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.

  • Resource
  • Published:

An atlas of rabbit development as a model for single-cell comparative genomics

Abstract

Traditionally, the mouse has been the favoured vertebrate model for biomedical research, due to its experimental and genetic tractability. However, non-rodent embryological studies highlight that many aspects of early mouse development, such as its egg-cylinder gastrulation and method of implantation, diverge from other mammals, thus complicating inferences about human development. Like the human embryo, rabbits develop as a flat-bilaminar disc. Here we constructed a morphological and molecular atlas of rabbit development. We report transcriptional and chromatin accessibility profiles for over 180,000 single cells and high-resolution histology sections from embryos spanning gastrulation, implantation, amniogenesis and early organogenesis. Using a neighbourhood comparison pipeline, we compare the transcriptional landscape of rabbit and mouse at the scale of the entire organism. We characterize the gene regulatory programmes underlying trophoblast differentiation and identify signalling interactions involving the yolk sac mesothelium during haematopoiesis. We demonstrate how the combination of both rabbit and mouse atlases can be leveraged to extract new biological insights from sparse macaque and human data. The datasets and computational pipelines reported here set a framework for a broader cross-species approach to decipher early mammalian development, and are readily adaptable to deploy single-cell comparative genomics more broadly across biomedical research.

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: Rabbit timeline of development and high-resolution imaging series of rabbit embryos.
Fig. 2: Rabbit transcriptional landscape of gastrulation and early organogenesis.
Fig. 3: Chromatin accessibility and gene expression along the trajectory of early SCT differentiation.
Fig. 4: Rabbit and mouse neighbourhood comparisons identify regions of similarity and dissimilarity across species.
Fig. 5: Automated annotation models trained on the rabbit atlas accurately classify cell types in sparse human and macaque data.
Fig. 6: Rabbit YS haematopoiesis exhibits conserved molecular markers with human in vitro development models and reveals signalling role of mesothelium.

Similar content being viewed by others

Data availability

Raw sequencing data are available through ArrayExpress with the following accessions: scRNA-seq: E-MTAB-11836; scATAC-seq: E-MTAB-11804. Raw histology and RNAscope imaging files are available through the EBI BioImage archive under accession S-BIAD604. All other links to the data are available at https://marionilab.github.io/RabbitGastrulation2022/. This includes links to processed single-cell transcriptomics and ATAC-seq data in a variety of formats for loading into both R and Python analysis pipelines. The transcriptomics and imaging data are also available to explore interactively via a web app accessible through the same link. Low-resolution thumbnail images for all generated imaging datasets are additionally provided. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

Codes are available at https://github.com/MarioniLab/RabbitGastrulation2022 and https://marionilab.github.io/RabbitGastrulation2022/.

References

  1. Leopardo, N. P. & Vitullo, A. D. Early embryonic development and spatiotemporal localization of mammalian primordial germ cell-associated proteins in the basal rodent Lagostomus maximus. Sci. Rep. 7, 594 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Yoshida, M. et al. Conserved and divergent expression patterns of markers of axial development in eutherian mammals. Dev. Dyn. 245, 67–86 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Lee, K. Y. & DeMayo, F. J. Animal models of implantation. Reproduction 128, 679–695 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. 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 

  5. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pijuan-Sala, B. et al. Single-cell chromatin accessibility maps reveal regulatory programs driving early mouse organogenesis. Nat. Cell Biol. 22, 487–497 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Argelaguet, R. et al. Multi-omics profiling of mouse gastrulation at single-cell resolution. Nature https://doi.org/10.1038/s41586-019-1825-8 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Ericsson, A. C., Crim, M. J. & Franklin, C. L. A brief history of animal modeling. Mo. Med. 110, 201–205 (2013).

    PubMed  PubMed Central  Google Scholar 

  9. Rabl, C. Édouard van Beneden und der gegenwärtige Stand der wichtigsten von ihm behandelten Probleme. Arch. Mikrosk. Anat. 88, 3–470 (1915).

    Article  Google Scholar 

  10. Hassoun, R., Schwartz, P., Rath, D., Viebahn, C. & Männer, J. Germ layer differentiation during early hindgut and cloaca formation in rabbit and pig embryos. J. Anat. 217, 665–678 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Püschel, B. et al. The rabbit (Oryctolagus cuniculus): a model for mammalian reproduction and early embryology. Cold Spring Harb. Protoc. 2010, db.emo139 (2010).

    Article  Google Scholar 

  12. Fischer, B., Chavatte-Palmer, P., Viebahn, C., Navarrete Santos, A. & Duranthon, V. Rabbit as a reproductive model for human health. Reproduction 144, 1–10 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Esteves, P. J. et al. The wide utility of rabbits as models of human diseases. Exp. Mol. Med. 50, 1–10 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Upham, N. S., Esselstyn, J. A. & Jetz, W. Inferring the mammal tree: species-level sets of phylogenies for questions in ecology, evolution, and conservation. PLoS Biol. 17, e3000494 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kumar, V., Hallström, B. M. & Janke, A. Coalescent-based genome analyses resolve the early branches of the euarchontoglires. PLoS ONE 8, e60019 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Esselstyn, J. A., Oliveros, C. H., Swanson, M. T. & Faircloth, B. C. Investigating difficult nodes in the placental mammal tree with expanded taxon sampling and thousands of ultraconserved elements. Genome Biol. Evol. 9, 2308–2321 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Płusa, B. & Piliszek, A. Common principles of early mammalian embryo self-organisation. Development 147, dev183079 (2020).

    Article  PubMed  Google Scholar 

  18. Wang, H. & Dey, S. K. Roadmap to embryo implantation: clues from mouse models. Nat. Rev. Genet. 7, 185–199 (2006).

    Article  PubMed  Google Scholar 

  19. Nishimura, M. Timing of implantation in New Zealand White rabbits. Congenit. Anom. 41, 198–203 (2001).

    Article  Google Scholar 

  20. Tyser, R. C. V. et al. Single-cell transcriptomic characterization of a gastrulating human embryo. Nature 600, 285–289 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Rostovskaya, M., Andrews, S., Reik, W. & Rugg-Gunn, P. J. Amniogenesis occurs in two independent waves in primates. Cell Stem Cell 29, 744–759.e6 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ivanovs, A. et al. Human haematopoietic stem cell development: from the embryo to the dish. Development 144, 2323–2337 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Imaz-Rosshandler, I. et al. Tracking early mammalian organogenesis—prediction and validation of differentiation trajectories at whole organism scale. Preprint at bioRxiv https://doi.org/10.1101/2023.03.17.532833 (2023).

  24. Williams, B. S. & Biggers, J. D. Polar trophoblast (Rauber’s layer) of the rabbit blastocyst. Anat. Rec. 227, 211–222 (1990).

    Article  CAS  PubMed  Google Scholar 

  25. Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Meinhardt, G. et al. Pivotal role of the transcriptional co-activator YAP in trophoblast stemness of the developing human placenta. Proc. Natl Acad. Sci. USA 117, 13562–13570 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Huang, D. et al. The role of Cdx2 as a lineage specific transcriptional repressor for pluripotent network during the first developmental cell lineage segregation. Sci. Rep. 7, 17156 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Chiu, Y. H. & Chen, H. GATA3 inhibits GCM1 activity and trophoblast cell invasion. Sci. Rep. 6, 21630 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Simmons, D. G. et al. Early patterning of the chorion leads to the trilaminar trophoblast cell structure in the placental labyrinth. Development 135, 2083–2091 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Heidmann, O., Vernochet, C., Dupressoir, A. & Heidmann, T. Identification of an endogenous retroviral envelope gene with fusogenic activity and placenta-specific expression in the rabbit: a new ‘syncytin’ in a third order of mammals. Retrovirology 6, 107 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Steingrímsson, E., Tessarollo, L., Reid, S. W., Jenkins, N. A. & Copeland, N. G. The bHLH-Zip transcription factor Tfeb is essential for placental vascularization. Development 125, 4607–4616 (1998).

    Article  PubMed  Google Scholar 

  32. Zadora, J. et al. Disturbed placental imprinting in preeclampsia leads to altered expression of DLX5, a human-specific early trophoblast marker. Circulation 136, 1824–1839 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dann, E., Henderson, N. C., Teichmann, S. A., Morgan, M. D. & Marioni, J. C. Differential abundance testing on single-cell data using k-nearest neighbor graphs. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-01033-z (2021).

    Article  PubMed  Google Scholar 

  34. Pereira, P. N. G. et al. Amnion formation in the mouse embryo: the single amniochorionic fold model. BMC Dev. Biol. 11, 48 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Foote, R. H. & Carney, E. W. The rabbit as a model for reproductive and developmental toxicity studies. Reprod. Toxicol. 14, 477–493 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. 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 

  37. Lawson, K. A. et al. Bmp4 is required for the generation of primordial germ cells in the mouse embryo. Genes Dev. 13, 424–436 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

  39. Kobayashi, T. & Surani, M. A. On the origin of the human germline. Development 145, dev150433 (2018).

  40. Nowotschin, S., Hadjantonakis, A.-K. & Campbell, K. The endoderm: a divergent cell lineage with many commonalities. Development 146, dev150920 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Alberio, R., Kobayashi, T. & Surani, M. A. Conserved features of non-primate bilaminar disc embryos and the germline. Stem Cell Rep. 16, 1078–1092 (2021).

    Article  CAS  Google Scholar 

  42. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kobayashi, T. et al. Tracing the emergence of primordial germ cells from bilaminar disc rabbit embryos and pluripotent stem cells. Cell Rep. 37, 109812 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. Yang, R. et al. Amnion signals are essential for mesoderm formation in primates. Nat. Commun. 12, 5126 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nakamura, T., Fujiwara, K., Saitou, M. & Tsukiyama, T. Non-human primates as a model for human development. Stem Cell Rep. 16, 1093–1103 (2021).

    Article  CAS  Google Scholar 

  46. Enders, A. C., Hendrickx, A. G. & Schlafke, S. Implantation in the rhesus monkey: initial penetration of endometrium. Am. J. Anat. 167, 275–298 (1983).

    Article  CAS  PubMed  Google Scholar 

  47. Dyer, M. A., Farrington, S. M., Mohn, D., Munday, J. R. & Baron, M. H. Indian hedgehog activates hematopoiesis and vasculogenesis and can respecify prospective neurectodermal cell fate in the mouse embryo. Development 128, 1717–1730 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Barminko, J., Reinholt, B. & Baron, M. H. Development and differentiation of the erythroid lineage in mammals. Dev. Comp. Immunol. 58, 18–29 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Cindrova-Davies, T. et al. RNA-seq reveals conservation of function among the yolk sacs of human, mouse, and chicken. Proc. Natl Acad. Sci. USA 114, E4753–E4761 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Damert, A., Miquerol, L., Gertsenstein, M., Risau, W. & Nagy, A. Insufficient VEGFA activity in yolk sac endoderm compromises haematopoietic and endothelial differentiation. Development 129, 1881–1892 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Bruveris, F. F., Ng, E. S., Stanley, E. G. & Elefanty, A. G. VEGF, FGF2, and BMP4 regulate transitions of mesoderm to endothelium and blood cells in a human model of yolk sac hematopoiesis. Exp. Hematol. 103, 30–39.e2 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Minot, C. S. & Taylor, E. Normal Plates of the Development of the Rabbit, Lepus cuniculus L. (Fischer, 1905).

  54. 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 

  55. Martínez, N. & Damiano, A. E. Aquaporins in fetal development. Adv. Exp. Med. Biol. 969, 199–212 (2017).

    Article  PubMed  Google Scholar 

  56. Chuva de Sousa Lopes, S. M., Roelen, B. A. J., Lawson, K. A. & Zwijsen, A. The development of the amnion in mice and other amniotes. Philos. Trans. R. Soc. Lond. B 377, 20210258 (2022).

    Article  Google Scholar 

  57. Roost, M. S. et al. KeyGenes, a tool to probe tissue differentiation using a human fetal transcriptional atlas. Stem Cell Rep. 4, 1112–1124 (2015).

    Article  CAS  Google Scholar 

  58. 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 

  59. Yagi, R. et al. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development. Development 134, 3827–3836 (2007).

    Article  CAS  PubMed  Google Scholar 

  60. Baczyk, D. et al. Bi-potential behaviour of cytotrophoblasts in first trimester chorionic villi. Placenta 27, 367–374 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Kwak, Y.-T., Muralimanoharan, S., Gogate, A. A. & Mendelson, C. R. Human trophoblast differentiation is associated with profound gene regulatory and epigenetic changes. Endocrinology 160, 2189–2203 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yu, C. et al. GCMa regulates the syncytin-mediated trophoblastic fusion. J. Biol. Chem. 277, 50062–50068 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Baczyk, D. et al. Glial cell missing-1 transcription factor is required for the differentiation of the human trophoblast. Cell Death Differ. 16, 719–727 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Kuckenberg, P., Kubaczka, C. & Schorle, H. The role of transcription factor Tcfap2c/TFAP2C in trophectoderm development. Reprod. Biomed. Online 25, 12–20 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. Knöfler, M. et al. Human placenta and trophoblast development: key molecular mechanisms and model systems. Cell. Mol. Life Sci. 76, 3479–3496 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Millonig, J. H., Millen, K. J. & Hatten, M. E. The mouse Dreher gene Lmx1a controls formation of the roof plate in the vertebrate CNS. Nature 403, 764–769 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Lewis, S. L. & Tam, P. P. L. Definitive endoderm of the mouse embryo: formation, cell fates, and morphogenetic function. Dev. Dyn. 235, 2315–2329 (2006).

    Article  PubMed  Google Scholar 

  68. Tarashansky, A. J. et al. Mapping single-cell atlases throughout Metazoa unravels cell type evolution. eLife 10, e66747 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Coifman, R. R. et al. Geometric diffusions as a tool for harmonic analysis and structure definition of data: diffusion maps. Proc. Natl Acad. Sci. USA 102, 7426–7431 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Haghverdi, L., Buettner, F. & Theis, F. J. Diffusion maps for high-dimensional single-cell analysis of differentiation data. Bioinformatics 31, 2989–2998 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Haghverdi, L., Büttner, M., Wolf, F. A., Buettner, F. & Theis, F. J. Diffusion pseudotime robustly reconstructs lineage branching. Nat. Methods 13, 845–848 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  73. Tosches, M. A. et al. Evolution of pallium, hippocampus, and cortical cell types revealed by single-cell transcriptomics in reptiles. Science 360, 881–888 (2018).

    Article  CAS  PubMed  Google Scholar 

  74. Shafer, M. E. R. Cross-species analysis of single-cell transcriptomic data. Front. Cell Dev. Biol. 7, 175 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Katarzyna and the CRUK CI Genomics Core for their help with the 10x Genomics sample processing; R. Argelaguet for bioinformatic support; N. Wilson for assistance with the facilities and technical support; M. Rostovskaya for discussions around eutherian amnion; M. Keller (Gehlenborg lab) for help in setting up the Vitessce visualization platform. We also thank H. D. Farizi and S. D. Høiberg (Novo Nordisk) for help with rabbit embryo sectioning. M.-L.N.T. is funded by a Herchel Smith PhD Fellowship in Science. D.K. is funded by the Wellcome Mathematical Genomics and Medicine Programme at the University of Cambridge (PFZH/158 RG92770). C.G. was funded by the Swedish Research Council (2017-06278) and by a Swedish Childhood Cancer Fund position grant (TJ2021-0009). I.I.-R. was funded by the Wellcome Mathematical Genomics and Medicine Programme at the University of Cambridge (203942/Z/16/Z; RDAG/426RG86191). Work in the Gottgens group is supported by Wellcome, Bloodwise, MRC and CRUK, and by core support grants from Wellcome to the Wellcome-MRC Cambridge Stem Cell Institute. Work in the Marioni group is supported by core funding from CRUK (C9545/A29580) and by the European Molecular Biology Laboratory. Work in the È.B.-G. lab is supported by CRUK (C9545/A29580). This work was funded as part of a Wellcome grant (220379/B/20/Z) awarded to B.G., J.N. and J.C.M. and the WSSS Dev Ext Wellcome Grant (220152_C_20_Z) awarded to B.G. and J.C.M.

Author information

Authors and Affiliations

Authors

Contributions

C.G., M.-L.N.T. and J.N. performed embryo dissections and generated the scRNA-seq atlas dataset. M.-L.N.T. and F.J.C.-N. generated the scATAC-seq atlas dataset. J.A.-R. And T.K.A. supplied the animals for the experiments and conducted histology and imaging of the embryos. M.-L.N.T. performed pre-processing and initial low-level analysis for both atlases. D.K. performed batch correction and global visualization of the scRNA-seq dataset. D.K. performed cross-species comparisons between the rabbit mouse, macaque and human datasets. D.K. implemented the website. B.T. performed analysis on the scATAC-seq dataset. D.K., M.-L.N.T., B.P.-S., C.G., B.T. and I.I.-R. annotated atlas cell types. M.-L.N.T., B.T. and D.K. analysed the atlas trophoblast cells. J.N., È.B.-G., J.C.M. and B.G. supervised the study. M.-L.N.T., D.K., B.T., B.G., J.C.M. and È.B.-G. wrote the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Èlia Benito-Gutiérrez, John C. Marioni or Berthold Göttgens.

Ethics declarations

Competing interests

Provision of time-mated rabbit embryos by Labcorp Early Development Laboratories Limited as well as scRNA-seq costs were supported by a research contract agreement with J.A.-R. and T.K.A. at Novo Nordisk A/S. È.B.-G. and J.C.M. have been employees of Genentech since September 2022.

Peer review

Peer review information

Nature Cell Biology thanks Janet Rossant and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Histology and dissection images reveals early dynamics of early germ layers of rabbit flat-disk embryo.

a) Representative RNAscope image of GD7 rabbit sagittal in utero section showing beginning of gastrulation. TFAP2C and OCT4 mark trophoblast and epiblast-derived cell types, respectively. Arrowheads indicate implantation on the anti-mesometrial side of the embryo. b) Representative RNAscope image of GD8 rabbit sagittal in utero sections showing germ layer differentiation. (i) Arrowheads indicate implantation sides in the mesometrial direction. TFAP2C marks trophoblast and trophoblast progenitors. (ii) Chorioamnion fold formation can be seen alongside early germ layer differentiation. Chorion consists of a mesoderm inner layer, and amnion has an mesoderm outer layer as captured in the RNAscope image. c) Representative histology images of rabbit cross-sections in utero and magnification of the embryo to show details and orientation of the embryo in the uterus. cm, cranial mesoderm. d) Summary of over 650 + H&E and RNAscope images of rabbit sections generated in this study of which the representative images are pulled from. e) RNAscope image of the vitelline artery (VA) and dorsal aorta (DA) region showing the RUNX1 + CDH5+ presumptive haemogenic endothelium in the ventral portion of the dorsal aorta. f) Representative images of the rabbit embryos and their extraembryonic tissues utilized in this study g) 13 somite and 16 somite GD9 rabbit embryos microdissected into anterior, mid, and posterior regions for scRNA-seq.

Extended Data Fig. 2 Updated transcriptome annotation improves the quality of processed scRNA-seq data.

a, The distances of intergenic reads to their nearest annotated gene in a single GD8 sample. i) The distances for a subset of intergenic reads aligned to a region of chromosome 1 are shown. The vertical line indicates the 600 bp extension added to the 3′ ends (see Methods). ii) Distances are also shown for intergenic reads whose closest annotated gene is GATA1 and SOX9. b) The read coverage for the same GD8 sample shows that many reads associated with FOXA2 transcripts are positioned off the 3′ end of the OryCun 2.0 reference annotation. These missing reads are captured by our 600 bp extension. c) The 3′ extended gene annotations substantially improve the signal obtained after processing with Cell Ranger. d) Examples of new annotations added from aligning human gene annotations with the rabbit transcriptome. These new annotations overlap positions with high read coverage. e) The signal captured as a result of the new human gene annotations are consistent with the known expression patterns of SIX3 (forebrain marker) and FOXC1 (mesodermal marker). Intergenic read distances and normalised expression values are available in the source data.

Source data

Extended Data Fig. 3 RNA-seq quality controls.

a) Number of high-quality cells sampled from each of the 26 scRNA-seq samples. Annotations refer to the anatomical dissections performed for GD9 samples. b) Distribution of UMIs (c) genes detected and (d) percentage of mitochondrial reads recorded across cells sampled from each of the 26 scRNA-seq samples (N = 19 embryos), coloured by developmental stage. The lower and upper hinges of the boxplots relate to the first and third quartiles respectively. The center line denotes the median and the whiskers extend to the largest/smallest value no further than 1.5 times the inter-quartile range from the upper/lower hinge respectively. Outlier points are plotted if they exceed the ranges defined by the the upper and lower whiskers. The number of cells used to calculate each statistic are shown in A) and are provided in the source data. e) Cells from samples taken at each developmental stage and anatomical dissection are highlighted in the UMAP embedding. Following batch correction, the samples are well mixed within the UMAP visualization. A - anterior section, M - mid section; P - posterior section; YS -yolk-sac/extraembryonic tissues; EP - embryo proper. QC values for each cell are available in the source data.

Source data

Extended Data Fig. 4 Automated cell type annotation.

a, SingleR predicted cell type annotations for each cell of the rabbit atlas (n = 146,133 cells), following training on the mouse atlas. Only a subset of predicted labels are annotated for visual clarity. Predicted labels for all cells are more easily visualized via the interactive web app, accessible at https://marionilab.github.io/RabbitGastrulation2022/. b) Normalised SingleR correlation scores for each rabbit cell against all possible mouse labels. Cells are ordered according to the label assigned by SingleR. The final annotated cell types are also displayed to show how the model predictions relate with those assigned after validation. c) Cells of the rabbit (top) and mouse (bottom) (Imaz-Rosshandler et al.23) datasets in a SAMap integrated UMAP embedding (n = 576,472 cells). Cells are coloured and annotated according to the cell type labels of each respective atlas. The rabbit (r) and mouse (m) label numberings refer to those in the legends of Fig. 2c and Extended Data Fig. 4a respectively. The full list of IDs can also be found in Supplementary Tables 3–7. Shortened labels for the rabbit extra-embryonic ectoderm cell types have also been added. rT = Trophoblast; rA1 = Amnion 1; rA2 = Amnion 2; rA3 = Amnion 3; rCY = Cytotrophoblast; rSP = SCT progenitors; rES = Early SCT. All panels depict rabbit cells integrated across all samples (N = 19 embryos). SingleR correlation scores and cell type predictions are provided in the source data, along with the integrated UMAP coordinates and final cell type annotations.

Source data

Extended Data Fig. 5 Cell type validation.

a) Cell type proportions across each scRNA-seq sample (N = 19 embryos). Cell types labels are organised based on the stage and dissection they are most abundant. Cell types with low abundance are not shown to preserve visual clairity. b) Row-normalised gene expression plot showing genes differentially expressed within each cell type. c) RNAscope images of SERPINC1 and DKK1 helped identify cells of the parietal endoderm (PE). Scale bars = 500 μm. d) Row-normalised gene expression plot of marker genes used to annotate rabbit extra-embryonic ectoderm cell types. e) Histology and RNAscope images of a synctial trophoblastic knob expressing TFAP2C at gestational day 8. Scale bar = 100μm. Normalised expression values and cell type annotations (across cells/samples) are available in the Source Data.

Source data

Extended Data Fig. 6 Whole embryo scATAC-seq and trophoblast annotation.

a) An illustration of the 8 samples (N = 4 embryos) processed for single-cell ATAC-sequencing. b) Quality control of whole embryo scATAC-seq nuclei based on transcription start site (TSS) enrichment score, total number of fragments per cell, and fraction of reads in peaks (FRIP) across samples. The lower and upper hinges of the boxplots relate to the first and third quartiles respectively. The center line denotes the median and the whiskers extend to the largest/smallest value no further than 1.5 times the inter-quartile range from the upper/lower hinge respectively. Number of nuclei per sample - BGRGP1:26250; BGRGP2 12246; BGRGP3 13008; BGRGP4 4887; BGRGP5 12192; BGRGP6 11982; BGRGP7 11313; BGRGP8 10368. c) The fragment size distribution plotted for each scATAC-seq sample. d) UMAP of 34,082 nuclei that passed quality control for scATAC-seq, colored by predicted cell type inferred from label transfer (see Methods). Inset shows the UMAP colored by developmental stage. e) Linear regression between mean RNA expression and mean ATAC gene scores across cell-types, for top 50 marker genes of annotated cell-types. Top left panel shows linear regression along with values across cell-types for SCT marker genes, other panels only show the linear model itself. Red dotted lines indicate a slope of 1 (see Methods). f) Gene Activity scores of marker genes for refined trophoblast to SCT cell-type annotation. g) Motif enrichment in marker peaks for refined trophoblast to SCT cell-type annotation. h) Genome browser views of the regions surrounding EOMES, CDH5, FOXA2, GATA1 & HDAC6, HBG1/2, PAX6, TBX6 for selected cell-types. N = 4 embryos in all panels. QC values, fragment size distributions, UMAP coordinates, predicted annotations, linear model parameters and heatmap values are all available in the source data.

Source data

Extended Data Fig. 7 Rabbit-mouse neighborhood comparisons.

a) Distribution of the number of cells within neighbourhoods of the rabbit (n = 5,253 neighborhoods) and mouse atlas (n = 14,034 neighborhoods). b) Same plot as in Fig. 3d shown for all cell types. c) Neighbourhoods of the mouse atlas (n = 14,034 neighborhoods) coloured according to their maximum correlation with any rabbit neighbourhood. General UMAP positions for a subset of cell types are shown. d) Maximum correlation scores, grouped by the scRNA-seq sample (N = 19 embryos) of each neighbourhood index cell. Samples are ordered by mean maximum correlation and coloured by the developmental stage.Annotations above the GD9 samples refer to the anatomical dissection. The lower and upper hinges of the boxplots relate to the first and third quartiles respectively. The center line denotes the median and the whiskers extend to the largest/smallest value no further than 1.5 times the inter-quartile range from the upper/lower hinge respectively. Outlier points are plotted if they exceed the ranges defined by the the upper and lower whiskers. The number of neighbourhoods linked to each sample and thus used to calculate each statistic is written along the x-axis. A - anterior section, M - mid section; P - posterior section; YS -yolk-sac/extraembryonic tissues; EP - embryo proper. Neighbourhood sizes, annotations and maximum correlation values are provided in the source data.

Source data

Extended Data Fig. 8 Neighbourhood comparisons along differentiation trajectories.

a–c) Neighbourhoods associated with a) allantois, b) amnion and c) blood differentiation trajectories are shown for the rabbit and mouse. Maximally correlated neighbourhood pairs are linked, with the strength of correlation indicated with line colour. Neighbourhoods are coloured by annotated cell type. Neighborhoods in b, c are represented in the UMAP embedding, whereas in a, neighborhoods are positioned according to a ForceAtlas2 graph layout. d) Alluvial plot showing the proportion of rabbit gut tube neighbourhoods which form maximally correlated mappings with mouse neighbourhoods of different cell types. Rabbit neighborhoods represented in all panels were constructed from cells integrated across all samples (N = 19 embryos). Details of the neighbourhood mappings are provided in the source data.

Source data

Extended Data Fig. 9 Human and macaque cell type prediction with SingleR.

a) Alluvial plot showing the changes to the original cell type labels after predicting annotations with a SingleR model trained on the rabbit atlas. Mappings only shown for predicted cell types with more than 10 cells. b) UMAP of the CS7 human dataset (Tyser et al.20) coloured according to the SingleR cell type predictions trained on the mouse atlas (1,195 cells, N = 1 embryo). c) Same as in a for the classification of cells from the macaque in-vitro dataset (Yang et al.44). d) Same as in b for the Yang et al.44 dataset (7,194 cells, N = 12 embryos). The predicted cell type annotations are available in the source data.

Source data

Extended Data Fig. 10 Ligand-receptor interactions and gene ontology reveal roles of visceral endoderm and mesothelium in yolk sac hematopoietic niche.

a) Extended dot plot of CellphoneDB output reveals a large network of extracellular matrix protein interactions between the mesothelium and endothelium, as well as VEGF and FGF complexes. The order of the cell types along the x-axis indicates the order of the molecule1:molecule2 expression along the y axis. The size of the dot indicates the -log10(pvalue), while the color of the dot indicates the log2 mean of the mean expression of the molecules. Of note, YS endothelium and mesothelium show a significant interaction between FLT1 and FLT family complexes with VEGFA, which plays a known role in haematopoiesis. Statistical analyses were performed using the one-sided permutation test as described in Efremova et al.49 b) Gene ontology analysis of the differential transcriptional expression of the visceral endoderm clusters and the mesothelium clusters reveal different roles in the yolk sac niche. Visceral endoderm cells score highly on nutrient transport roles like chylomicron assembly, cholesterol efflux, and cholesterol homeostasis (left). Mesothelium transcripts interact highly with the endothelium according to (A), and score highly on extracellular matrix organization and collagen fibril organization. This indicates a closer interaction with endothelium that is critical for the function of the yolk sac hematopoietic niche and the mesothelium provides a role in structure separate from the visceral endoderm cells. P-values are adjusted for multiple-testing. Differential expression results are available in the source data. Statistical analyses were performed using Fisher’s exact test on cells integrated across all samples (N = 19 embryos) where those cell types were present.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1. scRNA-seq sample submission data. Supplementary Table 2. scATAC sample submission data. Supplementary Table 3. List of cell type annotations with a one-to-one correspondance between the rabbit and mouse Imaz-Rosshandler et al.23 dataset. Supplementary Table 4. Cell type annotations with a one-to-many correspondance with cell types defined in the Imaz-Rosshandler23 mouse dataset. Supplementary Table 5. Cell type labels that are variations of those defined in the Imaz-Rosshandler et al.23 mouse dataset. Supplementary Table 6. Cell types defined exclusively in the rabbit dataset. Supplementary Table 7. Cell types defined exclusively in the Imaz-Rosshandler et al.23 mouse dataset.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig./Table 2

Statistical source data.

Source Data Extended Data Fig./Table 3

Statistical source data.

Source Data Extended Data Fig./Table 4

Statistical source data.

Source Data Extended Data Fig./Table 5

Statistical source data.

Source Data Extended Data Fig./Table 6

Statistical source data.

Source Data Extended Data Fig./Table 7

Statistical source data.

Source Data Extended Data Fig./Table 8

Statistical source data.

Source Data Extended Data Fig./Table 9

Statistical source data.

Source Data Extended Data Fig./Table 10

Statistical source data.

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

Ton, ML.N., Keitley, D., Theeuwes, B. et al. An atlas of rabbit development as a model for single-cell comparative genomics. Nat Cell Biol 25, 1061–1072 (2023). https://doi.org/10.1038/s41556-023-01174-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01174-0

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