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:

In vitro capture and characterization of embryonic rosette-stage pluripotency between naive and primed states

Abstract

Following implantation, the naive pluripotent epiblast of the mouse blastocyst generates a rosette, undergoes lumenogenesis and forms the primed pluripotent egg cylinder, which is able to generate the embryonic tissues. How pluripotency progression and morphogenesis are linked and whether intermediate pluripotent states exist remain controversial. We identify here a rosette pluripotent state defined by the co-expression of naive factors with the transcription factor OTX2. Downregulation of blastocyst WNT signals drives the transition into rosette pluripotency by inducing OTX2. The rosette then activates MEK signals that induce lumenogenesis and drive progression to primed pluripotency. Consequently, combined WNT and MEK inhibition supports rosette-like stem cells, a self-renewing naive-primed intermediate. Rosette-like stem cells erase constitutive heterochromatin marks and display a primed chromatin landscape, with bivalently marked primed pluripotency genes. Nonetheless, WNT induces reversion to naive pluripotency. The rosette is therefore a reversible pluripotent intermediate whereby control over both pluripotency progression and morphogenesis pivots from WNT to MEK signals.

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: Naive and primed markers in the embryonic rosette.
Fig. 2: WNT and MEK signals couple successive steps in morphogenesis to pluripotency progression.
Fig. 3: WNT controls the initial phase of the naive-primed transition.
Fig. 4: MEK and WNT inhibition maintains rosette characteristics.
Fig. 5: OTX2 enables MEK-driven transition to primed pluripotency.
Fig. 6: A primed chromatin landscape in RSCs.
Fig. 7: RSCs remodel pHC.
Fig. 8: Rosette-specific formatting of pHC.

Similar content being viewed by others

Data availability

Figures 3 and 4 have associated RNA-seq and single-cell RNA-seq data deposited in the GEO and are accessible through GEO series accession number GSE145727. Figure 6 has associated ChIP-seq data deposited in GEO with the accession number GSE112234 . Previously published RNA-seq data that were re-analysed here are available from ArrayExpress under accession codes E-MTAB-2958 and E-MTAB-5147. Previously published ChIP-seq data that were re-analysed here are available from the GEO under accession codes GSE23943 and GSE60204. Source data for Figs. 1, 2 and 48 and Extended Data Figs. 1, 36 and 8 are presented with the paper. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

Code availability

No custom computer code was used in this study.

References

  1. Weinberger, L., Ayyash, M., Novershtern, N. & Hanna, J. H. Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat. Rev. Mol. Cell Biol. 17, 155–169 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Bedzhov, I. & Zernicka-Goetz, M. Self-organizing properties of mouse pluripotent cells initiate morphogenesis upon implantation. Cell 156, 1032–1044 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Shahbazi, M. N. et al. Pluripotent state transitions coordinate morphogenesis in mouse and human embryos. Nature 552, 239–243 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Morgani, S., Nichols, J. & Hadjantonakis, A. K. The many faces of pluripotency: in vitro adaptations of a continuum of in vivo states. BMC Dev. Biol. 17, 7 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Smith, A. Formative pluripotency: the executive phase in a developmental continuum. Development 144, 365–373 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. ten Berge, D. et al. Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells. Nat. Cell Biol. 13, 1070–1075 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tuysuz, N. et al. Lipid-mediated Wnt protein stabilization enables serum-free culture of human organ stem cells. Nat. Commun. 8, 14578 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Yi, F. et al. Opposing effects of Tcf3 and Tcf1 control Wnt stimulation of embryonic stem cell self-renewal. Nat. Cell Biol. 13, 762–770 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wray, J. et al. Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat. Cell Biol. 13, 838–U246 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kemp, C., Willems, E., Abdo, S., Lambiv, L. & Leyns, L. Expression of all Wnt genes and their secreted antagonists during mouse blastocyst and postimplantation development. Dev. Dyn. 233, 1064–1075 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. ten Berge, D. et al. Wnt signaling mediates self-organization and axis formation in embryoid bodies. Cell Stem Cell 3, 508–518 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Wang, Q. T. et al. A genome-wide study of gene activity reveals developmental signaling pathways in the preimplantation mouse embryo. Dev. Cell 6, 133–144 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Albeck, J. G., Mills, G. B. & Brugge, J. S. Frequency-modulated pulses of ERK activity transmit quantitative proliferation signals. Mol. Cell 49, 249–261 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S. & Saitou, M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519–532 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Boroviak, T. et al. Lineage-specific profiling delineates the emergence and progression of naive pluripotency in mammalian embryogenesis. Dev. Cell 35, 366–382 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gardner, R. L., Lyon, M. F., Evans, E. P. & Burtenshaw, M. D. Clonal analysis of X-chromosome inactivation and the origin of the germ line in the mouse embryo. J. Embryol. Exp. Morphol. 88, 349–363 (1985).

