crosstalk from breeding ornaments impedes regeneration of injured male zebrafish fins. Dev. Cell 27, 19–31 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Kikuchi, K. et al. Primary contribution to zebrafish heart regeneration by gata4+ cardiomyocytes. Nature 464, 601–605 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wang, J. et al. The regenerative capacity of zebrafish reverses cardiac failure caused by genetic cardiomyocyte depletion. Development 138, 3421–3430 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Arif, M. et al. Integrative transcriptomic analysis of tissue-specific metabolic crosstalk after myocardial infarction. eLife 10, e66921 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Arguello, A. A. et al. CCAAT enhancer binding protein plays an essential role in memory consolidation and reconsolidation. J. Neurosci. 33, 3646–3658 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. De Heredia, L. L. & Magoulas, C. Lack of the transcription factor C/EBPδ impairs the intrinsic capacity of peripheral neurons for regeneration. Exp. Neurol. 239, 148–157 (2013).

    Article  CAS  Google Scholar 

  20. Valente, T. et al. CCAAT/enhancer binding protein δ regulates glial proinflammatory gene expression. Neurobiol. Aging 34, 2110–2124 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Kelicen, P. & Tindberg, N. Lipopolysaccharide induces CYP2E1 in astrocytes through MAP kinase kinase-3 and C/EBPβ and -δ. J. Biol. Chem. 279, 15734–15742 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Huang, G. N. et al. C/EBP transcription factors mediate epicardial activation during heart development and injury. Science 338, 1599–1603 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gemberling, M., Karra, R., Dickson, A. L. & Poss, K. D. Nrg1 is an injury-induced cardiomyocyte mitogen for the endogenous heart regeneration program in zebrafish. eLife 4, e05871 (2015).

    Article  PubMed Central  Google Scholar 

  24. Miller, W. L. Fluid volume overload and congestion in heart failure. Circ. Heart Fail. 9, e002922 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Dupont, M., Mullens, W. & Tang, W. H. W. Impact of systemic venous congestion in heart failure. Curr. Heart Fail. Rep. 8, 233–241 (2011).

    Article  PubMed  Google Scholar 

  26. Hentschel, D. M. et al. Acute renal failure in zebrafish: a novel system to study a complex disease. Am. J. Physiol. Ren. Physiol. 288, F923–F929 (2005).

    Article  CAS  Google Scholar 

  27. Lepilina, A. et al. A dynamic epicardial injury response supports progenitor cell activity during zebrafish heart regeneration. Cell 127, 607–619 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. McCampbell, K. K., Springer, K. N. & Wingert, R. A. Analysis of nephron composition and function in the adult zebrafish kidney. J. Vis. Exp. 9, e51644 (2014).

    Google Scholar 

  29. Thangaraju, M. et al. c/ebpδ Null mouse as a model for the double knock-out of slc5a8 and slc5a12 in kidney. J. Biol. Chem. 281, 26769–26773 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Harris, R. E., Setiawan, L., Saul, J. & Hariharan, I. K. Localized epigenetic silencing of a damage-activated WNT enhancer limits regeneration in mature Drosophila imaginal discs. eLife 5, e11588 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Goldman, J. A. & Poss, K. D. Gene regulatory programmes of tissue regeneration. Nat. Rev. Genet. 21, 511–525 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yang, H. et al. A map of cis-regulatory elements and 3D genome structures in zebrafish. Nature 588, 337–343 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sung, M.-H., Baek, S. & Hager, G. L. Genome-wide footprinting: ready for prime time? Nat. Methods 13, 222–228 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ko, C. Y., Chang, W. C. & Wang, J. M. Biological roles of CCAAT/Enhancer-binding protein delta during inflammation. J. Biomed. Sci. 22, 6 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lerch, J. K. Stress increases peripheral axon growth and regeneration through glucocorticoid receptor-dependent transcriptional programs. eNeuro 4, ENEURO.0246-17.2017 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Fuller, P. J. et al. Molecular evolution of the switch for progesterone and spironolactone from mineralocorticoid receptor agonist to antagonist. Proc. Natl Acad. Sci. USA 116, 18578–18583 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Katsu, Y., Oka, K. & Baker, M. E. Evolution of human, chicken, alligator, frog and zebrafish mineralocorticoid receptors: allosteric influence on steroid specificity. Sci. Signal. 11, eaao1520 (2018).

