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:

ETV2 functions as a pioneer factor to regulate and reprogram the endothelial lineage

Abstract

The vasculature is an essential organ for the delivery of blood and oxygen to all tissues of the body and is thus relevant to the treatment of ischaemic diseases, injury-induced regeneration and solid tumour growth. Previously, we demonstrated that ETV2 is an essential transcription factor for the development of cardiac, endothelial and haematopoietic lineages. Here we report that ETV2 functions as a pioneer factor that relaxes closed chromatin and regulates endothelial development. By comparing engineered embryonic stem cell differentiation and reprogramming models with multi-omics techniques, we demonstrated that ETV2 was able to bind nucleosomal DNA and recruit BRG1. BRG1 recruitment remodelled chromatin around endothelial genes and helped to maintain an open configuration, resulting in increased H3K27ac deposition. Collectively, these results will serve as a platform for the development of therapeutic initiatives directed towards cardiovascular diseases and solid tumours.

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: ETV2 promotes the endothelial program in both MEFs and EBs.
Fig. 2: ETV2 targets nucleosomes during reprogramming.
Fig. 3: ETV2 recruits BRG1 for chromatin remodelling.
Fig. 4: Phased nucleosomes are established surrounding ETV2 peaks.
Fig. 5: Brg1 knockdown results in a significant decrease in cells expressing FLK1 during reprogramming.
Fig. 6: ETV2 requires BRG1 to activate downstream genes during reprogramming.
Fig. 7: Brg1 conditional KO in ES/EBs.

Similar content being viewed by others

Data availability

The scRNA-seq, bulk RNA-seq, ATAC-seq, ChIP-seq and NOMe-seq data of Etv2-induced EB differentiation and MEF reprogramming are deposited at the NCBI Gene Expression Omnibus (GEO) database under accession no. GSE185684 (GSE168521, ChIP-seq of Etv2-induced MEF reprogramming and EB differentiation; GSE168636, ATAC-seq of Etv2-induced MEF reprogramming and EB differentiation; GSE185682, bulk RNA-seq of Etv2-induced EB differentiation; GSE185683, scRNA-seq of Etv2-induced MEFs reprogramming). All data will be made available upon request. All unique materials used in these studies are readily available from the authors or from commercial sources (Supplementary Tables 35). The MEF MNase-seq is from GSE90893. The MEF histone modification ChIP-seq of H3K9me3, H3K27me3, H3K9ac, H3K4me2, H3K4me1 and HDAC1 is from GSE90893. The H3K27ac ChIP-seq in Brg1 KD EB is from GSE71509. The scRNA-seq of iPSC reprogramming is from GSE100344. The scRNA-seq of cardiac fibroblast reprogramming is from GSE98567. The scRNA-seq of neural reprogramming is from GSE67310. The mass spectrometry data are available from GitHub (https://github.com/gongx030/Etv2_pioneer). Source data are provided with this paper.

Code availability

Codes pertaining to important analyses in this study are available from GitHub (https://github.com/gongx030/Etv2_pioneer).

