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:

Evaluating totipotency using criteria of increasing stringency

Abstract

Totipotency is the ability of a single cell to give rise to all of the differentiated cell types that build the conceptus, yet how to capture this property in vitro remains incompletely understood. Defining totipotency relies on a variety of assays of variable stringency. Here, we describe criteria to define totipotency. We explain how distinct criteria of increasing stringency can be used to judge totipotency by evaluating candidate totipotent cell types in mice, including early blastomeres and expanded or extended pluripotent stem cells. Our data challenge the notion that expanded or extended pluripotent states harbour increased totipotent potential relative to conventional embryonic stem cells under in vitro and in vivo conditions.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Gene expression analysis of candidate totipotent stem cells using RNA-seq.
Fig. 2: In vitro capacity and potential of candidate totipotent stem cells to give rise to trophoblast stem cells.
Fig. 3: Single-cell transcriptional comparison between blastoid cells, embryo cells and different stem cell states.
Fig. 4: In vivo potential of candidate totipotent stem cells to give rise to the TE lineage at E4.5.
Fig. 5: In vivo potential of candidate totipotent stem cells to give rise to the trophoblast lineage at E6.25.
Fig. 6: In vivo potential of candidate totipotent stem cells to give rise to the trophoblast lineage of the E12.5 placenta.

Similar content being viewed by others

Data availability

All raw and processed sequencing data, as well as .loom files for visualization in SCope, generated in this study, have been submitted to the NCBI GEO (http://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE145609 and also with further instructions at GitHub (https://github.com/pasquelab/totipotency). Furthermore, published datasets were downloaded as provided by Deng et al.41 (GEO: GSE45719), Posfai et al.4 (GEO: GSE84892), Mohammed et al.42 (GEO: GSE100597), Chen et al.43 (GEO: GSE74155), Li et al.39 (GEO: GSE135289, GSE135701), Yang et al.35 (ENA: ERP005641), Yang et al.36 (GEO: GSE89303) Pijuan-Sala et al.49 (https://github.com/MarioniLab/EmbryoTimecourse2018), Janiszewski et al.77 (GEO: GSE126229), Sozen et al.40 (GEO: GSE134240). All other data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

Codes pertaining to important analyses in this study are available from the Pasque laboratory GitHub webpage (https://github.com/pasquelab/totipotency).

References

  1. Solter, D. From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research. Nat. Rev. Genet. 7, 319–327 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Rossant, J. Investigation of the determinative state of the mouse inner cell mass. II. The fate of isolated inner cell masses transferred to the oviduct. J. Embryol. Exp. Morphol. 33, 991–1001 (1975).

    CAS  PubMed  Google Scholar 

  3. Tarkowski, A. K., Suwinska, A., Czolowska, R. & Ozdzenski, W. Individual blastomeres of 16- and 32-cell mouse embryos are able to develop into foetuses and mice. Dev. Biol. 348, 190–198 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Posfai, E. et al. Position- and hippo signaling-dependent plasticity during lineage segregation in the early mouse embryo. eLife 6, https://doi.org/10.7554/eLife.22906 (2017).

  5. Cockburn, K. & Rossant, J. Making the blastocyst: lessons from the mouse. J. Clin. Invest. 120, 995–1003 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kwon, G. S., Viotti, M. & Hadjantonakis, A. K. The endoderm of the mouse embryo arises by dynamic widespread intercalation of embryonic and extraembryonic lineages. Dev. Cell 15, 509–520 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nowotschin, S. et al. The emergent landscape of the mouse gut endoderm at single-cell resolution. Nature 569, 361–367 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. 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 