    CAS  PubMed  Google Scholar 

  18. Kurek, D. et al. Endogenous WNT signals mediate BMP-induced and spontaneous differentiation of epiblast stem cells and human embryonic stem cells. Stem Cell Reports 4, 114–128 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Masaki, H. et al. Interspecific in vitro assay for the chimera-forming ability of human pluripotent stem cells. Development 142, 3222–3230 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Veluscek, G., Li, Y., Yang, S. H. & Sharrocks, A. D. Jun-mediated changes in cell adhesion contribute to mouse embryonic stem cell exit from ground state pluripotency. Stem Cells 34, 1213–1224 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kalkan, T. et al. Tracking the embryonic stem cell transition from ground state pluripotency. Development 144, 1221–1234 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gardner, R. L. & Cockroft, D. L. Complete dissipation of coherent clonal growth occurs before gastrulation in mouse epiblast. Development 125, 2397–2402 (1998).

    CAS  PubMed  Google Scholar 

  23. Buecker, C. et al. Reorganization of enhancer patterns in transition from naive to primed pluripotency. Cell Stem Cell 14, 838–853 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, S. H. et al. Otx2 and Oct4 drive early enhancer activation during embryonic stem cell transition from naive pluripotency. Cell Rep. 7, 1968–1981 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Acampora, D., Di Giovannantonio, L. G. & Simeone, A. Otx2 is an intrinsic determinant of the embryonic stem cell state and is required for transition to a stable epiblast stem cell condition. Development 140, 43–55 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Habibi, E. et al. Whole-genome bisulfite sequencing of two distinct interconvertible DNA methylomes of mouse embryonic stem cells. Cell Stem Cell 13, 360–369 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Marks, H. et al. The transcriptional and epigenomic foundations of ground state pluripotency. Cell 149, 590–604 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Leitch, H. G. et al. Naive pluripotency is associated with global DNA hypomethylation. Nat. Struct. Mol. Biol. 20, 311–316 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ficz, G. et al. FGF signaling inhibition in ESCs drives rapid genome-wide demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 13, 351–359 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. von Meyenn, F. et al. Impairment of DNA methylation maintenance is the main cause of global demethylation in naive embryonic stem cells. Mol. Cell 62, 848–861 (2016).

    Article  CAS  Google Scholar 

  31. Galonska, C., Ziller, M. J., Karnik, R. & Meissner, A. Ground state conditions induce rapid reorganization of core pluripotency factor binding before global epigenetic reprogramming. Cell Stem Cell 17, 462–470 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Azuara, V. et al. Chromatin signatures of pluripotent cell lines. Nat. Cell Biol. 8, 532–538 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kurimoto, K. et al. Quantitative dynamics of chromatin remodeling during germ cell specification from mouse embryonic stem cells. Cell Stem Cell 16, 517–532 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Tosolini, M. et al. Contrasting epigenetic states of heterochromatin in the different types of mouse pluripotent stem cells. Sci. Rep. 8, 5776 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78, 7634–7638 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156 (1981).

