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:

Identification of the central intermediate in the extra-embryonic to embryonic endoderm transition through single-cell transcriptomics

Abstract

High-resolution maps of embryonic development suggest that acquisition of cell identity is not limited to canonical germ layers but proceeds via alternative routes. Despite evidence that visceral organs are formed via embryonic and extra-embryonic trajectories, the production of organ-specific cell types in vitro focuses on the embryonic one. Here we resolve these differentiation routes using massively parallel single-cell RNA sequencing to generate datasets from FOXA2Venus reporter mouse embryos and embryonic stem cell differentiation towards endoderm. To relate cell types in these datasets, we develop a single-parameter computational approach and identify an intermediate en route from extra-embryonic identity to embryonic endoderm, which we localize spatially in embryos at embryonic day 7.5. While there is little evidence for this cell type in embryonic stem cell differentiation, by following the extra-embryonic trajectory starting with naïve extra-embryonic endoderm stem cells we can generate embryonic gut spheroids. Exploiting developmental plasticity therefore offers alternatives to pluripotent cells and opens alternative avenues for in vitro differentiation.

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: Single-cell sequencing and analysis of embryonic Foxa2POS lineages.
Fig. 2: CAT and identification of the InterVE cluster as an intermediate population between VE and DE.
Fig. 3: Functional properties of VE in the transition to DE.
Fig. 4: ST reveals InterVE in E7.5 embryos.
Fig. 5: In vitro differentiation of endoderm lineages.
Fig. 6: An in vitro model for generating gut spheroids (nEnd spheroids) from extra-embryonic endoderm.

Similar content being viewed by others

Data availability

The single-cell RNA-seq data used in this study is in the Gene Expression Omnibus under accession number GSE164464. Previously published Nowotschin-2019 data that were re-analysed here are available under accession code GSE123046. Source data are provided with this study. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

All analyses, package versions as well as development environments for reproducibility purposes are publicly available at https://github.com/brickmanlab/rothova-et-al-2022. Implementation of CAT and the related pre-processed datasets are publicly available at https://github.com/brickmanlab/CAT. The interactive CAT app is available at https://align-clusters.com.

References

  1. Zorn, A. M. & Wells, J. M. Vertebrate endoderm development and organ formation. Annu. Rev. Cell Dev. Biol. 25, 221–251 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kraus, M. R. C. & Grapin-Botton, A. Patterning and shaping the endoderm in vivo and in culture. Curr. Opin. Genet. Dev. 22, 347–353 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Lawson, K. A., Meneses, J. J. & Pedersen, R. A. Cell fate and cell lineage in the endoderm of the presomite mouse embryo, studied with an intracellular tracer. Dev. Biol. 115, 325–339 (1986).

    Article  CAS  PubMed  Google Scholar 

  4. 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 

  5. Viotti, M., Nowotschin, S. & Hadjantonakis, A.-K. Afp::mCherry, a red fluorescent transgenic reporter of the mouse visceral endoderm. Genesis 49, 124–133 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Viotti, M., Nowotschin, S. & Hadjantonakis, A.-K. SOX17 links gut endoderm morphogenesis and germ layer segregation. Nat. Cell Biol. 16, 1146–1156 (2014).

    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. 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 

  9. Anderson, K. G. V. et al. Insulin fine-tunes self-renewal pathways governing naive pluripotency and extra-embryonic endoderm. Nat. Cell Biol. 19, 1164–1177 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Gardner, R. L. Investigation of cell lineage and differentiation in the extraembryonic endoderm of the mouse embryo. Development 68, 175–198 (1982).

    Article  CAS  Google Scholar 

  11. Verheijen, M. H. & Defize, L. H. Signals governing extraembryonic endoderm formation in the mouse: involvement of the type 1 parathyroid hormone-related peptide (PTHrP) receptor, p21Ras and cell adhesion molecules. Int. J. Dev. Biol. 43, 711–721 (1999).

    CAS  PubMed  Google Scholar 

  12. Lu, C. C., Brennan, J. & Robertson, E. J. From fertilization to gastrulation: axis formation in the mouse embryo. Curr. Opin. Genet. Dev. 11, 384–392 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Beddington, R. S. & Robertson, E. J. Axis development and early asymmetry in mammals. Cell 96, 195–209 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Belo, J. A. et al. Cerberus-like is a secreted factor with neuralizing activity expressed in the anterior primitive endoderm of the mouse gastrula. Mech. Dev. 68, 45–57 (1997).