References

  1. Ye, L., Zimmermann, W.-H., Garry, D. J. & Zhang, J. Patching the heart: cardiac repair from within and outside. Circ. Res. 113, 922–932 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Lugano, R., Ramachandran, M. & Dimberg, A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 77, 1745–1770 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Ginsberg, M. et al. Efficient direct reprogramming of mature amniotic cells into endothelial cells by ETS factors and TGFβ suppression. Cell 151, 559–575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Palikuqi, B. et al. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature 585, 426–432 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang, K. et al. Robust differentiation of human pluripotent stem cells into endothelial cells via temporal modulation of ETV2 with modified mRNA. Sci. Adv. 6, eaba7606 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Iwafuchi-Doi, M. & Zaret, K. S. Pioneer transcription factors in cell reprogramming. Genes Dev. 28, 2679–2692 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Gualdi, R. et al. Hepatic specification of the gut endoderm in vitro: cell signaling and transcriptional control. Genes Dev. 10, 1670–1682 (1996).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. Shi, X. et al. Cooperative interaction of Etv2 and Gata2 regulates the development of endothelial and hematopoietic lineages. Dev. Biol. 389, 208–218 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gong, W. Dpath software reveals hierarchicalÿ haemato-endothelial lineages of Etv2 progenitors based on single-cell transcriptome analysis. Nat. Commun. 8, 14362 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Koyano-Nakagawa, N. et al. Etv2 is expressed in the yolk sac hematopoietic and endothelial progenitors and regulates Lmo2 gene expression. Stem Cells 30, 1611–1623 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rasmussen, T. L. et al. Etv2 rescues Flk1 mutant embryoid bodies. Genesis 51, 471–480 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Koyano-Nakagawa, N. et al. Feedback mechanisms regulate Ets variant 2 (Etv2) gene expression and hematoendothelial lineages. J. Biol. Chem. 290, 28107–28119 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shi, X. et al. Foxk1 promotes cell proliferation and represses myogenic differentiation by regulating Foxo4 and Mef2. J. Cell Sci. 125, 5329–5337 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liu, F. et al. Induction of hematopoietic and endothelial cell program orchestrated by ETS transcription factor ER71/ETV2. EMBO Rep. 16, 654–669 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Singh, B. N. et al. A conserved HH-Gli1-Mycn network regulates heart regeneration from newt to human. Nat. Commun. 9, 4237 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Singh, B. N. et al. Etv2-miR-130a-Jarid2 cascade regulates vascular patterning during embryogenesis. PLoS ONE 12, e0189010 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Rasmussen, T. L. et al. VEGF/Flk1 signaling cascade transactivates Etv2 gene expression. PLoS ONE 7, e50103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sumanas, S. & Choi, K. ETS transcription factor ETV2/ER71/Etsrp in hematopoietic and vascular development. Curr. Top. Dev. Biol. 118, 77–111 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Lee, D. et al. ER71 acts downstream of BMP, Notch, and Wnt signaling in blood and vessel progenitor specification. Cell Stem Cell 2, 497–507 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lee, S. et al. Direct reprogramming of human dermal fibroblasts into endothelial cells using ER71/ETV2. Circ. Res. 120, 848–861 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Alexander, J. M. et al. Brg1 modulates enhancer activation in mesoderm lineage commitment. Development 142, 1418–1430 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Griffin, C. T., Curtis, C. D., Davis, R. B., Muthukumar, V. & Magnuson, T. The chromatin-remodeling enzyme BRG1 modulates vascular Wnt signaling at two levels. Proc. Natl Acad. Sci. USA 108, 2282–2287 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bultman, S. et al. A Brg1 null mutation in the mouse reveals functional differences among mammalian SWI/SNF complexes. Mol. Cell 6, 1287–1295 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Griffin, C. T., Brennan, J. & Magnuson, T. The chromatin-remodeling enzyme BRG1 plays an essential role in primitive erythropoiesis and vascular development. Development 135, 493–500 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. King, H. W. & Klose, R. J. The pioneer factor OCT4 requires BRG1 to functionally mature gene regulatory elements in mouse embryonic stem cells. eLife 6, e22631 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Treutlein, B. et al. Dissecting direct reprogramming from fibroblast to neuron using single-cell RNA-seq. Nature 534, 391–395 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Lee, J. et al. Activation of innate immunity is required for efficient nuclear reprogramming. Cell 151, 547–558 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sayed, N. et al. Transdifferentiation of human fibroblasts to endothelial cells. Circulation 131, 300–309 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Chanda, P. K. et al. Nuclear S-nitrosylation defines an optimal zone for inducing pluripotency. Circulation 140, 1081–1099 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Meng, S. et al. Reservoir of fibroblasts promotes recovery from limb ischemia. Circulation 142, 1647–1662 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhou, Y. et al. Comparative gene expression analyses reveal distinct molecular signatures between differentially reprogrammed cardiomyocytes. Cell Rep. 20, 3014–3024 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhou, G., Meng, S., Li, Y., Ghebre, Y. T. & Cooke, J. P. Optimal ROS signaling is critical for nuclear reprogramming. Cell Rep. 15, 919–925 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chronis, C. et al. Cooperative binding of transcription factors orchestrates reprogramming. Cell 168, 442–459.e20 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Guo, H. et al. DNA methylation and chromatin accessibility profiling of mouse and human fetal germ cells. Cell Res. 27, 165–183 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Sekiya, T., Muthurajan, U. M., Luger, K., Tulin, A. V. & Zaret, K. S. Nucleosome-binding affinity as a primary determinant of the nuclear mobility of the pioneer transcription factor FoxA. Genes Dev. 23, 804–809 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen, J. et al. Single-molecule dynamics of enhanceosome assembly in embryonic stem cells. Cell 156, 1274–1285 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Caravaca, J. M. et al. Bookmarking by specific and nonspecific binding of FoxA1 pioneer factor to mitotic chromosomes. Genes Dev. 27, 251–260 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Aydin, B. et al. Proneural factors Ascl1 and Neurog2 contribute to neuronal subtype identities by establishing distinct chromatin landscapes. Nat. Neurosci. 22, 897–908 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kingma, D. P. & Welling, M. Auto-encoding variational Bayes. Preprint at arXiv https://arxiv.org/abs/1312.6114 (2013).