  10. Tanaka, S., Kunath, T., Hadjantonakis, A. K., Nagy, A. & Rossant, J. Promotion of trophoblast stem cell proliferation by FGF4. Science 282, 2072–2075 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Kunath, T. et al. Imprinted X-inactivation in extra-embryonic endoderm cell lines from mouse blastocysts. Development 132, 1649–1661 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. Niakan, K. K., Schrode, N., Cho, L. T. & Hadjantonakis, A. K. Derivation of extraembryonic endoderm stem (XEN) cells from mouse embryos and embryonic stem cells. Nat. Protoc. 8, 1028–1041 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cho, L. T. et al. Conversion from mouse embryonic to extra-embryonic endoderm stem cells reveals distinct differentiation capacities of pluripotent stem cell states. Development 139, 2866–2877 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Papaioannou, V. E., McBurney, M. W., Gardner, R. L. & Evans, M. J. Fate of teratocarcinoma cells injected into early mouse embryos. Nature 258, 70–73 (1975).

    Article  CAS  PubMed  Google Scholar 

  16. Gardner, R. L. & Rossant, J. Investigation of the fate of 4-5 day post-coitum mouse inner cell mass cells by blastocyst injection. J. Embryol. Exp. Morphol. 52, 141–152 (1979).

    CAS  PubMed  Google Scholar 

  17. Niakan, K. K. et al. Sox17 promotes differentiation in mouse embryonic stem cells by directly regulating extraembryonic gene expression and indirectly antagonizing self-renewal. Genes Dev. 24, 312–326 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Canham, M. A., Sharov, A. A., Ko, M. S. & Brickman, J. M. Functional heterogeneity of embryonic stem cells revealed through translational amplification of an early endodermal transcript. PLoS Biol. 8, e1000379 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Beddington, R. S. & Robertson, E. J. An assessment of the developmental potential of embryonic stem cells in the midgestation mouse embryo. Development 105, 733–737 (1989).

    Article  CAS  PubMed  Google Scholar 

  20. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rodriguez-Terrones, D. et al. A molecular roadmap for the emergence of early-embryonic-like cells in culture. Nat. Genet. 50, 106–119 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Niwa, H. et al. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation. Cell 123, 917–929 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Ng, J. H., Heng, J. C., Loh, Y. H. & Ng, H. H. Transcriptional and epigenetic regulations of embryonic stem cells. Mutat. Res. 647, 52–58 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Kuckenberg, P. et al. The transcription factor TCFAP2C/AP-2γ cooperates with CDX2 to maintain trophectoderm formation. Mol. Cell Biol. 30, 3310–3320 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cambuli, F. et al. Epigenetic memory of the first cell fate decision prevents complete ES cell reprogramming into trophoblast. Nat. Commun. 5, 5538 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Yamanaka, S. & Takahashi, K. Induction of pluripotent stem cells from mouse fibroblast cultures. Tanpakushitsu Kakusan Koso 51, 2346–2351 (2006).

    CAS  PubMed  Google Scholar 

  27. Benchetrit, H. et al. Direct induction of the three pre-implantation blastocyst cell types from fibroblasts. Cell Stem Cell 24, 983–994 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

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

    Article  CAS  PubMed  Google Scholar 

  30. Hall, J. et al. Oct4 and LIF/Stat3 additively induce Kruppel factors to sustain embryonic stem cell self-renewal. Cell Stem Cell 5, 597–609 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. 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 

  32. Morgani, S. M. et al. Totipotent embryonic stem cells arise in ground-state culture conditions. Cell Rep. 3, 1945–1957 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Abad, M. et al. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 502, 340–345 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Choi, Y. J. et al. Deficiency of microRNA miR-34a expands cell fate potential in pluripotent stem cells. Science 355, https://doi.org/10.1126/science.aag1927 (2017).

  35. Yang, J. et al. Establishment of mouse expanded potential stem cells. Nature 550, 393–397 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yang, Y. et al. Derivation of pluripotent stem cells with in vivo embryonic and extraembryonic potency. Cell 169, 243–257 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, L. et al. G1 cyclins link proliferation, pluripotency and differentiation of embryonic stem cells. Nat. Cell Biol. 19, 177–188 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gao, X. et al. Establishment of porcine and human expanded potential stem cells. Nat. Cell Biol. 21, 687–699 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, R. et al. Generation of blastocyst-like structures from mouse embryonic and adult. Cell Cult. Cell 179, 687–702 (2019).

