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:

Permissive epigenomes endow reprogramming competence to transcriptional regulators

Abstract

Identifying molecular and cellular processes that regulate reprogramming competence of transcription factors broadens our understanding of reprogramming mechanisms. In the present study, by a chemical screen targeting major epigenetic pathways in human reprogramming, we discovered that inhibiting specific epigenetic roadblocks including disruptor of telomeric silencing 1-like (DOT1L)-mediated H3K79/K27 methylation, but also other epigenetic pathways, catalyzed by lysine-specific histone demethylase 1A, DNA methyltransferases and histone deacetylases, allows induced pluripotent stem cell generation with almost all OCT factors. We found that simultaneous inhibition of these pathways not only dramatically enhances reprogramming competence of most OCT factors, but in fact enables dismantling of species-dependent reprogramming competence of OCT6, NR5A1, NR5A2, TET1 and GATA3. Harnessing these induced permissive epigenetic states, we performed an additional screen with 98 candidate genes. Thereby, we identified 25 transcriptional regulators (OTX2, SIX3, and so on) that can functionally replace OCT4 in inducing pluripotency. Our findings provide a conceptual framework for understanding how transcription factors elicit reprogramming in dependency of the donor cell epigenome that differs across species.

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: Removing DOT1L-mediated H3K79 methylation enables reprogramming with OCT7, OCT8 and OCT9.
Fig. 2: Reduction of H3K27me1/2 through DOT1L inhibition contributes to reprogramming competence of OCT factors.
Fig. 3: Inhibiting multiple epigenetic pathways augments reprogramming competence of OCT factors in humans.
Fig. 4: Inhibiting multiple epigenetic pathways augments reprogramming competence of OCT factors in mice.
Fig. 5: A permissive epigenomic state enables reprogramming with a broad range of transcriptional regulators.
Fig. 6: Positive regulation of SIX3 and OTX2 in reprogramming.

Similar content being viewed by others

Data availability

Microarray, RNA-seq and ChIP-seq data have been deposited in the GEO under accession nos. GSE95608, GSE93706 and GSE149017. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    CAS  PubMed  Google Scholar 

  2. Adachi, K. & Scholer, H. R. Directing reprogramming to pluripotency by transcription factors. Curr. Opin. Genet. Dev. 22, 416–422 (2012).

    CAS  PubMed  Google Scholar 

  3. Apostolou, E. & Hochedlinger, K. Chromatin dynamics during cellular reprogramming. Nature 502, 462–471 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Buganim, Y., Faddah, D. A. & Jaenisch, R. Mechanisms and models of somatic cell reprogramming. Nat. Rev. Genet. 14, 427–439 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Cacchiarelli, D. et al. Integrative analyses of human reprogramming reveal dynamic nature of induced pluripotency. Cell 162, 412–424 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. dos Santos, R. L. et al. MBD3/NuRD facilitates induction of pluripotency in a context-dependent manner. Cell Stem Cell 15, 102–110 (2014).

    PubMed  PubMed Central  Google Scholar 

  7. Ebrahimi, A. et al. Bromodomain inhibition of the coactivators CBP/EP300 facilitate cellular reprogramming. Nat. Chem. Biol. 15, 519–528 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Mali, P. et al. Butyrate greatly enhances derivation of human induced pluripotent stem cells by promoting epigenetic remodeling and the expression of pluripotency-associated genes. Stem Cells 28, 713–720 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Onder, T. T. et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature 483, 598–602 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Rais, Y. et al. Deterministic direct reprogramming of somatic cells to pluripotency. Nature 502, 65–70 (2013).

    CAS  PubMed  Google Scholar 

  11. Zhang, Z., Xiang, D. & Wu, W. S. Sodium butyrate facilitates reprogramming by derepressing OCT4 transactivity at the promoter of embryonic stem cell-specific miR-302/367 cluster. Cell Reprogram. 16, 130–139 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. He, X. et al. Expression of a large family of POU-domain regulatory genes in mammalian brain development. Nature 340, 35–41 (1989).

    CAS  PubMed  Google Scholar 

  13. Rosenfeld, M. G. POU-domain transcription factors: pou-er-ful developmental regulators. Genes Dev. 5, 897–907 (1991).