    Article  PubMed  CAS  Google Scholar 

  40. Ben-Yair, R. et al. H3K27me3-mediated silencing of structural genes is required for zebrafish heart regeneration. Development 146, dev178632 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wang, Y. & Nicholas Advances and applications of single-cell sequencing technologies. Mol. Cell 58, 598–609 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Thompson, J. D. et al. Identification and requirements of enhancers that direct gene expression during zebrafish fin regeneration. Development 147, dev191262 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Vizcaya-Molina, E. et al. Damage-responsive elements in Drosophila regeneration. Genome Res. 28, 1852–1866 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Huang, W. C. et al. Treatment of glucocorticoids inhibited early immune responses and impaired cardiac repair in adult zebrafish. PLoS ONE 8, e66613 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Cutie, S., Payumo, A. Y., Lunn, D. & Huang, G. N. In vitro and in vivo roles of glucocorticoid and vitamin D receptors in the control of neonatal cardiomyocyte proliferative potential. J. Mol. Cell. Cardiol. 142, 126–134 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hirose, K. et al. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 364, 184–188 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ullman, M., Ullman, A., Sommerland, H., Skottner, A. & Oldfors, A. Effects of growth hormone on muscle regeneration and IGF-I concentration in old rats. Acta Physiol. Scand. 140, 521–525 (1990).

    Article  CAS  PubMed  Google Scholar 

  49. Lopez, J. et al. Growth hormone improves nerve regeneration, muscle re-innervation, and functional outcomes after chronic denervation injury. Sci. Rep. 9, 3117 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Elabd, C. et al. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 5, 4082 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Girard, C., Eychenne, B., Schweizer-Groyer, G. & Cadepond, F. Mineralocorticoid and glucocorticoid receptors in sciatic nerve function and regeneration. J. Steroid Biochem. Mol. Biol. 122, 149–158 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Nelson, C. M. et al. Glucocorticoids target ependymal glia and inhibit repair of the injured spinal cord. Front. Cell Dev. Biol. 7, 56 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Marshall, L. N. et al. Stage-dependent cardiac regeneration in Xenopus is regulated by thyroid hormone availability. Proc. Natl Acad. Sci. USA 116, 3614–3623 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gibbs, K. M., Chittur, S. V. & Szaro, B. G. Metamorphosis and the regenerative capacity of spinal cord axons in Xenopus laevis. Eur. J. Neurosci. 33, 9–25 (2011).

    Article  PubMed  Google Scholar 

  55. Chávez-Delgado, M. E. et al. Facial nerve regeneration through progesterone-loaded chitosan prosthesis. A preliminary report. J. Biomed. Mater. Res. B Appl. Biomater. 67B, 702–711 (2003).

    Article  CAS  Google Scholar 

  56. Routley, C. E. & Ashcroft, G. S. Effect of estrogen and progesterone on macrophage activation during wound healing. Wound Repair Regeneration 17, 42–50 (2009).

    Article  PubMed  Google Scholar 

  57. Herrera-Rincon, C. et al. Brief local application of progesterone via a wearable bioreactor induces long-term regenerative response in adult Xenopus hindlimb. Cell Rep. 25, 1593–1609 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Adán, N. et al. Prolactin promotes cartilage survival and attenuates inflammation in inflammatory arthritis. J. Clin. Invest. 123, 3902–3913 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Moreno-Carranza, B. et al. Prolactin promotes normal liver growth, survival and regeneration in rodents: effects on hepatic IL-6, suppressor of cytokine signaling-3, and angiogenesis. Am. J. Physiol. Regul. Integr. Comp. Physiol. 305, R720–R726 (2013).

    Article  CAS  PubMed  Google Scholar 

  60. Becker, T., Wullimann, M. F., Becker, C. G., Bernhardt, R. R. & Schachner, M. Axonal regrowth after spinal cord transection in adult zebrafish. J. Comp. Neurol. 377, 577–595 (1997).

    Article  CAS  PubMed  Google Scholar 

  61. Montague, T. G., Cruz, J. M., Gagnon, J. A., Church, G. M. & Valen, E. CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing. Nucleic Acids Res. 42, W401–W407 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Fivaz, J., Bassi, M. C., Pinaud, S. & Mirkovitch, J. RNA polymerase II promoter-proximal pausing upregulates c-fos gene expression. Gene 255, 185–194 (2000).