    CAS  Google Scholar 

  40. Sozen, B. et al. Self-organization of mouse stem cells into an extended potential blastoid. Dev. Cell 51, 698–712 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Deng, Q., Ramskold, D., Reinius, B. & Sandberg, R. Single-cell RNA-seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science 343, 193–196 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Mohammed, H. et al. Single-cell landscape of transcriptional heterogeneity and cell fate decisions during mouse early gastrulation. Cell Rep. 20, 1215–1228 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen, G. et al. Single-cell analyses of X chromosome inactivation dynamics and pluripotency during differentiation. Genome Res. 26, 1342–1354 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).

    Article  PubMed  Google Scholar 

  46. Rugg-Gunn, P. J., Cox, B. J., Ralston, A. & Rossant, J. Distinct histone modifications in stem cell lines and tissue lineages from the early mouse embryo. Proc. Natl Acad. Sci. USA 107, 10783–10790 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Murray, A., Sienerth, A. R. & Hemberger, M. Plet1 is an epigenetically regulated cell surface protein that provides essential cues to direct trophoblast stem cell differentiation. Sci. Rep. 6, 25112 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rivron, N. C. et al. Blastocyst-like structures generated solely from stem cells. Nature 557, 106–111 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Kime, C. et al. Induced 2C expression and implantation-competent blastocyst-like cysts from primed pluripotent stem cells. Stem Cell Rep. 13, 485–498 (2019).

    Article  CAS  Google Scholar 

  50. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Davie, K. et al. A single-cell transcriptome atlas of the aging Drosophila brain. Cell 174, 982–998 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Suwinska, A., Czolowska, R., Ozdzenski, W. & Tarkowski, A. K. Blastomeres of the mouse embryo lose totipotency after the fifth cleavage division: expression of Cdx2 and Oct4 and developmental potential of inner and outer blastomeres of 16- and 32-cell embryos. Dev. Biol. 322, 133–144 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Strnad, P. et al. Inverted light-sheet microscope for imaging mouse pre-implantation development. Nat. Methods 13, 139–142 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Strumpf, D. et al. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development 132, 2093–2102 (2005).

    Article  CAS  PubMed  Google Scholar 

  55. Home, P. et al. GATA3 is selectively expressed in the trophectoderm of peri-implantation embryo and directly regulates Cdx2 gene expression. J. Biol. Chem. 284, 28729–28737 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ralston, A. et al. Gata3 regulates trophoblast development downstream of Tead4 and in parallel to Cdx2. Development 137, 395–403 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Brulet, P., Babinet, C., Kemler, R. & Jacob, F. Monoclonal antibodies against trophectoderm-specific markers during mouse blastocyst formation. Proc. Natl Acad. Sci. USA 77, 4113–4117 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Avilion, A. A. et al. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 17, 126–140 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Watson, E. D. & Cross, J. C. Development of structures and transport functions in the mouse placenta. Physiology 20, 180–193 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Dumollard, R., Carroll, J., Duchen, M. R., Campbell, K. & Swann, K. Mitochondrial function and redox state in mammalian embryos. Semin. Cell Dev. Biol. 20, 346–353 (2009).

    Article  CAS  PubMed  Google Scholar 

  61. Whittington, N. C. & Wray, S. Suppression of red blood cell autofluorescence for immunocytochemistry on fixed embryonic mouse tissue. Curr. Protoc. Neurosci. 81, 2.28.1–2.28.12 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Sferruzzi-Perri, A. N., Higgins, J. S., Vaughan, O. R., Murray, A. J. & Fowden, A. L. Placental mitochondria adapt developmentally and in response to hypoxia to support fetal growth. Proc. Natl Acad. Sci. USA 116, 1621–1626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Albers, R. E. et al. Gestational differences in murine placenta: glycolytic metabolism and pregnancy parameters. Theriogenology 107, 115–126 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Tanaka, M., Hadjantonakis, A. K. & Nagy, A. Aggregation chimeras. Combining ES cells, diploid and tetraploid embryos. Methods Mol. Biol. 158, 135–154 (2001).