    CAS  PubMed  Google Scholar 

  14. Schöler, H. R., Hatzopoulos, A. K., Balling, R., Suzuki, N. & Gruss, P. A family of octamer-specific proteins present during mouse embryogenesis: evidence for germline-specific expression of an Oct factor. EMBO J. 8, 2543–2550 (1989).

    PubMed  PubMed Central  Google Scholar 

  15. Jerabek, S. et al. Changing POU dimerization preferences converts Oct6 into a pluripotency inducer. EMBO Rep. 18, e201642958 (2016).

  16. Maekawa, M. et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature 474, 225–229 (2011).

    CAS  PubMed  Google Scholar 

  17. Malik, V. et al. Pluripotency reprogramming by competent and incompetent POU factors uncovers temporal dependency for Oct4 and Sox2. Nat. Commun. 10, 3477 (2019).

    PubMed  PubMed Central  Google Scholar 

  18. Nakagawa, M. et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 26, 101–106 (2008).

    CAS  PubMed  Google Scholar 

  19. Esch, D. et al. A unique Oct4 interface is crucial for reprogramming to pluripotency. Nat. Cell Biol. 15, 295–301 (2013).

    CAS  PubMed  Google Scholar 

  20. Jin, W. et al. Critical POU domain residues confer Oct4 uniqueness in somatic cell reprogramming. Sci. Rep. 6, 20818 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Soufi, A., Donahue, G. & Zaret, K. S. Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell 151, 994–1004 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Aasen, T. et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat. Biotechnol. 26, 1276–1284 (2008).

    CAS  PubMed  Google Scholar 

  24. Blelloch, R. et al. Reprogramming efficiency following somatic cell nuclear transfer is influenced by the differentiation and methylation state of the donor nucleus. Stem Cells 24, 2007–2013 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Eminli, S. et al. Differentiation stage determines potential of hematopoietic cells for reprogramming into induced pluripotent stem cells. Nat. Genet. 41, 968–976 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Friedrich, R. P., Schlierf, B., Tamm, E. R., Bosl, M. R. & Wegner, M. The class III POU domain protein Brn-1 can fully replace the related Oct-6 during Schwann cell development and myelination. Mol. Cell Biol. 25, 1821–1829 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Jaegle, M. et al. The POU proteins Brn-2 and Oct-6 share important functions in Schwann cell development. Genes Dev. 17, 1380–1391 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Schreiber, J. et al. Redundancy of class III POU proteins in the oligodendrocyte lineage. J. Biol. Chem. 272, 32286–32293 (1997).

    CAS  PubMed  Google Scholar 

  29. Chang, Y. K. et al. Quantitative profiling of selective Sox/POU pairing on hundreds of sequences in parallel by Coop-seq. Nucleic Acids Res. 45, 832–845 (2017).

    CAS  PubMed  Google Scholar 

  30. Mistri, T. K. et al. Selective influence of Sox2 on POU transcription factor binding in embryonic and neural stem cells. EMBO Rep. 16, 1177–1191 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ferrari, K. J. et al. Polycomb-dependent H3K27me1 and H3K27me2 regulate active transcription and enhancer fidelity. Mol. Cell 53, 49–62 (2014).

    CAS  PubMed  Google Scholar 

  32. Lavarone, E., Barbieri, C. M. & Pasini, D. Dissecting the role of H3K27 acetylation and methylation in PRC2 mediated control of cellular identity. Nat. Commun. 10, 1679 (2019).

    PubMed  PubMed Central  Google Scholar 

  33. Montserrat, N. et al. Reprogramming of human fibroblasts to pluripotency with lineage specifiers. Cell Stem Cell 13, 341–350 (2013).

    CAS  PubMed  Google Scholar 

  34. Shu, J. et al. Induction of pluripotency in mouse somatic cells with lineage specifiers. Cell 153, 963–975 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Shu, J. et al. GATA family members as inducers for cellular reprogramming to pluripotency. Cell Res. 25, 169–180 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, K. et al. Gadd45a is a heterochromatin relaxer that enhances iPS cell generation. EMBO Rep. 17, 1641–1656 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Gao, Y. et al. Replacement of Oct4 by Tet1 during iPSC induction reveals an important role of DNA methylation and hydroxymethylation in reprogramming. Cell Stem Cell 12, 453–469 (2013).