    Article  CAS  PubMed  Google Scholar 

  63. Traver, D. et al. Transplantation and in vivo imaging of multilineage engraftment in zebrafish bloodless mutants. Nat. Immunol. 4, 1238–1246 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Landt, S. G. et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Jiang, M., Anderson, J., Gillespie, J. & Mayne, M. uShuffle: a useful tool for shuffling biological sequences while preserving the k-let counts. BMC Bioinformatics 9, 192 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Mayor, C. et al. VISTA: visualizing global DNA sequence alignments of arbitrary length. Bioinformatics 16, 1046–1047 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Wang, Y. et al. The 3D Genome Browser: a web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol. 19, 151 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Sun, F., Shoffner, A. R. & Poss, K. D. A genetic cardiomyocyte ablation model for the study of heart regeneration in zebrafish. Methods Mol. Biol. 2158, 71–80 (2021).

    Article  CAS  PubMed  Google Scholar 

  69. Plaut, I. Effects of fin size on swimming performance, swimming behaviour and routine activity of zebrafish Danio rerio. J. Exp. Biol. 203, 813–820 (2000).

    Article  CAS  PubMed  Google Scholar 

  70. Lee, Y., Grill, S., Sanchez, A., Murphy-Ryan, M. & Poss, K. D. Fgf signaling instructs position-dependent growth rate during zebrafish fin regeneration. Development 132, 5173–5183 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Duke Zebrafish Core for animal care, Duke Center for Genomic and Computational Biology for advice, A. Dickson and K. Oonk for assistance with ISH, J. Kang, Y. Diao, J. A. Goldman, M. Pronobis, V. Cigliola, R. Yan, K. Ando, L. Slota-Burtt and R. Karra for comments on the manuscript and Y. Diao, J. Rawls, B. Black and N. Bursac for discussions. We acknowledge research support from NIH (R01 HL155607 to J.C.; R35 GM 124820 to F.Y.; R35 HL150713 and R01 HL136182 to K.D.P.) and from AHA and Fondation Leducq to K.D.P.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization was provided by F.S. and K.D.P., wet lab investigations by F.S., A.R.S., J.C. and A.S., bioinformatic analysis by J.O., L.S., Y.L., H.Y., F.Y. and G.E.C., writing of the manuscript by F.S. and K.D.P., funding acquisition by K.D.P., G.E.C. and F.Y., and supervision by K.D.P., G.E.C. and F.Y.

Corresponding author

Correspondence to Kenneth D. Poss.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Josep Saura and the other, anonymous, reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Bioinformatic analysis of brain RNA-seq during cardiac regeneration.

a, Gene Ontology analysis demonstrating biological pathways with gene enrichment. p < 0.05. Counts indicate the number of genes with significantly changed expression in each biological pathway. b, HOMER analysis demonstrating enriched transcription factor binding motifs at the promoter regions (upstream 2 kb to downstream 500 bp) of genes with increased brain RNA levels during heart regeneration.

Extended Data Fig. 2 Expression of cebpd is not induced in several tissues in adult zebrafish during heart regeneration.

ISH on sections of nasal epithelium, spleen, liver, skeletal muscle, and intestine, indicating that cebpd expression is not noticeably induced in these tissues during heart regeneration. Tissues were harvested 7 days after tamoxifen administration, which induced CM ablation in CreER + animals. n = 10 animals for all groups. Scale bars: 100 µm.

Extended Data Fig. 3 Cardiac muscle regeneration and fluid regulation in cebpd mutant animals.

a, Representative sections of cardiac ventricles 30 days after resection (dpa) in wild-type and cebpd-/- animals, stained with AFOG. Scale bar: 100 µm. n reported in (b). b, Semiquantitative assessment of cardiac injuries based on muscle and scar morphology, indicating no significant difference between cebpd-/- animals and their wild-type siblings. 1: Robust regeneration. 2: Partial regeneration 3: Blocked regeneration. A single trial was performed, with n = 11 animals for each group. Data were analysed using a Fisher’s exact test. c, Sections of 7 days post resection (dpa) ventricles staining with markers for CM nuclei (Mef2; green) and cell cycle entry (EdU; red). Dashed lines outline the approximate resection injury site. Scale bars: 100 µm. d, Quantification of the CM cycling index at 7 dpa indicating no significant differences in cebpd knockout animals compared to their wild-type siblings. A single trial was performed with n = 8 cebpd+/+animals, 9 cebpd+/− animals, and 12 cebpd−/− animals. Mean ± s.d.. Data were analysed using an unpaired two-tailed Student’s t-test. e, Fluorescence images of tailfin vasculature indicating that significantly less fluorescent dextran was able to reach the circulatory system of cebpd-/- fish after cardiac injuries. Data are quantified in Fig. 3. n = 16 CreER- and 21 CreER + animals for cebpd+/+, and n = 22 CreER- and 18 CreER + animals for cebpd-/-. Scale bars: 500 µm.