    CAS  PubMed  Google Scholar 

  65. Nagy, A., Gertsenstein, M., Vintersten, K. & Behringer, R. Assembling aggregates between embryonic stem (ES) cells and tetraploid embryos. CSH Protoc. 2006, https://doi.org/10.1101/pdb.prot4426 (2006).

  66. Tomoda, K. et al. Reprogramming epiblast stem cells into pre-implantation blastocyst cell-like cells. Preprint at bioRxiv https://doi.org/10.1101/2020.09.29.318279 (2020).

  67. Tremble, K. et al. Sox2 modulation increases naïve pluripotency plasticity. Preprint at bioRxiv https://doi.org/10.1101/2020.01.14.906933 (2020).

  68. Yachie-Kinoshita, A. et al. Modeling signaling-dependent pluripotency with Boolean logic to predict cell fate transitions. Mol. Syst. Biol. 14, e7952 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  70. Blij, S., Parenti, A., Tabatabai-Yazdi, N. & Ralston, A. Cdx2 efficiently induces trophoblast stem-like cells in naive, but not primed, pluripotent stem cells. Stem Cells Dev. 24, 1352–1365 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Hackett, J. A. et al. Tracing the transitions from pluripotency to germ cell fate with CRISPR screening. Nat. Commun. 9, 4292 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Hadjantonakis, A. K. & Papaioannou, V. E. Dynamic in vivo imaging and cell tracking using a histone fluorescent protein fusion in mice. BMC Biotechnol. 4, 33 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Nagy, A., Rossant, J., Nagy, R., Abramow-Newerly, W. & Roder, J. C. Derivation of completely cell culture-derived mice from early-passage embryonic stem cells. Proc. Natl Acad. Sci. USA 90, 8424–8428 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Rugg-Gunn, P. J. et al. Cell-surface proteomics identifies lineage-specific markers of embryo-derived stem cells. Dev. Cell 22, 887–901 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  76. Liao, Y., Smyth, G. K. & Shi, W. The R package rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads. Nucleic Acids Res. 47, e47 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Janiszewski, A. et al. Dynamic reversal of random X-chromosome inactivation during iPSC reprogramming. Genome Res. 29, 1659–1672 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. 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  Google Scholar 

  79. Petropoulos, S. et al. Single-cell RNA-seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 167, 285 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Picelli, S. et al. Tn5 transposase and tagmentation procedures for massively scaled sequencing projects. Genome Res. 24, 2033–2040 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Ramskold, D., Wang, E. T., Burge, C. B. & Sandberg, R. An abundance of ubiquitously expressed genes revealed by tissue transcriptome sequence data. PLoS Comput. Biol. 5, e1000598 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Huynh-Thu, V. A., Irrthum, A., Wehenkel, L. & Geurts, P. Inferring regulatory networks from expression data using tree-based methods. PLoS ONE https://doi.org/10.1371/journal.pone.0012776 (2010).

  84. Moerman, T. et al. GRNBoost2 and Arboreto: efficient and scalable inference of gene regulatory networks. Bioinformatics 35, 2159–2161 (2019).