    Article  CAS  PubMed  Google Scholar 

  15. Costello, I. et al. Lhx1 functions together with Otx2, Foxa2, and Ldb1 to govern anterior mesendoderm, node, and midline development. Genes Dev. 29, 2108–2122 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Robb, L. & Tam, P. P. L. Gastrula organiser and embryonic patterning in the mouse. Semin. Cell Dev. Biol. 15, 543–554 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Burtscher, I. & Lickert, H. Foxa2 regulates polarity and epithelialization in the endoderm germ layer of the mouse embryo. Development 136, 1029–1038 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Scheibner, K. et al. Epithelial cell plasticity drives endoderm formation during gastrulation. Nat. Cell Biol. 23, 692–703 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chan, M. M. et al. Molecular recording of mammalian embryogenesis. Nature 570, 77–82 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chu, L.-F. et al. Single-cell RNA-seq reveals novel regulators of human embryonic stem cell differentiation to definitive endoderm. Genome Biol. 17, 173 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Morrison, G. M. et al. Anterior definitive endoderm from ESCs reveals a role for FGF signaling. Cell Stem Cell 3, 402–415 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Yasunaga, M. et al. Induction and monitoring of definitive and visceral endoderm differentiation of mouse ES cells. Nat. Biotechnol. 23, 1542–1550 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Tada, S. et al. Characterization of mesendoderm: a diverging point of the definitive endoderm and mesoderm in embryonic stem cell differentiation culture. Development 132, 4363–4374 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Zhong, W. et al. Wnt and Nodal signaling simultaneously induces definitive endoderm differentiation of mouse embryonic stem cells. Rom. J. Morphol. Embryol. Rev. Roum. Morphol. Embryol. 58, 527–535 (2017).

    Google Scholar 

  25. Ang, S. L. et al. The formation and maintenance of the definitive endoderm lineage in the mouse: involvement of HNF3/forkhead proteins. Dev. Camb. Engl. 119, 1301–1315 (1993).

    CAS  Google Scholar 

  26. Burtscher, I., Barkey, W. & Lickert, H. Foxa2-venus fusion reporter mouse line allows live-cell analysis of endoderm-derived organ formation. Genesis 51, 596–604 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Jaitin, D. A. et al. Massively parallel single cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. MGI-Mouse Gene Expression Database (GXD) (MGI); (Acc. date 12/04/2019) http://www.informatics.jax.org/expression.shtml

  29. Kojima, Y. et al. The transcriptional and functional properties of mouse epiblast stem cells resemble the anterior primitive streak. Cell Stem Cell 14, 107–120 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Snow, M. H. L. Gastrulation in the mouse: growth and regionalization of the epiblast. Development 42, 293–303 (1977).

    Article  Google Scholar 

  31. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  32. McKnight, K. D., Hou, J. & Hoodless, P. A. Dynamic expression of thyrotropin-releasing hormone in the mouse definitive endoderm. Dev. Dyn. 236, 2909–2917 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. 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 

  34. Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Barkas, N. et al. Joint analysis of heterogeneous single-cell RNA-seq dataset collections. Nat. Methods 16, 695–698 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. g:Profiler – a web server for functional enrichment analysis and conversions of gene lists; (Acc. date 3/9/2029) https://biit.cs.ut.ee/gprofiler/gost

  40. molecular_function Gene Ontology Term (GO:0003674); (Acc. date 3/9/2019) http://www.informatics.jax.org/vocab/gene_ontology

  41. Duncan, S. A. et al. Expression of transcription factor HNF-4 in the extraembryonic endoderm, gut, and nephrogenic tissue of the developing mouse embryo: HNF-4 is a marker for primary endoderm in the implanting blastocyst. Proc. Natl Acad. Sci. USA 91, 7598–7602 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mfopou, J. K. et al. Efficient definitive endoderm induction from mouse embryonic stem cell adherent cultures: a rapid screening model for differentiation studies. Stem Cell Res. 12, 166–177 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Villegas, S. N. et al. PI3K/Akt1 signalling specifies foregut precursors by generating regionalized extra-cellular matrix. eLife 2, e00806 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Rothová, M., Hölzenspies, J. J., Livigni, A., Villegas, S. N. & Brickman, J. M. Differentiation of mouse embryonic stem cells into ventral foregut precursors. Curr. Protoc. Stem Cell Biol. 36, 1G.3.1–1G.3.12 (2016).