    Article  CAS  PubMed  Google Scholar 

  39. Brons, I. G. et al. Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 448, 191–195 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Tesar, P. J. et al. New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature 448, 196–199 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Lyashenko, N. et al. Differential requirement for the dual functions of β-catenin in embryonic stem cell self-renewal and germ layer formation. Nat. Cell Biol. 13, 753–U365 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Biechele, S., Cockburn, K., Lanner, F., Cox, B. J. & Rossant, J. Porcn-dependent Wnt signaling is not required prior to mouse gastrulation. Development 140, 2961–2971 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Augustin, I. et al. Autocrine Wnt regulates the survival and genomic stability of embryonic stem cells. Sci. Signal. 10, eaah6829 (2017).

    Article  PubMed  CAS  Google Scholar 

  44. Bao, S. et al. Epigenetic reversion of post-implantation epiblast to pluripotent embryonic stem cells. Nature 461, 1292–1295 (2009).

    Article  CAS  PubMed  Google Scholar 

  45. Guo, G. et al. Klf4 reverts developmentally programmed restriction of ground state pluripotency. Development 136, 1063–1069 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Watanabe, A., Yamada, Y. & Yamanaka, S. Epigenetic regulation in pluripotent stem cells: a key to breaking the epigenetic barrier. Philos. Trans. R. Soc. Lond. B Biol. Sci. 368, 20120292 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Messerschmidt, D. M., Knowles, B. B. & Solter, D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev. 28, 812–828 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Becker, J. S., Nicetto, D. & Zaret, K. S. H3K9me3-dependent heterochromatin: barrier to cell fate changes. Trends Genet. 32, 29–41 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Bogdanovic, O. & Lister, R. DNA methylation and the preservation of cell identity. Curr. Opin. Genet. Dev. 46, 9–14 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Saksouk, N. et al. Redundant mechanisms to form silent chromatin at pericentromeric regions rely on BEND3 and DNA methylation. Mol. Cell 56, 580–594 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Chazaud, C., Yamanaka, Y., Pawson, T. & Rossant, J. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2–MAPK pathway. Dev. Cell 10, 615–624 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Chazaud, C. & Yamanaka, Y. Lineage specification in the mouse preimplantation embryo. Development 143, 1063–1074 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Boroviak, T., Loos, R., Bertone, P., Smith, A. & Nichols, J. The ability of inner-cell-mass cells to self-renew as embryonic stem cells is acquired following epiblast specification. Nat. Cell Biol. 16, 516–528 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hoshino, H., Shioi, G. & Aizawa, S. AVE protein expression and visceral endoderm cell behavior during anterior–posterior axis formation in mouse embryos: asymmetry in OTX2 and DKK1 expression. Dev. Biol. 402, 175–191 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Ang, S. L. et al. A targeted mouse Otx2 mutation leads to severe defects in gastrulation and formation of axial mesoderm and to deletion of rostral brain. Development 122, 243–252 (1996).

    CAS  PubMed  Google Scholar 

  56. Chazaud, C. & Rossant, J. Disruption of early proximodistal patterning and AVE formation in Apc mutants. Development 133, 3379–3387 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Moser, A. R. et al. Homozygosity for the Min allele of Apc results in disruption of mouse development prior to gastrulation. Dev. Dyn. 203, 422–433 (1995).

    Article  CAS  PubMed  Google Scholar 

  58. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10, 48 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Supek, F., Bosnjak, M., Skunca, N. & Smuc, T. REVIGO summarizes and visualizes long lists of Gene Ontology terms. PLoS ONE 6, e21800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Okabe, M., Ikawa, M., Kominami, K., Nakanishi, T. & Nishimune, Y. ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett. 407, 313–319 (1997).

    Article  CAS  PubMed  Google Scholar 

  61. Ostermeier, G. C., Wiles, M. V., Farley, J. S. & Taft, R. A. Conserving, distributing and managing genetically modified mouse lines by sperm cryopreservation. PLoS ONE 3, e2792 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Bedzhov, I., Leung, C. Y., Bialecka, M. & Zernicka-Goetz, M. In vitro culture of mouse blastocysts beyond the implantation stages. Nat. Protoc. 9, 2732–2739 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Iacovino, M., Roth, M. E. & Kyba, M. Rapid genetic modification of mouse embryonic stem cells by Inducible Cassette Exchange recombination. Methods Mol. Biol. 1101, 339–351 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kyba, M., Perlingeiro, R. C. & Daley, G. Q. HoxB4 confers definitive lymphoid-myeloid engraftment potential on embryonic stem cell and yolk sac hematopoietic progenitors. Cell 109, 29–37 (2002).