  47. Schep, A. N. et al. Structured nucleosome fingerprints enable high-resolution mapping of chromatin architecture within regulatory regions. Genome Res. 25, 1757–1770 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wapinski, O. L. et al. Rapid chromatin switch in the direct reprogramming of fibroblasts to neurons. Cell Rep. 20, 3236–3247 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Marquez-Vilendrer, S. B., Thompson, K., Lu, L. & Reisman, D. Mechanism of BRG1 silencing in primary cancers. Oncotarget 7, 56153–56169 (2016).

    Article  PubMed  Google Scholar 

  50. Ho, L. et al. An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency. Proc. Natl Acad. Sci. USA 106, 5181–5186 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Durkin, M., Qian, X., Popescu, N. & Lowy, D. Isolation of mouse embryo fibroblasts. Bio Protoc 3, e908 (2013).

    Article  PubMed  Google Scholar 

  52. Caprioli, A. et al. Nkx2-5 represses Gata1 gene expression and modulates the cellular fate of cardiac progenitors during embryogenesis. Circulation 123, 1633–1641 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Singh, B. N. et al. ETV2 (Ets variant transcription factor 2)-Rhoj cascade regulates endothelial progenitor cell migration during embryogenesis. Arterioscler. Thromb. Vasc. Biol. 40, 2875–2890 (2020).

    Article  CAS  PubMed  Google Scholar 

  54. Luger, K., Rechsteiner, T. J. & Richmond, T. J. Preparation of nucleosome core particle from recombinant histones. Methods Enzymol. 304, 3–19 (1999).

    Article  CAS  PubMed  Google Scholar 

  55. Tanaka, Y. et al. Expression and purification of recombinant human histones. Methods 33, 3–11 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Leavitt, T. et al. Prrx1 fibroblasts represent a pro-fibrotic lineage in the mouse ventral dermis. Cell Rep. 33, 108356 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Magli, A. et al. Time-dependent Pax3-mediated chromatin remodeling and cooperation with Six4 and Tead2 specify the skeletal myogenic lineage in developing mesoderm. PLoS Biol. 17, e3000153 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Das, S. et al. Protection of retinal cells from ischemia by a novel gap junction inhibitor. Biochem. Biophys. Res. Commun. 373, 504–508 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tiscornia, G., Singer, O. & Verma, I. M. Production and purification of lentiviral vectors. Nat. Protoc. 1, 241–245 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  62. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lopez, R., Regier, J., Cole, M. B., Jordan, M. I. & Yosef, N. Deep generative modeling for single-cell transcriptomics. Nat. Methods 15, 1053–1058 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2019).

    Article  CAS  Google Scholar 

  66. Choi, K.-D. et al. Identification of the hemogenic endothelial progenitor and its direct precursor in human pluripotent stem cell differentiation cultures. Cell Rep. 2, 553–567 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. 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  CAS  Google Scholar 

  68. 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 

  69. Magli, A. et al. Pax3 cooperates with Ldb1 to direct local chromosome architecture during myogenic lineage specification. Nat. Commun. 10, 2316 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Furlan-Magaril, M. & Recillas-Targa, F. DNA-protein interactions, principles and protocols. Methods Mol. Biol. 1334, 205–218 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Furlan-Magaril, M., Rincón-Arano, H. & Recillas-Targa, F. DNA-protein interactions, principles and protocols, third edition. Methods Mol. Biol. 543, 253–266 (2009).