    Article  Google Scholar 

  45. Bhaduri, A. et al. Cell stress in cortical organoids impairs molecular subtype specification. Nature 578, 1–7 (2020).

    Article  CAS  Google Scholar 

  46. Chen, Y.-J. J. et al. Single-cell RNA sequencing identifies distinct mouse medial ganglionic eminence cell types. Sci. Rep. 7, 45656 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hwang, Y. S. et al. Reconstitution of prospermatogonial specification in vitro from human induced pluripotent stem cells. Nat. Commun. 11, 5656 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Simunovic, M. & Brivanlou, A. H. Embryoids, organoids and gastruloids: new approaches to understanding embryogenesis. Dev. Camb. Engl. 144, 976–985 (2017).

    CAS  Google Scholar 

  49. Amadei, G. et al. Inducible stem-cell-derived embryos capture mouse morphogenetic events in vitro. Dev. Cell 56, 366–382 (2020).

    Article  PubMed  CAS  Google Scholar 

  50. 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 

  51. Zhang, R.-R. et al. Human iPSC-derived posterior gut progenitors are expandable and capable of forming gut and liver organoids. Stem Cell Rep. 10, 780–793 (2018).

    Article  CAS  Google Scholar 

  52. Aggarwal, C. C., Hinneburg, A. & Keim, D. A. in Database Theory — ICDT 2001 (eds. Van den Bussche, J. & Vianu, V.) 420–434 (Springer, 2001). https://doi.org/10.1007/3-540-44503-X_27

  53. Vershynin, R (ed.) High-Dimensional Probability: An Introduction with Applications in Data Science 38–69 (Cambridge University Press, 2018). https://doi.org/10.1017/9781108231596.006

  54. Blum, A., Hopcroft, J. & Kannan, R. Foundations of Data Science (Cambridge University Press, 2020). https://doi.org/10.1017/9781108755528

  55. Maaten, Lvander & Hinton, G. Visualizing Data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  56. Pearson, K. LIII. On lines and planes of closest fit to systems of points in space. https://doi.org/10.1080/14786440109462720 (1901).

  57. McLean, A. B. et al. Activin A efficiently specifies definitive endoderm from human embryonic stem cells only when phosphatidylinositol 3-kinase signaling is suppressed. Stem Cells 25, 29–38 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Ortmann, D. et al. Naive pluripotent stem cells exhibit phenotypic variability that is driven by genetic variation.Cell Stem Cell 27, 470–481.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ang, S. L. & Rossant, J. HNF-3 beta is essential for node and notochord formation in mouse development. Cell 78, 561–574 (1994).

    Article  CAS  PubMed  Google Scholar 

  60. Ptashne, M. A genetic switch: phage [lambda] and higher organisms (Cell Press: Blackwell Scientific Publications, 1992).

  61. Lawson, K. A., Meneses, J. J. & Pedersen, R. A. Clonal analysis of epiblast fate during germ layer formation in the mouse embryo. Dev. Camb. Engl. 113, 891–911 (1991).

    CAS  Google Scholar 

  62. Riveiro, A. R. & Brickman, J. M. From pluripotency to totipotency: an experimentalist’s guide to cellular potency. Development 147, dev189845 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. 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 

  64. Linneberg-Agerholm, M. et al. Naïve human pluripotent stem cells respond to Wnt, Nodal and LIF signalling to produce expandable naïve extra-embryonic endoderm. Development 146, dev180620 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. Linneberg-Agerholm, M. & Brickman, J. M. in Human Naïve Pluripotent Stem Cells (ed. Rugg-Gunn, P.) 105–116 (Springer US, 2022). https://doi.org/10.1007/978-1-0716-1908-7_8

  66. McCracken, K. W., Howell, J. C., Wells, J. M. & Spence, J. R. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat. Protoc. 6, 1920–1928 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rodrigues, O. R. & Monard, S. A rapid method to verify single-cell deposition setup for cell sorters.Cytometry A 89, 594–600 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Keren-Shaul, H. et al. MARS-seq2.0: an experimental and analytical pipeline for indexed sorting combined with single-cell RNA sequencing. Nat. Protoc. 14, 1841–1862 (2019).