    CAS  PubMed  Google Scholar 

  38. Hu, X. et al. Tet and TDG mediate DNA demethylation essential for mesenchymal-to-epithelial transition in somatic cell reprogramming. Cell Stem Cell 14, 512–522 (2014).

    CAS  PubMed  Google Scholar 

  39. Dunn, S. J., Martello, G., Yordanov, B., Emmott, S. & Smith, A. G. Defining an essential transcription factor program for naive pluripotency. Science 344, 1156–1160 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Takashima, Y. et al. Resetting transcription factor control circuitry toward ground-state pluripotency in human. Cell 158, 1254–1269 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Theunissen, T. W. et al. Systematic identification of culture conditions for induction and maintenance of naive human pluripotency. Cell Stem Cell 15, 471–487 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mise, N. et al. Differences and similarities in the developmental status of embryo-derived stem cells and primordial germ cells revealed by global expression profiling. Genes Cells 13, 863–877 (2008).

    CAS  PubMed  Google Scholar 

  43. Sabour, D. et al. Identification of genes specific to mouse primordial germ cells through dynamic global gene expression. Hum. Mol. Genet. 20, 115–125 (2011).

    CAS  PubMed  Google Scholar 

  44. Sugawa, F. et al. Human primordial germ cell commitment in vitro associates with a unique PRDM14 expression profile. EMBO J. 34, 1009–1024 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Chia, N. Y. et al. A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature 468, 316–320 (2010).

    CAS  PubMed  Google Scholar 

  46. Hernandez, C. et al. Dppa2/4 Facilitate epigenetic remodeling during reprogramming to pluripotency. Cell Stem Cell 23, 396–411 e398 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Heng, J. C. et al. The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell 6, 167–174 (2010).

    CAS  PubMed  Google Scholar 

  48. Buganim, Y. et al. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 150, 1209–1222 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Mai, T. et al. NKX3-1 is required for induced pluripotent stem cell reprogramming and can replace OCT4 in mouse and human iPSC induction. Nat. Cell Biol. 20, 900–908 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Zuber, M. E., Gestri, G., Viczian, A. S., Barsacchi, G. & Harris, W. A. Specification of the vertebrate eye by a network of eye field transcription factors. Development 130, 5155–5167 (2003).

    CAS  PubMed  Google Scholar 

  51. Braam, S. R. et al. Feeder-free culture of human embryonic stem cells in conditioned medium for efficient genetic modification. Nat. Protoc. 3, 1435–1443 (2008).

    CAS  PubMed  Google Scholar 

  52. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    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 

Download references

Acknowledgements

We thank M. Sinn, I. Gelker, H.W. Choi and M. Haustein for technical assistance. We also thank J. Müller-Keuker for creating the graphical abstract. This work was supported by the Max Planck Society.

Author information

Authors and Affiliations

Authors

Contributions

K.K. conceived the study, performed the experiments, interpreted the data and wrote the manuscript. J.Y. and B.S. contributed to the plasmid construction and western blotting. J.B. contributed to the chemical screening. Jonghun K. and D.W.H. contributed to the karyotyping. M.J.A. contributed to the microarray. Johnny K. interpreted the data and wrote the manuscript. G.W. and D.H. contributed to the teratoma and chimera assays. J.C. and P.C. contributed to the ChIP-seq. H.R.S. supervised this study, interpreted the data and wrote the manuscript.

Corresponding author

Correspondence to Hans R. Schöler.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Reprogramming competence of OCT factors in humans.

a, A schematic representation of eight OCT proteins. The numbers indicate the length of amino acids. N-TAD, N-terminal transactivation domain; DBD, DNA-binding domain; C-TAD, C-terminal transactivation domain. The DBD has a bipartite structure with two subdomains, POU-specific domain (red) and POU-homeodomain (blue), which are connected by a linker (green). b, Among eight OCT family members, only OCT4 and OCT6 were reprogramming-competent. P value was 0.0002. Data are presented as mean values ± s.d. (b, n = 3 biologically independent samples). Differences between samples were compared using a two-tailed Student’s t-test (***P < 0.001).