Source data

Extended Data Fig. 4 Transcriptome profiling of cebpd mutant brain and kidney during heart regeneration.

a,b, Volcano plot showing differential gene expression in the cebpd-/- brain (a) and kidney (b) during heart regeneration. 521 genes (158 genes increased and 363 genes decreased) and 1662 genes (787 increased and 875 decreased) were differentially regulated in the brain and kidney of cebpd-/- animals, respectively. Full lists of differentially expressed genes are shown in Supplementary Tables 2 and 3. Pink dots: highlighted genes. Blue dots: genes with decreased RNA levels (p < 0.05, FC < −1.2). Grey dots: genes with no significant changes. Orange dots: genes with increased RNA levels (p < 0.05, FC > 1.2). c, RNA-seq browser track of aqp7 showing decreased transcript levels of aqp7 (Log2FC = −1.062, p = 0.0012) in cebpd-/- kidney during heart regeneration. d, RNA-seq browser track of aqp10a showing decreased transcript levels of aqp10a (Log2FC = −1.045, p = 0.0020) in cebpd-/- kidney during heart regeneration. e, RNA-seq browser track of slc5a8l showing decreased transcript levels of slc5a8l (Log2FC = −0.515, p = 0.0034) in cebpd-/- kidney during heart regeneration. f,g, Gene Ontology analysis of differentially expressed genes in cebpd-/- brain during heart regeneration demonstrating top biological pathways (f) and molecular functions (g) with gene enrichment. Counts indicate the number of genes with significantly changed expression in Gene Ontology term. h,i, Gene Ontology analysis of differentially expressed genes in cebpd-/- kidney during heart regeneration demonstrating top biological pathways (h) and molecular functions (i) with gene enrichment. Counts indicate the number of genes with significantly changed expression in Gene Ontology term.

Extended Data Fig. 5 Two BAC sequences direct distinct gene expression patterns in zebrafish.

a, Two BAC transgenic lines show distinct larval EGFP fluorescence patterns. Boxed area is magnified on bottom left, and a magnified dorsal view is shown on bottom right. 108cebpd12:EGFP is prominent in skeleton and strong in jaw, indicated by yellow arrows. 35cebpd84:EGFP is weak in skin with low expression in jaw, indicated by yellow arrows. Scale bars: 500 µm. This expression is consistent in all animals used in this study. b, Two BAC transgenic lines show distinct adult EGFP fluorescence patterns. 108cebpd12:EGFP is strong in caudal fin rays and nasal cavity, indicated by yellow arrows. 35cebpd84:EGFP is weak in skin, indicated by yellow arrows. Scale bars: 500 µm. This expression is consistent in all animals used in this study.

Extended Data Fig. 6 Transcriptome and epigenetic analysis of whole kidney marrow.

a, Schematic of CM ablation and whole kidney marrow (WKM) collection for RNA-seq and ATAC-seq. b, Volcano plot showing differential gene expression in WKM after cardiac injury. Blue dots: genes with decreased RNA levels (p < 0.05, FC < −1.2). Grey dots: genes without significant changes. Orange dots: genes with increased RNA levels (p < 0.05, FC > 1.2). c, Gene ontology analysis of top biological pathways with gene enrichment. p < 0.05. Counts indicate the number of genes with significantly changed expression in each biological pathway. d, Heatmap of chromatin regions with changes in accessibility in the brain and kidney after CM ablation. p < 0.05. e, Heatmaps of RNA-seq and ATAC-seq data representing putative enhancer elements linked to genes with significant transcriptional changes in WKM after a cardiac injury. f, BiFET analysis indicating enriched transcription factor binding to open regions in WKM chromatin regions cardiac injury. Red: p < 0.05. Blue: p ≥ 0.05.

Extended Data Fig. 7 Epigenetic features of CEN and its sequence conservation across species.

a, A/B compartment analysis indicates a chromatin B to A compartment switch at cebpd and CEN loci. Dashed lines indicate cebpd and CEN region. b, Browser tracks indicating assays from this study and others. Brain ATAC-seq and Brain RNA-seq, uninjured and 7 days after CM ablation (7 dpi; this study); CM H3.3 occupancy, uninjured and 7 dpi (GSE81862); H3K27Ac occupancy, uninjured and 7 dpi (GSE75894); H3K27me3 and H3K4me3 occupancy in ventricular Gata4+ CMs, uninjured and 5 days post resection (GSE96928). Dashed lines indicate CEN. c, mVista plot of genomic regions around cebpd indicating high conservation of zebrafish CEN with cyprinid fish and low conservation with amphibians and mammals. Calculation window = 100 bp, conservation identity = 70%.

Extended Data Fig. 8 Expression of corticosteroid receptors in the brain and kidney.

a,b, RNA-seq browser track of nr3c1 (a) and nr3c2 (b) indicating that expression of nr3c1 in both brain and whole kidney marrow (WKM) are not significantly changed during heart regeneration, indicated as ‘injured’ in tracks. c, ISH on sections of brain and kidney from uninjured animals demonstrating nr3c1 and nr3c2 expression in multiple regions including ependymal cells and renal tubules. n = 5 animals for each group. Scale bar: 100 µm.

Extended Data Fig. 9 CEN is required for fluid homeostasis during heart regeneration.

Fluorescence images of tailfin vasculature indicating that less fluorescent dextran transferred to the circulatory system of CEN−/− fish after cardiac injuries. A single trial with n = 9 animals for both groups of CEN+/+, and n = 11 CreER- and n = 10 CreER + animals for CEN−/− animals, was performed. Data are quantified in Fig. 6. Scale bars: 500 µm.

Extended Data Fig. 10 Model describing CEN element functions in local and remote tissues.

(Top) Cardiac injury induces cebpd expression locally, regulating epicardial activation during heart regeneration. CEN is sufficient but not required for directing local cebpd induction, likely due to existence of redundant enhancers. (Bottom) Cardiac injury leads to transcriptional activation by bound corticosteroid receptors in remote tissues. CEN exists in an open, permissive structure, topologically close to its promoter and poised for corticosteroid receptor binding. Ligand-bound corticosteroid receptors trigger CEN-directed cebpd expression in distant tissues, contributing to fluid homeostasis in animals undergoing injury-induced regeneration.

Supplementary information

Reporting Summary.

Peer Review File.

Supplementary Tables

Supplementary Table 1 Differentially expressed transcripts from brain RNA-seq after cardiomyocyte ablation (FC > 1.2, P < 0.05). Two-sided Wald significance test performed with DESeq2. Supplementary Table 2 Differentially expressed transcripts from RNA-seq of wild-type and cebpd−/− brains after cardiomyocyte ablation (FC > 1.2, P < 0.05). Two-sided Wald significance test performed with DESeq2. Supplementary Table 3 Differentially expressed transcripts from RNA-seq of wild-type and cebpd−/− kidneys after cardiomyocyte ablation (FC > 1.2, P < 0.05). Two-sided Wald significance test performed with DESeq2. Supplementary Table 4 Differentially accessible chromatin regions from brain ATAC-seq after cardiomyocyte ablation (P < 0.05). Two-sided Wald significance test performed with DESeq2. Supplementary Table 5 Differentially expressed transcripts from whole kidney marrow RNA-seq after cardiomyocyte ablation (FC > 1.2, P < 0.05). Two-sided Wald significance test performed with DESeq2. Supplementary Table 6 Differentially accessible chromatin regions from whole kidney marrow ATAC-seq after cardiomyocyte ablation (P < 0.05). Supplementary Table 7 BiFET analysis of transcription factor binding sites in open regions of whole kidney marrow chromatin samples. MACS2-called peaks were analysed based on the hypergeometric distribution and P values were adjusted by the Benjamin–Hochberg procedure. Supplementary Table 8 Differential ATAC-seq chromatin regions paired with differentially expressed genes in the whole kidney marrow after cardiomyocyte ablation. Supplementary Table 9 BiFET analysis of transcription factor binding sites in open regions of brain chromatin samples. MACS2-called peaks were analysed based on the hypergeometric distribution and P values were adjusted by the Benjamin–Hochberg procedure. Supplementary Table 10 Differential ATAC-seq chromatin regions paired with differentially expressed genes in the brain after cardiomyocyte ablation. Supplementary Table 11 Footprint analysis of enriched transcription factor occupancy in brain during heart regeneration. Two-sided Fisher’s exact test. Supplementary Table 12 Summary of reagents and strains. a, List of experimental models. b, List of antibodies. c, List of chemicals. d, List of recombinant DNA. e, List of primers for generating ISH probes. f, List of primers for qPCR. g, List of software and algorithms.

Source data

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, F., Ou, J., Shoffner, A.R. et al. Enhancer selection dictates gene expression responses in remote organs during tissue regeneration. Nat Cell Biol 24, 685–696 (2022). https://doi.org/10.1038/s41556-022-00906-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00906-y

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