    Article  CAS  PubMed  Google Scholar 

  69. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  71. cell-cycle genes (cc.genes) for mouse · Issue #2493 · satijalab/seurat (GitHub) https://github.com/satijalab/seurat/issues/2493

  72. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Giladi, A. et al. Single-cell characterization of haematopoietic progenitors and their trajectories in homeostasis and perturbed haematopoiesis. Nat. Cell Biol. 20, 836–846 (2018).

    Article  CAS  PubMed  Google Scholar 

  74. Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods 17, 261–272 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Groiss, S. et al. Highly resolved spatial transcriptomics for detection of rare events in cells. https://doi.org/10.1101/2021.10.11.463936 (2021).

  76. Guilliams, M. et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell 185, 379–396.e38 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. D’Gama, P. P. et al. Diversity and function of motile ciliated cell types within ependymal lineages of the zebrafish brain. Cell Rep. 37, 109775 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Palla, G. et al. Squidpy: a scalable framework for spatial single cell analysis. https://doi.org/10.1101/2021.02.19.431994 (2021).

  81. Chen, T., & Guestrin, C. (2016). XGBoost: A Scalable Tree Boosting System. In Proceedings of the 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining (pp. 785–794). New York, NY, USA: ACM. https://doi.org/10.1145/2939672.2939785

  82. Bergen, V. et al. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

  83. Bergen, V. et al. RNA velocity—current challenges and future perspectives. Mol. Syst. Biol. 17, e10282 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Lickert for the kind gift of the FOXA2Venus reporter mice, the DanStem Genomics Platform (H. Neil, M. Michaut and H. Wollmann), DanStem Flow Cytometry Platform (G. dela Cruz and P. van Dieken), Stem Cell Culture Platform (A. Meligkova, E. F. Rebollo and M. Paulsen), DanStem Imaging Platform (J. Bulkescher and A. Shrestha) for technical expertise, support and the use of instruments, M.S. Paulsen for facilitating ST, Brickman laboratory members for critical discussions, K. J. Won for comments and T. Machet for graphical advice and help. Raw sequencing data were converted from bcl to fastq using the DeiC National Life Science Supercomputer at DTU (www.computerome.dk).

Work in our groups was supported by grants from the Lundbeck Foundation (R198-2015-412), J.M.B.; Independent Research Fund Denmark (8020-00100B and 6110-00009), J.M.B.; the Novo Nordisk Foundation (NNF17OC0028218), J.M.B.; the Danish National Research Foundation (DNRF116), J.M.B. and A.T. The Novo Nordisk Foundation Center for Stem Cell Medicine is supported by Novo Nordisk Foundation (NNF21CC0073729), and its predecessor The Novo Nordisk Foundation Center for Stem Cell Biology was also supported by the Novo Nordisk Foundation (NNF17CC0027852).

Author information

Authors and Affiliations

Authors

Contributions

M.M.R., J.M.B. and A.T. conceived the study. M.M.R., J.M.B., A.T., A.V.N., A.R.R., M.L-A. and Y.F.W. designed and interpreted experiments. M.M.R. performed all experiments, Y.F.W, A.R.R. and M.L.-A. contributed to the nEnd-spheroid experiments. The pre-processing and initial filtering of the raw sequencing data was done by E.D. All subsequent data analysis was carried out by A.V.N. and M.P. with input from I.A. M.M.R., J.M.B. and A.T. wrote the manuscript with input from all other authors.

Corresponding authors

Correspondence to Ala Trusina or Joshua Mark Brickman.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Heiko Lickert, Manu Setty, and Patrick Tam 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 Reproducibility and gene expression in Foxa2-Venus populations.

(a) Sample representation within each developmental stage. The samples are evenly distributed across the appropriate clusters. (b) Expression of known lineage markers agrees with the assigned cluster identities. aDE – anterior definitive endoderm, AVE – anterior visceral endoderm, DE1- early definitive endoderm, DE2 – later definitive endoderm, EmVE – embryonic visceral endoderm, ExVE – extra-embryonic endoderm, FG – foregut, FP – floorplate, HG – hindgut, InterVE – intermediate visceral endoderm, MG – midgut, PE – parietal endoderm, PS1 – early primitive streak, PS2 – later primitive streak.