    Article  CAS  PubMed  Google Scholar 

  72. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Amemiya, H. M., Kundaje, A. & Boyle, A. P. The ENCODE Blacklist: identification of problematic regions of the genome. Sci. Rep. 9, 9354 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Yu, G., Wang, L.-G. & He, Q.-Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Lay, F. D., Kelly, T. K. & Jones, P. A. DNA methylation protocols. Methods Mol. Biol. 1708, 267–284 (2017).

    Article  CAS  Google Scholar 

  77. Statham, A. L., Taberlay, P. C., Kelly, T. K., Jones, P. A. & Clark, S. J. Genome-wide nucleosome occupancy and DNA methylation profiling of four human cell lines. Genom. Data 3, 94–96 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Bailey, T. L. & Machanick, P. Inferring direct DNA binding from ChIP-seq. Nucleic Acids Res. 40, e128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lesluyes, T., Johnson, J., Machanick, P. & Bailey, T. L. Differential motif enrichment analysis of paired ChIP-seq experiments. BMC Genomics 15, 752 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Bailey, T. L. et al. MEME Suite: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–W208 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from the Department of Defense (W81XWH2110606) and Minnesota Regenerative Medicine. We thank N. Koyano, K.-D. Choi and B. N. Singh for technical assistance and discussions. We acknowledge G. R. Crabtree and B. Bruneau for providing the Brg1f/f;ActinCreER ES cells. We acknowledge the University of Minnesota Genomics Center for their technical assistance and the Minnesota Supercomputing Institute for providing computational resources. We recognize C. Faraday for assistance with figure layout and preparation.

Author information

Authors and Affiliations

Authors

Contributions

W.G., S.D., J.E.S.-P. and D.J.G. conceived the project and W.G., S.D., J.E.S.-P., K.S.Z., M.G.G. and D.J.G. wrote the manuscript. W.G., S.D., J.E.S.-P., E.S., N.D., T.A.L. and E.L.-M., designed, performed experiments and analysed the data. K.S.Z., M.G.G. and D.J.G. supervised the project. All authors commented on and edited the final version of the paper.

Corresponding author

Correspondence to Daniel J. Garry.

Ethics declarations

Competing interests

D.J.G. and M.G.G. are co-founders of NorthStar Genomics. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks John Cooke and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Characterization of mouse embryonic fibroblasts that overexpress ETV2 (iHA-Etv2 MEFs) following the addition of doxycycline to reprogram MEFs to endothelial cells.

(a) The gating strategy for iHA-Etv2 MEF FACS characterization is outlined in the five profiles. (b) We isolated embryonic fibroblasts from this mouse line and demonstrated that this cell population uniformly expressed fibroblast markers (Thy1.2, CD44 and CD29). (c) The western blot analysis showed that ETV2 was robustly expressed within 3 hrs post-Dox treatment [representative blots (c) from 3 independent experiments with similar results]. (d-g) ETV2 overexpression resulted in an increase in cells expressing FLK1/TIE2, as measured by FACS (n = 3 biological replicates; *P < 0.05). (h) The biological processes that are significantly associated with the up-regulated genes in cluster 7 (FLK1+ cells at day 7 of reprogramming) compared with cluster 1 (undifferentiated MEFs). (I-m) qPCR experiments showed the increased expression levels of endothelial genes (i) Etv2 (*P = 0.0423, *P = 0.0377, ****P < 1×10-4), (j) Lmo2 (*P = 0.0496), (k) Flk1 (****P < 1×10-4), (l) CD31 (**P = 0.0044) and (m) Tie2 (****P < 1×10-4) at 1 day, 2 days, and 7 days post induction of ETV2, as well as the no Dox control (n = 3 biological replicates; one-way ANOVA with multiple comparison). Data are presented as mean ± SEM.

Source data

Extended Data Fig. 2 Expression of endothelial transcripts in the FLK1+ cell population at day 7 post-ETV2 induction during MEF reprogramming.