    Article  CAS  PubMed  Google Scholar 

  85. Vintersten, K. et al. Mouse in red: red fluorescent protein expression in mouse ES cells, embryos, and adult animals. Genesis 40, 241–246 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Gertsenstein, M. Mouse embryos’ fusion for the tetraploid complementation assay. Methods Mol. Biol. 1313, 41–59 (2015).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the contribution of the Model Production Core staff at the Center for Phenogenomics for technical support. Single-cell sequencing was performed at National Genomics Infrastructure in Stockholm at the Science for Life Laboratory (funded by the Knut and Alice Wallenberg Foundation and the Swedish Research Council) with assistance from SNIC/Uppsala Multidisciplinary Center for Advanced Computational Science with massively parallel sequencing and access to the UPPMAX computational infrastructure. Bulk RNA-seq was performed at the KU Leuven Genomics Core. We thank S. Aerts, S. Aibar, C. Davie and C. Flerin for discussions and support; and J. Wu for the C1-12 tdTomato-expressing D-EPSCs. This work was funded by CIHR (FDN-143334), Genome Canada and Ontario Genomics (OGI-099), Programme de bourses de chercheur-boursier FRQS Junior 1 (FRQS 268829, 280187), the Swedish Research Council, Ragnar Söderberg Foundation, Ming Wai Lau Center for Reparative Medicine, Center for Innovative Medicine, Wallenberg Academy Fellow, NSERC (2014-04497). Research in the Pasque laboratory is supported by The Research Foundation–Flanders (FWO; Odysseus Return Grant G0F7716N to V.P.; FWO grants G0C9320N and G0B4420N to V.P.), the KU Leuven Research Fund (BOFZAP starting grant StG/15/021BF to V.P. C1 grant C14/16/077 to V.P. and project financing) and FWO PhD fellowships to A.Janiszewski (1158318N), I.T. (1S72719N) and S.K.T. (1S75720N).

Author information

Authors and Affiliations

Authors

Contributions

J.P.S., A.M., A.Janiszewski, T.Y., T.P., M.E.B., N.D.G. and S.K.T. performed in vitro stem cell experiments. E.P., I.R. and B.B. performed in vivo chimera experiments. J.P.S., A.Janiszewski, P.K., S.P., M.E.B., I.T., F.L. and V.P. performed transcriptional analyses. E.P., V.P., F.L., J.P.S., A.Jurisicova and J.R. planned experiments, analysed data and wrote the manuscript. V.P., F.L., E.P. and J.R. conceived the study.

Corresponding authors

Correspondence to Eszter Posfai, Vincent Pasque, Fredrik Lanner or Janet Rossant.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Peer reviewer reports are available. Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Culture conditions and morphologies of ESCs, L-EPSCs and D-EPSCs.

Scale bar 100 µ. At least 5 brightfield images were taken of cells in each culture condition.

Extended Data Fig. 2 Transcriptional profiling of L- and D-EPSCs related to Fig. 1.

a, Number of DEGs between 2iLif ESCs and L-EPSCs at different days of conversion. b, Gene expression comparison of pluripotency and somatic genes between 2iLif ESCs and L-EPSCs and mouse embryonic fibroblasts (MEFs) as control. 1 sample. c, Changes in expression levels between 2iLif ESCs and L-EPSCs for genes reported to change in Yang et al.35. 1 sample. d, GSEA analysis of 4-cell stage signature genes in bulk RNA-seq data of L-EPSCs generated in this study (2iLif n = 1, EPSC n = 5) and published bulk L-EPSCs RNA-seq data (Yang et al. 35, (2iLif n = 5, EPSC n = 5), Yang et al.36 (2iLif n = 2, EPSC n = 2)). e, UMAP from Fig. 1e showing the resource from which each cell was merged, and lists the number of cells used from each. f, FeaturePlots projecting expression of representative 2-cell, EPI, PE, and TE marker genes, overlaying Fig. 1e UMAP. g, GSEA analysis of 4-cell stage signature genes in scRNA-seq data of L-EPSCs and D-EPSCs generated in this study (2iLif n = 191, L-EPSCs n = 182, D-EPSCs n = 186) and published L-EPSCs scRNA-seq data (Yang et al.35 (2iLif n = 96, EPSC=96)). *: p-value 0,05-0,01, **: p-value 0,01-0,001. h, Heatmap of genes previously reported to be upregulated in D-EPSCs compared to ESCs cultured in Lif/serum conditions, which are similarly upregulated in D-EPSCs (generated in this study) when compared to naïve 2iLif ESCs. i, Violin plot using Module A & B from Yang Y. et al., 2017, Figure 7C36 Fig. 7c, showing significant upregulation in both L-EPSCs and D-EPSCs compared to 2i. N = number of cells in each condition.