Extended Data Fig. 2 shRNA-mediated DOT1L inhibition enables reprogramming with OCT7, OCT8 and OCT9.

a, Knockdown efficiency of two shRNA targeting DOT1L mRNA was determined by qPCR. The shLuc construct was used as a control. P values were 0.0006, 0.0024 and 0.0009 from left to right. b, shRNA-mediated DOT1L inhibition resulted in a dramatic reduction of H3K79me1/2 levels. c, shRNA-mediated DOT1L inhibition not only dramatically enhanced OCT4- and OCT6-based reprogramming but also enabled iPSC generation in O7SKM-, O8SKM- and O9SKM-transduced cells. P values were 0.0005, 0.007, 0.0046, 0.0011, 0.0037, 0.0038, 0.0267, 0.004, 0.0037, 0.0081, 0.0004, 0.0023, 0.0026, 0.0003, 0.0011 from left to right. Data are presented as mean values ± s.d. (a,c, n = 3 biologically independent samples). Differences between samples were compared using a two-tailed Student’s t-test (***P < 0.001, **P < 0.01, *P < 0.05). The experiments were repeated independently three times with similar results and representative images are shown (b). See Source Data for uncropped blot images.

Source data

Extended Data Fig. 3 SGC0946-mediated DOT1L inhibition facilitates reprogramming process.

a, SGC0946-mediated DOT1L inhibition did not alter transactivation activities of TADs of OCT factors. b, SGC0946-mediated DOT1L inhibition resulted in the activation of LIN28A, NANOG, SALL4 and SOX2 on day 6 of OCT7-, OCT8- and OCT9-based reprogramming (n = 2 biologically independent samples). c, Coverage plots of OCT4FLAG, OCT7FLAG and OCT7FLAG + SGC0946 ChIP-seqs around + /-500bp from the center of OCT4FLAG peaks. Rows represent OCT4FLAG binding sites. d, SGC0946-mediated DOT1L inhibition did not alter levels of the indicated histone marks. e, SGC0946-mediated DOT1L inhibition did not change expression levels of EED, EZH2 and SUZ12. Data are presented as mean values ± s.d. (a, n = 3 biologically independent samples). The experiments were repeated independently three times with similar results and representative images are shown (d,e). See Source Data for uncropped blot images.

Source data

Extended Data Fig. 4 Inhibiting multiple epigenetic pathways enhances human and mouse reprogramming.

a, Inhibition of multiple epigenetic pathways dramatically enhanced reprogramming efficiencies in humans. P values were <0.0001, 0.0003, <0.0001, 0.0004, <0.0001, <0.0001, <0.0001, 0.0035, <0.0001, <0.0001, <0.0001, <0.0001, 0.0001, <0.0001, <0.0001, <0.0001, <0.0001, <0.0001, <0.0001, <0.0001, <0.0001, 0.0004, <0.0001, <0.0001, <0.0001, <0.0001, <0.0001, 0.0004, <0.0001, <0.0001, <0.0001, 0.0003 from left to right. b, Inhibition of multiple epigenetic pathways dramatically enhanced reprogramming efficiencies in mice. P values were 0.0008, 0.0014, 0.0003, <0.0001, 0.0002, 0.0009, 0.0004, <0.0001, 0.0006, <0.0001, <0.0001, 0.0002, <0.0001, <0.0001, <0.0001, <0.0001 from left to right. Data are presented as mean values ± s.d. (a,b, n = 3 biologically independent samples). Differences between samples were compared using a two-tailed Student’s t-test (***P < 0.001, **P < 0.01, *P < 0.05). S, SGC0946; RG, RG-108; SB, SB431542; C, CI-994; RN, RN-1.