(a) The violin plots show the scaled expression levels of endothelial markers such as Etv2, Emcn, Lmo2, Flk1/Kdr, Cdh5 and Sox18 in MEFs, day 1, day 2, day 7 post-ETV2 induction, as well as the FLK1+ cells from day 7. The y-axis indicates the gene expression levels scaled and normalized by Seurat. (b) The violin plots show the scaled expression levels of endothelial markers in seven cell clusters. The y-axis indicates the gene expression levels scaled and normalized by Seurat. The one-sided enrichment test was used to evaluate the significance of pathway enrichment. The p-value adjustment was performed by B-H Procedure. (c) The biological processes that are significantly associated with the up-regulated in genes in cluster 1 (undifferentiated MEFs) compared with the rest of the cell populations. There are 3,562, 948, 2,936, 7,202 and 827 single cells from undifferentiated MEFs, MEFs with day 1, day 2, day 7 post-ETV2 induction, and FLK1+ cell population of day 7 post-ETV2 induction, respectively. (d) GSEA plot indicates significant upregulation of the inflammatory response in MEFs (cluster 1). The y-axis representing the enrichment score (ES) for each gene and x-axis indicates the gene rank in the ordered list. The default GSEA permutation test was used to evaluate the significance of gene set enrichment. No p-value adjustment was performed. (e) Heatmap representing the gene expression levels scaled by Seurat for upregulated (red) and downregulated (blue) genes in cluster 1 and cluster 2. (f-g) The bar plots show top 10 significant pathways for cluster 1 and cluster 2 for MEFs. The y-axis represents the p adjusted values obtained from over-representation test in Gene Ontology enrichment analysis. The one-sided enrichment test was used to evaluate the significance of Gene Ontology enrichment. The p-value adjustment was performed by B-H Procedure.

Extended Data Fig. 3 Expression profile of immune response related genes and significance of pathways in reprogrammed MEFs are upregulated post induction of ETV2.

(a) The UMAP shows expression profiles of Tlr3, Nfkb1 and Vav3, Cd38 and Abl1 (members of B cell receptor signaling pathway) in undifferentiated MEFs and post Etv2 induction day 1, day 2, day 7 and Flk1+ cells at day 7. (b-c) The bar plot shows immune response related pathways significantly upregulated in (b) Flk1+ cells from day 7 and (c) day 1 post Etv2 induction compared to MEFs. The y-axis indicates the p-value showing significance for each pathway obtained from Gene Ontology enrichment. The one-sided enrichment test was used to evaluate the significance of Gene Ontology enrichment. The p-value adjustment was performed by B-H Procedure.

Extended Data Fig. 4 ETV2 overexpression promotes endothelial lineage development in iHA-Etv2 ES/EBs.

(a) Etv2 endogenous expression during differentiation of ES/EBs (n = 3 biological replicates; one-way ANOVA with multiple comparison ***P = 2×10-4). (b-c) FACS analysis of iHA-Etv2 EBs after 3 h (D2.125) or 12 h (D2.5) of the overexpression of ETV2 ( + Dox). Note a significant induction of the endothelial lineage (FLK1+ cells) following 12 h of ETV2 overexpression (n = 3 biological replicates; 2way ANOVA with multiple comparison ****P < 1×10-4). (d-h) qPCR experiments showed increased expression levels of endothelial genes (d) Etv2 (****P < 1×10-4, ***P = 0.0004), (e) Cdh5 (***P = 0.0001), (f) Lmo2 (*P = 0.041, ****P < 1×10-4), (g) Flk1 (***P = 0.0002, ****P < 1×10-4) and (h) Sox18 (***P = 0.0005) following the induction of ETV2 ( + Dox). (n = 3 biological replicates; one-way ANOVA with multiple comparison). Data are presented as mean ± SEM.

Source data

Extended Data Fig. 5 Commonly up- and down-regulated genes in FLK1 + cell populations from ETV2 induced ES/EB differentiation and MEF reprogramming.

(a-b) The Venn diagrams show the overlap of commonly up- and down-regulated genes during EB differentiation and MEF reprogramming. (c-d) Top commonly up- and down-regulated genes during EB differentiation and MEF reprogramming. The y-axis shows the log2 fold change of gene expression between FLK1+ cell populations and the baseline conditions (D2.5 EB in ES/EB differentiation, and undifferentiated MEFs during MEF reprogramming). (e-f) The pathways that are significantly associated with commonly up- and down-regulated genes during ES/EB differentiation and MEF reprogramming are highlighted. The one-sided enrichment test was used to evaluate the significance of Gene Ontology enrichment. The p-value adjustment was performed by B-H Procedure.

Extended Data Fig. 6 Combined RNA-seq and ATAC-seq analysis during EB and MEF reprogramming.