    Article  CAS  PubMed  Google Scholar 

  65. Willert, K. et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448–452 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Muraro, M. J. et al. A single-cell transcriptome atlas of the human pancreas. Cell Syst. 3, 385–394.e3 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-seq. Genome Biol. 17, 77 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Grun, D., Kester, L. & van Oudenaarden, A. Validation of noise models for single-cell transcriptomics. Nat. Methods 11, 637–640 (2014).

    Article  PubMed  CAS  Google Scholar 

  69. 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 

  70. Kroeze, L. I. et al. Characterization of acute myeloid leukemia based on levels of global hydroxymethylation. Blood 124, 1110–1118 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. van de Werken, C. et al. A universal method for sequential immunofluorescent analysis of chromatin and chromatin-associated proteins on chromosome spreads. Chromosome Res. 21, 475–489 (2013).

    Article  PubMed  CAS  Google Scholar 

  72. Grewal, S. I. & Jia, S. Heterochromatin revisited. Nat. Rev. Genet. 8, 35–46 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the following colleagues for providing cell lines used in this study: B. Merrill (Tcf7l1–/–), A. Smith (RGd2), J. Wysocka (Tbx3-RFP;Oct6-GFP), A. Simeone (Otx2KO and Otx2OE) and M. Kyba (A2lox.Cre). We thank M. Muraro from Single Cell Discoveries for help with single-cell sequencing and data analysis, and R. van Albada and A. Korporaal for technical assistance. NWO ECHO.10.B1.064, TI Pharma D5-402, Marie Curie FP7-PEOPLE-2009-RG-256560, ZonMW 116006104 (all to D.t.B.), NWO-VIDI 864.12.007 (to H.M.) and FES NIRM (Dutch Innovation Award) supported this study.

Author information

Authors and Affiliations

Authors

Contributions

A.N., I.E., E.v.G., L.V., D.K., J.L., J.S., M.M. and D.t.B. designed and performed experiments and analysed data. A.N., I.E., D.K., J.L., J.S., R.A.M.D., G.v.M., M.M., M.D., E.B.B., H.M. and D.t.B. prepared the manuscript. J.S., D.t.B., R.A.M.D., G.v.M. and H.M. designed, performed and analysed the ChIP-seq experiments. D.t.B., J.S., A.T.d.D., R.W.W.B. and W.F.J.v.I. performed and analysed the RNA-seq experiments. E.v.G., D.t.B. and N.C.R. performed and analysed the single-cell sequencing experiments. A.N., C.E., E.v.G. and E.B.B. prepared and analysed the chromosome spreads. A.M. assisted with the mouse experiments and Y.G. performed additional experiments. H.M. and J.H.J. designed, performed and analysed nucleoside mass spectrometry analyses. D.t.B. conceived the study and coordinated the work.

Corresponding author

Correspondence to Derk ten Berge.

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 Naive and primed markers in the embryonic rosette.

a,b, Counts of embryos of the indicated stages displaying a rosette or lumen and KLF4 (a) or OTX2 (b) expression status. a, and (b) show different litters. c, Individual colour channels for Fig. 1h (representative of 5 experiments). d, Individual colour channels for Fig. 1i (representative of 8 experiments). Scale bars 20 µm.

Source data

Extended Data Fig. 2 WNT and MEK signals couple successive steps in morphogenesis to pluripotency progression.