Extended Data Fig. 2 Foxa2-Venus populations.

(a) Single cell FACS strategy. The sorted cells were consecutively gated in order to remove cell debris, to select singlet from multiplet cells and to select live cells. A general gate was created in the FSC-A vs SSC-A plot to include the population of interest and minimize low FSC and high SSC events. The events in this gate were then gated for single cells based on FSC-W to exclude doublets and aggregates. The single cells were then gated for DAPI exclusion. Single-color controls were used to determine the gates for the different fluorophores. The FOXA2POS cells were mixed with pre-labelled carrier cells (labelled with CellVue Maroon) and the FOXA2POS cells (negative for CellVue Maroon staining) were used for the final index single cell sort. Each cell specifications (such as forward scatter - cell size and FOXA2-Venus protein levels) were recorded and linked to a specific sorted cell. (b) Bar plot representation of samples per cluster. Each colour represents a time point between E6.5 and E9.5. (c) Images of E6.5 - E9.5 FOXA2POS embryos, the E8.5 and E9.5 embryos were dissected as illustrated. (d) Re-clustered UMAP dimensional embedding of gastrulation stage lineages. Here, the Node segregates into two clusters. (e) FACS and cell cycle chart for gastrulation stage lineages representing the cell size (radius of circles), Foxa2 mRNA level (X-axis), FOXA2 protein level (Y-axis) and cell cycle phase (colour of pie charts, G1-phase in red, G2M-phase in green, and S-phase in blue) for specific clusters. The Node is represented by a proliferative and non-proliferative cell populations. AVE – anterior visceral endoderm, DE1- early definitive endoderm, DE2 – later definitive endoderm, EmVE – embryonic visceral endoderm, ExVE – extra-embryonic endoderm, InterVE – intermediate visceral endoderm, PS1 – early primitive streak, PS2 – later primitive streak.

Source data

Extended Data Fig. 3 CAT analysis of different endoderm populations.

(a) A heatmap of Euclidean distances between the clusters in our in vivo and Now-2019 dataset. (b) An example of bootstrapped distance histograms illustrating that our InterVE cluster aligns to Now-EmVE2 (in green) and not other clusters in Now-2019 dataset (red). (c-e) Sankey diagrams representing alignments between clusters in: (c) our in vivo to Now-2019 dataset (before sub-clustering); (d) Now-2019 to our in vivo dataset; (e) our in vivo dataset aligned to itself. (c-e) Thickness of the line is inversely proportional to the pairwise distance. (f) PCA of all FOXA2POS cells places the InterVE between the extra-embryonic VE and embryonic DE lineages. AVE – anterior visceral endoderm, DE1- early definitive endoderm, DE2 – later definitive endoderm, EmVE – embryonic visceral endoderm, ExVE – extra-embryonic endoderm, FP – floorplate, InterVE – intermediate visceral endoderm, PE – parietal endoderm, PS1 – early primitive streak, PS2 – later primitive streak.

Extended Data Fig. 4 PAGA trajectory inference on DE and VE lineages.

(a-b) Partition-based graph abstraction (PAGA) of VE and DE FOXA2POS lineages. Similar to PCA, the PAGA trajectory inference highlights the position of InterVE in between the VE and DE lineages. AVE – anterior visceral endoderm, DE1- early definitive endoderm, DE2 – later definitive endoderm, EmVE – embryonic visceral endoderm, ExVE – extra-embryonic endoderm, InterVE – intermediate visceral endoderm.

Extended Data Fig. 5 Molecular basis for VE-InterVE-DE transition.