Source data

Extended Data Fig. 3 Scattering of cells during single-cell integration and gene regulatory networks analysis, related to Fig. 1.

a, Cells are binned according to their position in the integration UMAP in Fig. 1e depending on whether they occupy a position defined as ICM, TE, PE, EPI E4.5, EPI E5.5 or EPI E6.75. ScRNAseq profiles of L-EPSCs generated in this study are merged with published EPSCs signatures from the Liu lab (Liu EPSCs)35. Modular scores are then presented for each subcluster of each stem cell type. The modular scores are based on the top100 differentially expressed gene signatures defined by the corresponding embryo data. The number of cells in each group can be found in Source data Extended Fig. 3. b, UMAP constructed based on the activity of gene regulatory networks in 2iLif ESCs, embryonic stages E3.5 ICM, E4.5 EPI, E5.5 EPI, L-EPSCs and D-EPSCs.

Source data

Extended Data Fig. 4 Flow cytometry Gating strategy.

a, Displayed is an example of the gating strategy implemented to generate flow cytometry data. The data displayed uses the iCdx2 Elf5.2A.mCherry ESCs, which are eGFP+, contain the Elf5.2A.mCherry reporter, and are stained for both CD40 and Plet1.The plots are pseudo-colour plots and the x-axis labels state markers and fluorochromes used. Laser (V=violet; B=blue; G=green; R=red) and filter bandpass information is also included on x-axis in brackets. Ancestry plots are included to show how cells were gated: majority of cells were included in FSC/SSC gate due to size discrepancy between cells of interest and underlying feeder cells. Single cells were determined by FSC-W x FSC-H and SSC-W x SSC-H plots. Live cells were gated using dead cell stain Sytox blue. Cells of interest were discriminated from underlying feeder cells (EGFP negative) by expression of eGFP. Fluorescence minus one (FMO) controls were used to establish gates in order to separate positive and negative populations.

Extended Data Fig. 5 Re-analysis of transcriptional profiling data of blastoid cells related to Fig. 3.

a, UMAP from Fig. 3a identifying original datasets4,35,39,40,42,43,50 for each cell. b, UMAP from Fig. 3a, highlighting ZG-blastoid EPI, ZG-blastoid ZG, B-blastoid TE, and ZG-blastoid intermediate. c, UMAP from Fig. 3a, highlighting ZG-blastoid populations produced with 2i/Lif vs. EPSCs. d, UMAP from Fig. 3a, highlighting B-blastoid EPI, B-blastoid PE, B-blastoid TE, and B-blastoid intermediates. e, Pie chart showing percent of each cell type category based on our re-analysis of all B-blastoid cells.

Extended Data Fig. 6 Gene regulatory network atlas spanning mouse embryo stages from morula to gastrulation and in vitro blastoid models.

a, UMAP analysis based on the activity of gene regulatory networks. b, Heatmap representing the activity of selected regulons associated with lineage-specific TFs averaged across cells from each cluster. c, Dot plot showing expression levels of NANOG, GATA4, GATA3, T and CDX2 target genes derived from gene regulatory network analysis of selected cell types.

Extended Data Fig. 7 E4.5 and E6.25 chimeras using different L-EPSC or D-EPSC cell lines.

ad, Stacked bar charts showing percent of E4.5 and E6.25 chimeric embryos with different lineage contributions (epiblast – EPI, trophoblast (ExE and ectoplacental cone) –TB, epiblast and trophoblast position – EPI + TB position). mScarlet-NLS expressing cells were cultured in either 2i/Lif (11 chimeras analyzed at E4.5 and 8 at E6.25) or L-EPSC (15 chimeras analyzed at E4.5 and 24 at E6.25) conditions and tdTomato expressing cells were cultured in 2i/Lif (9 chimeras analyzed at E6.25) or D-EPSC (26 chimeras analyzed at E4.5 and 6 chimeras analyzed at E6.25) conditions. The number of chimeras showing different lineage contributions is indicated withing the bar chart. e-h, Representative immunofluorescent stainings of E4.5 or E6.25 chimeric embryos generated using L-EPSCs (mScarlet-NLS) or D-EPSCs (tdTomato), stained for mScarlet (detected using an mCherry antibody), tdTomato (using a DsRed antibody), SOX2 (EPI), CDX2 (TE), or ELF5 (ExE). Whole embryo images show maximum intensity projections in panels e, g and h. Single planes shown in panel f and in all magnified views. Scale bar = 20 µ. Number of chimeras analyzed: 15 in panel E, 24 in panel F, 26 in panel G, and 6 in panel H.

Extended Data Fig. 8 Analysis of chimeric E12.5 placentas generated with totipotent blastomeres.

Immunofluorescent stainings of E12.5 chimeric placentas generated with a single blastomere of an 8-cell stage embryo that expressed either H2B-GFP or DsRed and a wild-type host embryo. H2B-GFP expressing placentas were stained for TFAP2C, TPBPA, MCT4, CD31 and GFP (top panel). DsRed expressing placentas were stained for KRT8, MCT1, CDH3 and mCherry/RFP (bottom panel). Images shown at different magnifications. Scale bars: 500 µ (4x), 200 µ (7x), 50 µ (40x). Sections from 4 chimeric placentas were analyzed.

Extended Data Fig. 9 Full panel of chimeric E12.5 placentas generated using tetraploid complementation.

Immunofluorescent staining of E12.5 chimeric placentas generated with H2B-GFP expressing tetraploid host embryo and wild-type ESCs (left panel), wild-type tetraploid host embryo and H2B-GFP expressing ESCs (middle panel) and wild-type host embryo (diploid) and H2B-GFP expressing L-EPSCs. Placenta sections are immuno-stained for GFP and the following trophoblast markers: CDH3, TPBPA, and MCT4. A total of 14 H2B-GFP positive L-EPSCs placentas were collected from two females, and 3 to 4 placentas were analyzed for each marker. Images shown at different magnifications. Scale bars: 500 µ (4x), 200 µ (7x), 50 µ (40x). Sections from 4 chimeric placentas were analyzed.

Supplementary information

Supplementary Information

Supplementary Fig. 1: SCENIC analysis in mouse embryo stages from morula to gastrulation and in vitro blastoid models.

Reporting Summary

Peer Review Information

Supplementary Tables 1–7

Supplementary Table 1: differential gene expression analysis of bulk RNA-seq data. Supplementary Table 2: Gene Ontology analysis. Supplementary Table 3: Panther pathway enrichment analysis on differentially expressed genes between ESCs and EPSCs. Supplementary Table 4: four-cell stage signature associated with Fig.1d. Supplementary Table 5: GSEA signatures for each stage. Supplementary Table 6: List of all of the primers used. Supplementary Table 7: list of all of the antibodies used.

Source data

Source Data Fig. 1

Source data Fig. 1f.

Source Data Fig. 2

Statistical source data Fig. 2b and Fig. 2d,e.

Source Data Fig. 2

Flow cytometry gating strategy related to Fig. 2.

Source Data Fig. 3

Source data Fig. 3c.

Source Data Fig. 4

Source data Fig. 4b,d.

Source Data Fig. 5

Source data Fig. 5c.

Source Data Fig. 6

Source data Fig. 6b,c.

Source Data Extended Data Fig. 2

Source data for the number of differentially expressed genes Extended Data Fig. 2a.

Source Data Extended Data Fig. 3

Source data for the number of cells per group Extended Data Fig. 3a.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Posfai, E., Schell, J.P., Janiszewski, A. et al. Evaluating totipotency using criteria of increasing stringency. Nat Cell Biol 23, 49–60 (2021). https://doi.org/10.1038/s41556-020-00609-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-00609-2

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