Extended Data Fig. 5 Characterization of human iPSC lines that were generated with OCT7, OCT8 and OCT9 under S/SB/RN treatment.

a, Expression of pluripotency makers in iPSC lines. The O7-1 and O7-2 lines were generated with O7SKM; the O8-1 and O8-2 lines were generated with O8SKM; and the O9-1 and O9-2 iPSC lines were generated with O9SKM. DAPI was used as a nuclear counterstain. Scale bars, 500 μm. b, Karyotype analysis demonstrated correct chromosome content and no obvious deletions or duplications in all iPSC lines. c, No OCT4 transgene was detected in iPSC lines generated with other OCT factors. Instead, only transgenes that were used for the generation of the respective iPSC lines were detected in corresponding iPSC lines. d, All iPSC lines displayed gene expression profiles similar to H1 hESCs but distinct from fibroblasts. e, OCT4 and NANOG promoters were methylated in fibroblasts but largely unmethylated in the iPSC lines and H1 hESCs. f, iPSC lines formed teratomas consisting of all three embryonic germ layers, providing evidence for in vivo differentiation potential. Scale bars, 100 μm. The experiments were repeated independently three times with similar results and representative images are shown (a,c,f). See Source Data for uncropped blot images.

Source data

Extended Data Fig. 6 Characterization of mouse iPSC lines that were generated with Oct6, Oct7, Oct8, Oct9 and Oct11 under S/CI/RN treatment.

a, All iPSC lines were stained positive for Oct4-GFP, NANOG and SSEA1. The mO6-1 and mO6-2 lines were generated with O6SKM; the mO7-1 and mO7-2 lines were generated with O7SKM; the mO8-1 and mO8-2 lines were generated with O8SKM; the mO9-1 and mO9-2 lines were generated with O9SKM; and the mO4 line was generated with O4SKM. b, iPSC lines produced chimeras as determined by PCR. C, corresponding iPSC lines were used as positive controls. H2O was used as a negative control. L, DNA ladder. c, Karyotype analysis demonstrated correct chromosome content and no obvious deletions or duplications in the iPSC lines. d, iPSC lines formed teratomas consisting of all three embryonic germ layers, providing evidence for in vivo differentiation potential. e, mO11-1 and mO11-2 lines, which were generated with O11SKM, were stained positive for Oct4-GFP, NANOG and SSEA1. f, mO11-1 and mO11-2 lines formed teratomas. g, All iPSC lines displayed gene expression profiles similar to mESCs but distinct from MEFs. h, No Oct4 transgene was detected in mO11-1 and mO11-2 lines. Instead, Oct11 transgene was integrated in these iPSC lines. i, mO11-1 and mO11-2 lines produced chimeras as determined by PCR. C, the mO11-1 iPSC line was used as a positive control. H2O was used as a negative control. L, DNA ladder. Scale bar, 250 μm (a,e), 100 μm (d,f). The experiments were repeated independently three times with similar results and representative images are shown (a,b,d,e,f,h,i). See Source Data for uncropped blot images.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–8 and Table 1.

Reporting Summary

Supplementary Dataset 1

The list of epigenetic compounds.

Supplementary Dataset 2

The list of candidate genes.

Supplementary Dataset 3

The list of primers.

Supplementary Dataset 4

Compound ID.

Supplementary Data 1

Unprocessed blots for Supplementary Fig. 4a.

Supplementary Data 2

Unprocessed blots for Supplementary Fig. 4b.

Supplementary Data 3

Unprocessed blots for Supplementary Fig. 4c.

Supplementary Data 4

Unprocessed blots for Supplementary Fig. 4d.

Source data

Source Data Fig. 1b

Unprocessed western blots.

Source Data Fig. 1e

Unprocessed western blots.

Source Data Fig. 2f

Unprocessed western blots.

Source Data Fig. 2g

Unprocessed western blots.

Source Data Fig. 3d

Unprocessed gels.

Source Data Fig. 4d

Unprocessed gels.

Source Data Extended Fig. 2b

Unprocessed western blots.

Source Data Extended Fig. 3d

Unprocessed western blots.

Source Data Extended Fig. 3e

Unprocessed western blots.

Source Data Extended Fig. 5c

Unprocessed gels.

Source Data Extended Fig. 6b

Unprocessed gels.

Source Data Extended Fig. 6h

Unprocessed gels.

Source Data Extended Fig. 6i

Unprocessed gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, KP., Choi, J., Yoon, J. et al. Permissive epigenomes endow reprogramming competence to transcriptional regulators. Nat Chem Biol 17, 47–56 (2021). https://doi.org/10.1038/s41589-020-0618-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0618-6

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