(a) Heatmap representing DEGs in the InterVE cluster. Gene expression in this heatmap includes both DE-expressed genes (first part of the heatmap) and VE-expressed genes (the second part of heatmap), which are co-expressed in the InterVE cluster. Genes are hierarchically clustered. (b) Cumulative gene expression of Smoothened GO-FC. Boxplots show median value, bordered by upper/lower (75/25) percentile, whiskers showing max/min value in 1.5 * IQR (interquartile range) and outliers as black dots. Two-sided Mann-Whitney U test was used to determine the significance between AVE and InterVE as well as DE2 and InterVE; *=<0.05; **=<0.01; ***=<0.001; ns=non-significant. (c) CAT networks representing examples of GO-FC, where InterVE is fully separated from VE-lineages and aligned with DE-lineages. The VE-lineages are in yellow, DE-lineages in blue, InterVE in green, and other lineages in pink. (d) CAT networks representing examples of GO-FC, where InterVE is aligned with VE-lineages and separated from DE-lineages. The VE-lineages are in yellow, DE-lineages in blue, InterVE in green, and other lineages in pink. AVE – anterior visceral endoderm, DE1- early definitive endoderm, DE2 – later definitive endoderm, EmVE – embryonic visceral endoderm, ExVE – extra-embryonic endoderm, FG – foregut, FP – floorplate, HG – hindgut, InterVE – intermediate visceral endoderm, LIV – liver, MG – midgut, NOTO – notochord, PE – parietal endoderm, PS1 – early primitive streak, PS2 – later primitive streak.

Extended Data Fig. 6 Spatial transcriptomics revealing InterVE in E7.5 embryos.

(a) Feature importance plot generated by decision tree showing the most important genes to enable the identification of the InterVE population. (b) Heatmap representing InterVE cells and 25 genes used for spatial transcriptomics. (c) Similar pattern as in (b) is obtained when the same 25 genes are projected in a heatmap of the identified InterVE cells in the embryo section in Fig. 4d. (d) Multiple sections of E7.5 embryos showing InterVE cells (in red, probability>0.5) identified by the decision tree. The InterVE cells are present on both anterior and posterior sides of the embryo but at this stage they are found predominantly in the anterior side. Colour-code represents the probability of a cell being InterVE. (e) PAGA trajectory inference of selected clusters in Nowotschin-2019 dataset. EmVE2 (corresponding to our InterVE) connects to various gut clusters along the anterior posterior axis of the developing gut. (f) Dotplot of AVE markers in the subclustered Nowotchin-2019 dataset. Based on CAT alignment (Fig. 2c), embryonic stage (Supplementary Dataset 4) and markers shown here, EmVE0 represents AVE at E6.5, EmVE3 and EmVE4 represent AVE at E5.5. (g) Sankey diagram representing alignments in between the Now-EmVE sub-clusters. EmVE2 (equivalent to our InterVE, Fig. 2c) is aligning with EmVE0 (equivalent to our AVE) suggesting a link between InterVE and AVE. DE - definitive endoderm, VE – visceral endoderm.

Extended Data Fig. 7 Differentiation of ESCs in vitro to generate distinct in vivo-like populations.

(a) Violin plots represent gene expression of the four most common genes used to purify DE in vitro. Two-sided Mann-Whitney U was used to determine the significance between VE clusters (AVE, EmVE, ExVE1 and ExVE2) and DE clusters (DE1 and DE2); *=<0.05; **=<0.01; ***=<0.001; ns=non-significant. (b) FACS plots showing the sorting strategy for isolating specific cell populations at various stages of 2D-ESC differentiation using the Gsc-GFP/Hhex-RedStar double reporter. (c) Sample representation within each differentiation protocol. Samples distribute evenly across the appropriate clusters. (d) Expression of known lineage markers agrees with the assigned cluster identities. DE- definitive endoderm, PrE – primitive endoderm, PS- primitive streak, VE –visceral endoderm.

Extended Data Fig. 8 Comparison of ESC in vitro differentiation protocols to in vivo populations.

(a) UMAP dimensional embedding of 2D-ESC and 2D-ESC-Pi3ki datasets. (b-c) Sankey diagrams representing significant alignments between clusters from 2D-ESC + PI3Ki protocols and our in vivo dataset. The alignments to DE2 and hindgut lineages are highlighted in red in (b) for fraction of 2D-ESC cells and in (c) for fraction of 2D-ESC-PI3Ki cells. The height of the cluster-bars to the left represents the proportion of cells in each cluster from (b) 2D-protocol and (c) PI3Ki-protocol. The width of the lines is given by the cluster-height divided by the number of alignments. D4 – Day 4, D5 – Day 5, D6 – Day 6. (d) UMAP dimensional embedding of 2D-ESC and 3D-ESC protocol highlighting that even when 2 different cell lines are used, the 2D and 3D protocols cluster together. (e) Scheme of calculating similarity measure S = 1-n_DE/N_E (1-n_EE/(n_DE (n_DE-1))). In this example, the total number of in vivo lineages, N_E = 5; in the bottom right panel, number of alignments, n_DE = 4 (red and green arrows); number of alignments among the four in vivo lineages n_EE = 2. DE1- early definitive endoderm, DE – definitive endoderm, DE2 – later definitive endoderm, FP – floorplate progenitors, PS2 – later primitive streak.