(a, b) Representative images of rosettes and aggregates generated by ESCs 48 hrs after seeding in BME in the indicated conditions and stained for the indicated markers. a, KLF4 (green) and OCT6 (red). b, OTX2 (green) and KLF4 (red). 3 independent experiments with similar results. c, Rosettes generated by ESCs 48 hrs after seeding in BME and stained for PODXL live or after fixation and permeabilization. Permeabilized samples showed PODXL staining, incorrectly suggesting presence of a lumen, while live staining revealed that the PODXL was not exposed at the cell surface and no lumen was actually present (illustrative examples, repeated > 5 times). d, Examples to illustrate criteria for calling ESC aggregates displaying a lumen, using live PODXL staining combined with phalloidin to reveal polarization (illustrative examples, repeated > 5 times). e, Individual colour channels for Fig. 2n (representative of 3 experiments). f, Individual colour channels for Fig. 2o (representative of 4 experiments). Scale bars 20 µm (a–f).

Extended Data Fig. 3 WNT controls the initial phase of the naive-primed transition.

(a, b) Immunostaining for the indicated markers of ESCs plated in EpiLC conditions (a) or N2B27 medium (b) for the indicated number of days (single experiments). c, Dispersed colonies (dashed white circles) obtained after passaging of a blastocyst grown out in the presence of IWP2, MEK inhibitor and LIF (LIM). 15 from 18 and 12 from 13 embryos yielded cell lines. d, Chimeras (brown), obtained after blastocyst injection of newly derived LIM cell lines (LIMA2, LIMB1 and LIMB3), with mate (black) and pups (only for LIMA2). Brown pups indicate that germline transmission occurred. e, RT-PCR analysis of ESCs maintained for 6 passages in L2i or MEK/WNT-inhibited conditions and of EpiSCs. n = 3 (LIF/2i and LIM) or n = 2 (EpiSC) biological replicates. Scale bars 20 µm (a,b), 100 µm (c).

Source data

Extended Data Fig. 4 MEK/WNT-inhibition maintains rosette characteristics.

a, Immunostainings and western blot for the indicated markers of R1 ESCs maintained in L2i or LIM for 6 passages or as indicated (3 independent repeats with similar results; western blot 1 experiment, full blot in Source Data). b, Plating efficiencies of LIM cells in the presence and absence of LIF (n = 3 biological replicates, R1, FN3 and IB10 cells). c, Female SV8 L2i ESCs, LIM cells and EpiSCs stained for Xist by RNA-FISH. Puncta indicating Xist expression (arrowheads) are visible in both L2i and LIM cells, while only EpiSCs display Xist clouds indicating X-chromosome inactivation (arrows) (two independent experiments). d, Doubling times of the indicated cell lines in the indicated conditions. p12 and p24 indicate passage numbers of the cell lines. e, t-SNE plot of single cell RNA-Seq data, and log2 expression levels of the indicated genes plotted into the t-SNE plot. n = 159, 178, 102 (R1) and 184, 149 and 164 (IB10) cells for L2i, LIM and primed, respectively. f, RSCs were cultured for 10 days in the indicated conditions and immunostained for OTX2 (red). Representative images from 3 independent replicates. g, To determine apicobasal polarity, the Golgi apparatus was marked by Gm130 antibody staining, imaged by confocal microscopy and colorized according to z-stack level. Basal side of the cells correspond to low z-levels (green), and apical side to high levels (red). The data show apical Golgi localization in LIM cells, indicating their apicobasal polarity. Three experiments with similar results. h, Live tracking of individual cell locations during 24 hrs of L2i ESCs, LIM cells on serum-coating, and LIM cells on BME-coating. 80, 47 and 20 cells, respectively. Representative of 4 experiments. Scale bars 20 µm (a,g), 10 µm (c,f), 100 µm (h).

Source data

Extended Data Fig. 5 Otx2 enables MEK-driven transition to primed pluripotency.

a, RGd2 ESCs and RSCs were plated in primed conditions and analysed at several time points for GFP by flow cytometry. n = 3 independent replicates (3rd is shown in main Fig. 5a). b, ESCs and RSCs, carrying the Tbx3-RFP;Oct6-GFP reporters, were plated in primed conditions and analysed daily for expression of the reporters. Lines go through means, n = 3 independent experiments. c, Western blots showing OCT6 (only for IB10), ERK and phospho-ERK in IB10 and R1 ESCs and RSCs upon transfer to primed medium. Two (for OCT6) and three biological replicates. Full blots in Source Data. d, Immunostaining for OTX2 in TRE-Otx2 ESCs following induction by doxycycline for the indicated duration (single experiment). Scale bar 40 µm.