(a) The number of transcription factors whose motifs associated chromatin accessibility were significantly increased or decreased in the FLK1+ cell populations at 12 hours post-Etv2 induction compared with D2.5 EBs. (b) The number of transcription factors whose motifs associated chromatin accessibility were significantly increased or decreased in the FLK1+ cell population at day 7 post-ETV2 induction compared with undifferentiated MEFs. (c-d) The number of transcription factors whose motif-associated chromatin accessibility that were commonly increased or decreased during EB and MEF reprogramming. (e) The transcription factors whose RNA-seq expression levels and motifs associated chromatin accessibility that were both up-regulated or down-regulated during EB reprogramming (FLK1+ cell from EBs at 12 hours post-ETV2 induction vs. day 2.5 EB). (f) The transcription factors whose RNA-seq expression levels and motifs associated chromatin accessibility that were both up-regulated or down-regulated during MEF reprogramming (FLK1+ cell population at day 7 post-ETV2 induction vs. undifferentiated MEFs). (g-h) The commonly up- and down-regulated genes between EBs and MEFs. (i) The transcription factor motifs that are significantly enriched in 5k region surrounding the transcription start sites of the commonly up- and down-regulated genes in EBs and MEFs. The TF enrichment was evaluated by the two-sided χ2 test in chromVAR. Data shown in 6i represent the average of two biological replicates. The p-value adjustment was performed by B-H Procedure.

Extended Data Fig. 7 The ETV2 bound sites at day 1 post-Etv2 induction in MEFs target the nucleosomes and the analysis of ETV2 ChIP-seq peaks during EB and MEF reprogramming.

(a) The MNase-seq, BRG1, H3K27ac, H3, H3K9me3, H3K27me3, H3K9ac, H3K4me3, H4K7me1 and Hdac1 ChIP-seq signals surrounding the ETV2 bound sites at day 1 post-ETV2 induction during MEF reprogramming. The ETV2 binding sites were split into nucleosome and nucleosome free region (NFR) according to the MNase-seq signals in undifferentiated MEFs. Data shown here represent the average of two biological replicates. (b) The latent representation of ATAC-seq V-plots (-320bp to + 320 bp) where the centers are nucleosome free or occupied by mono nucleosome. (c) The aggregated ETV2 bound sites centric V-plot whose centers were occupied by mono nucleosomes or nucleosome free. (d) The fragment size profile of ETV2 bound sites centric region (-320bp to +320 bp) where the centers are nucleosome free or occupied by mono nucleosomes. (e) Motif analysis of ETV2 bound sites during EB reprogramming (3 hours post-ETV2 induction) and MEF reprogramming (24 hours post-ETV2 induction). The table shows the significantly enriched motifs in ETV2 bound sites during EB and MEF reprogramming. (f) The overlap of ETV2 bound sites at day 1, day 2 and day 7 post-ETV2 induction during MEF reprogramming. (g) The overlap of ETV2 bound sites at 3 hours and 12 hours post-ETV2 induction during EB reprogramming. (h) The bar plot shows the percent of genes located near the late, early and sustained ETV2 bound sites related to blood vessel development.

Extended Data Fig. 8 Brg1 knockdown in iHA-Etv2 MEFs using shRNA lentiviral particles.

(a) Schematic diagram of shRNA lentiviral knockdown of Brg1 in iHA-Etv2 MEFs. Briefly, MEFs were exposed to shRNA particles 72 hrs before reprogramming was started and cells were collected for analysis and sequencing at various time points throughout the reprogramming process (D1, D2 and D7). shRNA particles were added throughout the reprogramming process to ensure BRG1 expression was not increased. (b) Western blot analysis of BRG1 expression in iHA-Etv2 MEFs exposed to normal reprogramming media versus media with shRNA particles against Brg1 [representative blots (b) from 3 independent experiments with similar results]. (c-l) Compared to control, shRNA knockdown of (d) Brg1 (****P < 1×10-4) in the context of (c) Etv2 overexpression (****P < 1×10-4) leads to a significant decrease in the expression of (e) Flk1 (****P < 1×10-4, ***P = 0.0005), (f) Hopx (***P = 0.0004, **P = 0.0052), (g) Otor (***P = 0.0001, ***P = 0.0009), (h) Emcn (****P < 1×10-4), (i) Sox18 (****P < 1×10-4, ***P = 0.0003), (j) Lmo2 (****P < 1×10-4), (k) Mmp9 (****P < 1×10-4, ***P = 0.001) and (l) Lax1 (****P < 1×10-4, ***P = 0.0003) transcripts, which are upregulated at D7 following overexpression of ETV2 in iHA-Etv2 MEFs (n = 3 biological replicates; one-way ANOVA with multiple comparison). Data are presented as mean ± SEM.