Extended Data Fig. 9 Characterization of nEnd and nEnd vs ESC differentiation.

(a) Full heatmap representing differentially expressed PrE genes from Now-2019 dataset. All clusters besides nEnd1 and nEnd2 are from Now-2019 dataset. The nEnd1, nEnd2, Now-PrE and Now-ParE form a hierarchal cluster. (b) Immuno-fluorescence staining of nEnd-spheroids for CDX2, FOXA2 and E-CAD. Results are representative of 4 independent experiments. (c) RT-qPCR of InterVE, nEnd and gut markers during the nEnd spheroid differentiation. The bars represent standard error of the mean over n = 4 biologically independent experiments. Unpaired t-test was used to determine the significance; * = <0.05; **=<0.01; ***=<0.001; ****=<0.0001. (d) Dotplot showing nEnd, InterVE and gut markers in MARS-seq dataset consisting of nEnd cells differentiating into nEnd spheroids. (e) RNA velocity on nEnd differentiation into nEnd spheroids. (f) RNA velocity on ESC differentiation using the 2D protocol. (g) Sankey diagram representing alignments between the spheroid gut differentiation and our in vivo dataset. DE - definitive endoderm, InterVE – intermediate visceral endoderm, nEnd – naïve extra-embryonic endoderm, PS – primitive streak. AVE – anterior visceral endoderm, DE2 – later definitive endoderm, EmVE – embryonic visceral endoderm, ExVE – extra-embryonic endoderm, InterVE – Intermediate visceral endoderm, PE – parietal endoderm.

Source data

Supplementary information

Reporting Summary

Supplementary Data 1

List of in vivo DEGs for all defined cell types. Gene significance and fold-change (log10 base) was determined using Mann–Whitney U test, followed by Bonferroni correction for multiple testing. List of markers were filtered as follows: average log fold change >0.5 and adjusted P value <0.05.

Supplementary Data 2

Distribution of individual samples across each cluster in the in vivo dataset.

Supplementary Data 3

DEGs of subclustered in vivo dataset including PS, DE, node and visceral endoderm. Gene significance and fold-change (log10 base) was determined using Mann–Whitney U test, followed by Bonferroni correction for multiple testing. List of markers were filtered as follows: average log fold change >0.5 and adjusted P value <0.05.

Supplementary Data 4

Distribution of E5.5–E7.5 samples across EmVE clusters in Nowotschin-2019 dataset.

Supplementary Data 5

DEGs between E7.5 and E8.5 DE clusters in the in vivo dataset.

Supplementary Data 6

GO analysis of DEGs between InterVE and other VE clusters and between InterVE and DE2.

Supplementary Data 7

List of genes included in each GO-FC.

Supplementary Data 8

Overview of in vitro protocols used in this study.

Supplementary Data 9

Number of cells FACS-sorted for each in vitro protocol used in this study.

Supplementary Data 10

Overview of genes contributing to the low similarity measure for each GO-FC.

Supplementary Data 11

List of antibodies used in this study.

Supplementary Data 12

List of primers and UPL probes used in this study.

Supplementary Data 13

Details for single-cell resolution including barcodes, reads, mapping and genes.

Supplementary Data 14

Gene names and catalogue numbers for the specific probes designed by Resolve BioSciences.

Source data

Source Data Fig. 2

Statistical source data for bar plots in Fig. 2e.

Source Data Fig. 3

Source data for Fig. 3b.

Source Data Fig. 5

Source data for Fig. 5f.

Source Data Extended Data Fig. 2

Source data for Extended Data Fig. 2b.

Source Data Extended Data Fig. 9

Statistical source data for RT–qPCR in Extended Data Fig. 9c.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rothová, M.M., Nielsen, A.V., Proks, M. et al. Identification of the central intermediate in the extra-embryonic to embryonic endoderm transition through single-cell transcriptomics. Nat Cell Biol 24, 833–844 (2022). https://doi.org/10.1038/s41556-022-00923-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00923-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