Source data

Extended Data Fig. 6 A primed chromatin landscape in rosette-like stem cells.

a, Immunofluorescence and western blot analysis for DNMT3L expression in the indicated cell types (3 independent repeats with similar results; western blot 1 experiment). b, Individual colour channels for Fig. 6b, right panel. Representative of 5 experiments. c, Representative input plot for H3K27me3 ChIP-Seq for the bivalent cluster. (d,e) H3K4me3 (d) or H3K27me3 (e) ChIP-Seq correlogram of ESCs, RSCs and EpiLCs. Two biological repeats for each condition. (f, g) Average intensity plots of H3K4me3 and H3K27me3 in the H3K4me3 (f) or H3K27me3 (g) enriched cluster. Two biological repeats for each condition. h, Screenshots from the UCSC genome browser showing H3K4me3 (blue) and H3K27me3 (red) peaks on the Pou3f1 and Fgf5 genes. Scale bars 20 µm (a,b).

Source data

Extended Data Fig. 7 Rosette-like stem cells remodel pericentric heterochromatin.

(a–d) Metaphase chromosome spreads of ESCs, RSCs and primed cells stained for the indicated markers. DAPI (blue), centromeres (red). e, Female RSCs (LIMA6) were cultured for 3 days in primed conditions and immunostained for the indicated markers. Arrowheads indicate H3K27me3-positive inactive X chromosomes demonstrating transition to primed pluripotency. f, RSCs were cultured for 10 days in the indicated conditions and immunostained for the indicated markers. Five (a–d) or three (e, f) biological replicates. Scale bars 20 µm (a-d), 10 µm (e, f).

Extended Data Fig. 8 Rosette-specific formatting of pericentric heterochromatin.

a, Counts of embryos of the indicated stages displaying pericentric H3K27me3 accumulation, and in which the presence of a rosette, KLF4 or OTX2 expression is detected. Embryos are the same as those of Extended Data Fig. 1a, b. b, Confocal microscopy images of E5.1 embryo stained for H3K27me3 (green), centromeres (magenta), OTX2 (red), and DAPI (blue). Some H3K27me3-positive pHC foci are indicated by white arrows (light blue, overlap of H3K27me3 and DAPI). Representative example from the embryos analysed in Extended Data Fig. 8a. Scale bars 20 µm.

Source data

Supplementary information

Reporting Summary

Supplementary Tables 1–6

Supplementary Table 1. Related to Fig. 3c,d. Excel file containing RNA-seq data of R1 ESC time course in LIM and primed conditions. Supplementary Table 2. Related to Fig. 3e–g. Excel file containing RNA-seq data of R1 and CGR8 ESCs in L2i, LIM, 35 h in primed, and 4 days in primed conditions. Differential gene expression fold change and significance were estimated from biological duplicates using DESeq2 Bioconductor package v.1.18.1. The DESeq2 package uses a Wald test to establish P values, which undergo Benjamini and Hochberg adjustment for multiple testing. Supplementary Table 3. Related to Fig. 4. Contribution to chimeras of male LIM-derived cell lines. Supplementary Table 4. Related to Fig. 4f. Excel file containing GO analysis of genes differentially expressed between L2i ESCs and LIM cells (two biological replicates). The P values were calculated from minimum hypergeometric scores from a ranked gene list using GOrilla. Supplementary Table 5. Related to Fig. 6c–h. Excel file of genes with bivalent promoters in L2i ESCs, RSCs and Epiblast-like cells. Supplementary Table 6. Oligonucleotide primers, antibodies and cell lines used in the study.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Neagu, A., van Genderen, E., Escudero, I. et al. In vitro capture and characterization of embryonic rosette-stage pluripotency between naive and primed states. Nat Cell Biol 22, 534–545 (2020). https://doi.org/10.1038/s41556-020-0508-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0508-x

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