Source data

Extended Data Fig. 9 Dek, Znhit1 and Cdh8 knockdown does not impact ETV2 mediated endothelial reprogramming.

(a-c) The expression profiles of (a) Dek, (b) Znhit1 and (c) Cdh8 expression during MEF reprogramming. (d-e) iHA-Etv2 MEFs were exposed to Chd8, Dek and Znhit shRNA particles 72 hrs before ETV2 mediated reprogramming was started and cells were collected for analysis 24 hrs (D1) following ETV2 overexpression. Flow cytometry analysis shows that knockdown of Chd8, Dek and Znhit does not affect MEF reprogramming mediated by ETV2 (n = 3 biological replicates). (f-h) qPCR analysis shows efficient knockdown of Chd8 (**P = 0.0068), Dek (**P = 0.0037) and Znhit (**P = 0.0017) in iHA-Etv2 MEFs (n = 3 biological replicates; one-tailed unpaired t test ****P < 1.0×10-4). Data are presented as mean ± SEM.

Source data

Extended Data Fig. 10 ETV2 requires BRG1 to activate downstream genes during reprogramming.

(a) The heatmap shows the ATAC-seq signal surrounding the summit of 12,170 sustained ETV2 ChIP-seq peaks that were present at day 1 and day 7 post-induction of ETV2 in control MEFs (the sustained Etv2 peaks). The ATAC-seq data include undifferentiated MEFs, day 7 post-ETV2 induction, and FLK1+ cells from day 7 post-ETV2 induction in MEFs. We also include the ATAC-seq data from undifferentiated Brg1 KD (knockdown) in MEFs and day 7 post-ETV2 induction with Brg1 KD in MEFs. The sustained ETV2 peaks were divided into two groups: open (red) or closed (black) at day 7 post- induction of Brg1 KD in MEFs. (b) Heatmap shows ETV2 ChIP-seq signal surrounding 4,965 sustained ETV2 ChIP-seq peaks present in the D7 post ETV2 induction in WT MEFs and Brg1 KD MEFs. (c) UCSC genome browser tracks show the ETV2 ChIP-seq signal surrounding Etv2 ChIP-seq peaks at the promoter region of two endothelial genes Rhoj and Kdr. Data shown in 10a and 10b represent the average of two biological replicates.

Supplementary information

Supplementary Information

Supplementary Figs. 1–6.

Reporting Summary

Supplementary Tables 1–5

Supplementary Table 1 The transcription factors whose motif-associated chromatin accessibility and expression were upregulated in both EBs and MEFs following ETV2 induction. Supplementary Table 2 The transcription factors whose motif-associated chromatin accessibility and expression were downregulated in both EBs and MEFs following ETV2 induction. Supplementary Table 3. Taqman qPCR probes. Supplementary Table 4 Re-ChIP primers. Supplementary Table 5 ChIP qPCR primers for ELK3

Source data

Source Data Fig. 3

Unprocessed western blot membranes. (a) Unprocessed GST blot membrane for Fig. 3d. (b) Unprocessed BRG1 blot membrane for Fig. 3e. (c) Unprocessed BRG1 blot membrane for Fig. 3f.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blot membranes. (a) Unprocessed BRG1 blot membrane for Fig. 7b. (b) Unprocessed GAPDH blot membrane for Fig. 7b.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blot membranes. (a) Unprocessed HA blot membrane for Extended Data Fig. 1c. (b) Unprocessed GAPDH blot membrane for Extended Data Fig. 1c.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blot membranes. (a) Unprocessed BRG1 blot membrane for Extended Data Fig. 8b. (b) Unprocessed GAPDH blot membrane for Extended Data Fig. 8b.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gong, W., Das, S., Sierra-Pagan, J.E. et al. ETV2 functions as a pioneer factor to regulate and reprogram the endothelial lineage. Nat Cell Biol 24, 672–684 (2022). https://doi.org/10.1038/s41556-022-00901-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00901-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing