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:

Comprehensive spatiotemporal mapping of single-cell lineages in developing mouse brain by CRISPR-based barcoding

Abstract

A fundamental interest in developmental neuroscience lies in the ability to map the complete single-cell lineages within the brain. To this end, we developed a CRISPR editing-based lineage-specific tracing (CREST) method for clonal tracing in Cre mice. We then used two complementary strategies based on CREST to map single-cell lineages in developing mouse ventral midbrain (vMB). By applying snapshotting CREST (snapCREST), we constructed a spatiotemporal lineage landscape of developing vMB and identified six progenitor archetypes that could represent the principal clonal fates of individual vMB progenitors and three distinct clonal lineages in the floor plate that specified glutamatergic, dopaminergic or both neurons. We further created pandaCREST (progenitor and derivative associating CREST) to associate the transcriptomes of progenitor cells in vivo with their differentiation potentials. We identified multiple origins of dopaminergic neurons and demonstrated that a transcriptome-defined progenitor type comprises heterogeneous progenitors, each with distinct clonal fates and molecular signatures. Therefore, the CREST method and strategies allow comprehensive single-cell lineage analysis that could offer new insights into the molecular programs underlying neural specification.

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: A Cre-driven lineage recording mouse model based on CRISPR barcoding.
Fig. 2: Transcriptional profiling of En1-derived cells in the developing mouse vMB.
Fig. 3: CREST barcode-based lineage relationships among cell clusters in the developing mouse vMB.
Fig. 4: Clonal analyses reveal variable fate outcomes for individual En1-derived vMB progenitors.
Fig. 5: Distinct differentiation trajectories and molecular programs for DA and GLU specification.
Fig. 6: CREST barcoding combined with ex vivo brain organoid culture and clonal splitting strategy reveals fate potentials of transcriptome-defined progenitors.

Similar content being viewed by others

Data availability

Sequencing raw data and counts matrices are available under accession number NCBI GEO: GSE210139. The reference genome mm10 is available from the 10X Genomics website https://support.10xgenomics.com/single-cell-gene-expression/software/release-notes/build. Raw imaging data have been deposited in Mendeley Data at https://data.mendeley.com/datasets/6b36kp78g5. Source data are provided with this paper.

Code availability

Computer code for single-cell transcriptome analysis and lineage analysis is available at https://github.com/Y0NEKO/ChenLab-CREST.

References

  1. Gao, P. et al. Deterministic progenitor behavior and unitary production of neurons in the neocortex. Cell 159, 775–788 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mayer, C. et al. Clonally related forebrain interneurons disperse broadly across both functional areas and structural boundaries. Neuron 87, 989–998 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Price, J., Turner, D. & Cepko, C. Lineage analysis in the vertebrate nervous system by retrovirus-mediated gene transfer. Proc. Natl Acad. Sci. USA 84, 156–160 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sultan, K. T. et al. Clonally related GABAergic interneurons do not randomly disperse but frequently form local clusters in the forebrain. Neuron 92, 31–44 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tabata, H. & Nakajima, K. Efficient in utero gene transfer system to the developing mouse brain using electroporation: visualization of neuronal migration in the developing cortex. Neuroscience 103, 865–872 (2001).

    Article  CAS  PubMed  Google Scholar 

  6. Ge, M. et al. A spacetime odyssey of neural progenitors to generate neuronal diversity. Neurosci. Bull. https://doi.org/10.1007/s12264-022-00956-0 (2022).

  7. Wang, Z. & Zhu, J. MEMOIR: a novel system for neural lineage tracing. Neurosci. Bull. 33, 763–765 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Zong, H., Espinosa, J. S., Su, H. H., Muzumdar, M. D. & Luo, L. Mosaic analysis with double markers in mice. Cell 121, 479–492 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Wagner, D. E. & Klein, A. M. Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet. 21, 410–427 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Biddy, B. A. et al. Single-cell mapping of lineage and identity in direct reprogramming. Nature 564, 219–224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rodriguez-Fraticelli, A. E. et al. Single-cell lineage tracing unveils a role for TCF15 in haematopoiesis. Nature 583, 585–589 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science https://doi.org/10.1126/science.aaw3381 (2020).

  13. You, Z. et al. Mapping of clonal lineages across developmental stages in human neural differentiation. Cell Stem Cell 30, 473–487.e479 (2023).

    Article  CAS  PubMed  Google Scholar 

  14. Bandler, R. C. et al. Single-cell delineation of lineage and genetic identity in the mouse brain. Nature 601, 404–409 (2022).

    Article  CAS  PubMed  Google Scholar 

  15. Ratz, M. et al. Clonal relations in the mouse brain revealed by single-cell and spatial transcriptomics. Nat. Neurosci. 25, 285–294 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Delgado, R. N. et al. Individual human cortical progenitors can produce excitatory and inhibitory neurons. Nature 601, 397–403 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Ma, J., Shen, Z., Yu, Y. C. & Shi, S. H. Neural lineage tracing in the mammalian brain. Curr. Opin. Neurobiol. 50, 7–16 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. McKenna, A. et al. Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science 353, aaf7907 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Kalhor, R. et al. Developmental barcoding of whole mouse via homing CRISPR. Science https://doi.org/10.1126/science.aat9804 (2018).

  20. Alemany, A., Florescu, M., Baron, C. S., Peterson-Maduro, J. & van Oudenaarden, A. Whole-organism clone tracing using single-cell sequencing. Nature 556, 108–112 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Raj, B. et al. Simultaneous single-cell profiling of lineages and cell types in the vertebrate brain. Nat. Biotechnol. 36, 442–450 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Spanjaard, B. et al. Simultaneous lineage tracing and cell-type identification using CRISPR-Cas9-induced genetic scars. Nat. Biotechnol. 36, 469–473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. He, Z. et al. Lineage recording in human cerebral organoids. Nat. Methods 19, 90–99 (2022).

    Article  PubMed  Google Scholar 

  24. Quinn, J. J. et al. Single-cell lineages reveal the rates, routes, and drivers of metastasis in cancer xenografts. Science https://doi.org/10.1126/science.abc1944 (2021).

  25. Simeonov, K. P. et al. Single-cell lineage tracing of metastatic cancer reveals selection of hybrid EMT states. Cancer Cell 39, 1150–1162.e1159 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yang, D. et al. Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution. Cell 185, 1905–1923.e1925 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bowling, S. et al. An engineered CRISPR-Cas9 mouse line for simultaneous readout of lineage histories and gene expression profiles in single cells. Cell 181, 1693–1694 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zeng, H. et al. An inducible and reversible mouse genetic rescue system. PLoS Genet. 4, e1000069 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Chi, C. L., Martinez, S., Wurst, W. & Martin, G. R. The isthmic organizer signal FGF8 is required for cell survival in the prospective midbrain and cerebellum. Development 130, 2633–2644 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Ciemerych, M. A. & Sicinski, P. Cell cycle in mouse development. Oncogene 24, 2877–2898 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. La Manno, G. et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell 167, 566–580.e519 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Bayer, S. A., Wills, K. V., Triarhou, L. C. & Ghetti, B. Time of neuron origin and gradients of neurogenesis in midbrain dopaminergic neurons in the mouse. Exp. Brain Res. 105, 191–199 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gong, W. et al. Benchmarked approaches for reconstruction of in vitro cell lineages and in silico models of C. elegans and M. musculus developmental trees. Cell Syst. 12, 810–826.e814 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. He, L. et al. Enhancing the precision of genetic lineage tracing using dual recombinases. Nat. Med. 23, 1488–1498 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Birtele, M. et al. Single-cell transcriptional and functional analysis of dopaminergic neurons in organoid-like cultures derived from human fetal midbrain. Development https://doi.org/10.1242/dev.200504 (2022).

  39. Alves dos Santos, M. T. & Smidt, M. P. En1 and Wnt signaling in midbrain dopaminergic neuronal development. Neural Dev. 6, 23 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Arenas, E. Wnt signaling in midbrain dopaminergic neuron development and regenerative medicine for Parkinson’s disease. J. Mol. Cell. Biol. 6, 42–53 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Ásgrímsdóttir, E. S. & Arenas, E. Midbrain dopaminergic neuron development at the single cell level: in vivo and in stem cells. Front. Cell Dev. Biol. 8, 463 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Li, L. et al. A mouse model with high clonal barcode diversity for joint lineage, transcriptomic, and epigenomic profiling in single cells. Preprint at bioRxiv https://doi.org/10.1101/2023.01.29.526062 (2023).

  43. Joksimovic, M. et al. Spatiotemporally separable Shh domains in the midbrain define distinct dopaminergic progenitor pools. Proc. Natl Acad. Sci. USA 106, 19185–19190 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Aguirre, A. J. et al. Genomic copy number dictates a gene-independent cell response to CRISPR/Cas9 targeting. Cancer Discov. 6, 914–929 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kimmel, R. A. et al. Two lineage boundaries coordinate vertebrate apical ectodermal ridge formation. Genes Dev. 14, 1377–1389 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Xu, Q., Tam, M. & Anderson, S. A. Fate mapping Nkx2.1-lineage cells in the mouse telencephalon. J. Comp. Neurol. 506, 16–29 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. 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 

  49. Puelles, E., Martínez-de-la-Torre, M., Watson, C. & Puelles, L. in The Mouse Nervous System (eds Watson, C. et al.) 337-359 (Academic Press, 2012).

  50. Madrigal, M. P., Moreno-Bravo, J. A., Martínez-López, J. E., Martínez, S. & Puelles, E. Mesencephalic origin of the rostral substantia nigra pars reticulata. Brain Struct. Funct. 221, 1403–1412 (2016).

    Article  PubMed  Google Scholar 

  51. Kala, K. et al. Gata2 is a tissue-specific post-mitotic selector gene for midbrain GABAergic neurons. Development 136, 253–262 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Morales, M. & Root, D. H. Glutamate neurons within the midbrain dopamine regions. Neuroscience 282, 60–68 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Waite, M. R., Skidmore, J. M., Billi, A. C., Martin, J. F. & Martin, D. M. GABAergic and glutamatergic identities of developing midbrain Pitx2 neurons. Dev. Dyn. 240, 333–346 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nakatani, T., Minaki, Y., Kumai, M. & Ono, Y. Helt determines GABAergic over glutamatergic neuronal fate by repressing Ngn genes in the developing mesencephalon. Development 134, 2783–2793 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Puelles, E. et al. Otx2 regulates the extent, identity and fate of neuronal progenitor domains in the ventral midbrain. Development 131, 2037–2048 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Moreno-Bravo, J. A., Perez-Balaguer, A., Martinez, S. & Puelles, E. Dynamic expression patterns of Nkx6.1 and Nkx6.2 in the developing mes-diencephalic basal plate. Dev. Dyn. 239, 2094–2101 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Prakash, N. et al. Nkx6-1 controls the identity and fate of red nucleus and oculomotor neurons in the mouse midbrain. Development 136, 2545–2555 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bonilla, S. et al. Identification of midbrain floor plate radial glia-like cells as dopaminergic progenitors. Glia 56, 809–820 (2008).

    Article  PubMed  Google Scholar 

  59. Martinez-Lopez, J. E., Moreno-Bravo, J. A., Madrigal, M. P., Martinez, S. & Puelles, E. Mesencephalic basolateral domain specification is dependent on Sonic Hedgehog. Front. Neuroanat. 9, 12 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Arenas, E., Denham, M. & Villaescusa, J. C. How to make a midbrain dopaminergic neuron. Development 142, 1918–1936 (2015).

    Article  CAS  PubMed  Google Scholar 

  61. Blaess, S. et al. Temporal-spatial changes in Sonic Hedgehog expression and signaling reveal different potentials of ventral mesencephalic progenitors to populate distinct ventral midbrain nuclei. Neural Dev. 6, 29 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Ono, Y. et al. Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells. Development 134, 3213–3225 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Dumas, S. & Wallén-Mackenzie, Å. Developmental co-expression of Vglut2 and Nurr1 in a mes-di-encephalic continuum preceeds dopamine and glutamate neuron specification. Front. Cell Dev. Biol. 7, 307 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Villaescusa, J. C. et al. A PBX1 transcriptional network controls dopaminergic neuron development and is impaired in Parkinson’s disease. EMBO J. 35, 1963–1978 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wang, L., Xiang, B., Liu, H. & Wei, W. LinTInd: lineage tracing by indels. R package version 1.2.0. Bioconductor https://doi.org/10.18129/B9.bioc.LinTInd (2022).

  66. Smith, T., Heger, A. & Sudbery, I. UMI-tools: modeling sequencing errors in unique molecular identifiers to improve quantification accuracy. Genome Res. 27, 491–499 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J. https://doi.org/10.14806/ej.17.1.200 (2011).

  68. Magoč, T. & Salzberg, S. L. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinforma. 27, 2957–2963 (2011).

    Article  Google Scholar 

Download references

Acknowledgements

We thank J. Zhou in Institute of Neuroscience, Z. Yang in Fudan University Shanghai Medical College and B. Zhou in Institute of Biochemistry and Cell Biology for providing us En1-Cre, Nkx2.1-Cre and NR1 mouse lines. We thank M. Poo and S. Yang for intensive discussions on the logic and flow of the manuscript’s content. We thank J. He for comments on the manuscript. We thank D. Xiang, X. Chen and Q. Hu in Optical Imaging facility, Z. Zhou, M. Zhang, H. Hai and L. Quan in Molecular and Cellular Biology Core Facility, M. Zhang and P. Bao in Neural Stem Cell Core Facility at the Institute of Neuroscience. This research was supported in part by STI2030-Major Projects (grant no. 2021ZD0200900 to Y.C.), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDB32030200 to Y.C.), the Shanghai Municipal Science and Technology Major Project (grant nos. 2018SHZDZX05 to Y.C. and 2018SHZDZX01 to M.X.), the National Natural Science Foundation of China (grant nos. 32170806 to Y.C.; 82222021, 32270849 and 81974174 to M.X. and 81870187 to W.W.), Program of Shanghai Academic/Technology Research leader (grant no. 22XD1420800 to M.X.) and UniXell R&D Innovation Cooperation Program (grant no. DA001-RD202101 to Y.C.).

Author information

Authors and Affiliations

Authors

Contributions

Y.C. conceived the project. Y.C. and W.W. supervised the project. Y.C., Z.Y. and L.X. designed CREST mouse lines. L.X. performed bulk experiments, scRNA-seq experiments, library construction and ex vivo culture. L.X. and X.Z. performed transcriptome analysis. H.L., L.W. and L.X. performed lineage analysis. L.X., X.Z., Z.W. and W.Z. performed plasmid construction. Y.L., X.J., H.H., M.X., L.X. and T.Y. performed immunohistochemistry and imaging. L.X., Y.C., H.L. and W.W. wrote the manuscript.

Corresponding authors

Correspondence to Wu Wei or Yuejun Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks James Gagnon and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Madhura Mukhopadhyay, in collaboration with the Nature Methods team. 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 Generation of CERST mouse lines.

a, Gene targeting of the TIGRE locus. The gRNA cassette and the V1 recorder cassette were targeted into the TIGRE locus of mouse embryonic stem cells (mESCs). The upper diagram shows the wild-type TIGRE locus, the lower shows the donor plasmid used for gene targeting. The probes used for southern blot are shown. Neo, neomycin resistance gene driven by PGK promoter; Frt, recognition site of Flp recombinase; DTA, diphtheria toxin A. b, Southern blot of selected mESC clones. The probes used are indicated in Extended Data Fig. 1a. Successful targeting of clone 3-H03 was confirmed using restriction fragments of different sizes: 19.7 kb for the 5′ probe, 6.3 kb for the 3′ probe, and 11.7 kb for the Neo probe. c, BFP expression was observed in the 3-H03 mESC clone, which was used to generate the TIGRECREST-gRNAs mouse line. d, Gene targeting of Rosa26 locus. The Cas9 cassette and the V2 recorder cassette were targeted into the Rosa26 locus of mouse zygotes. The upper diagram shows the wild-type Rosa26 locus, and the lower shows the donor plasmid used for gene targeting. e, Successful targeting of Rosa26CREST-Cas9 mouse was confirmed through genotyping using the primers indicated in Extended Data Fig. 1d. f, Retrieval of CREST barcodes for individual cells. CREST barcodes are enriched from cDNA libraries of single cells, classified as V1 or V2, and assigned to individual cells by 10X cell barcodes for clonal analysis (see Methods).

Source data

Extended Data Fig. 2 Validation of CERST in vivo.

a, Immunohistochemistry images at E12.5 and P0 showing that En1-driven Cre recombinase activates EGFP expression, which indicates Cas9 expression in the midbrain and anterior hindbrain. The boxed areas are magnified. Scale bars, 200 µm. b, FACS results of En1-Cre-CREST embryos between E7.5–E8.5. c, d, Edited array proportion (c) and CREST potential (d) in targeted (EGFP+) and non-targeted (EGFP) cells at E8.5. CREST libraries from mice without Cas9 or without sgRNA were used as control for V1 or V2, respectively. Library preparation error is limited (V1, 0.20%; V2, 0.38%). e, InDels (insertions and deletions) of varying lengths were observed in both recorder arrays. The occurrence is calculated as [the number of InDel types with the indicated length in V1 or V2] / [total number of InDel types of V1 or V2]. f, g, The 20 most frequently observed CREST barcodes in V1 (f) and V2 (g) recorders in each dataset before blacklist filtering. Numerical postfixes represent biological replicates; alphabetical postfixes represent technical replicates. Heatmaps (right) show the proportion of the indicated CREST barcode in each sample. h, Edit type frequency at each target site of V1 and V2 among all En1-Cre-CREST bulk samples. i, Frequencies of InDel types spanning different numbers of target sites in all En1-Cre-CREST bulk samples. Data are presented as mean +/− SEM. j, Length (bp) of V1 and V2 barcodes among all En1-Cre-CREST bulk samples. The lower and upper hinges are the first and third quartiles, the central line is the median, and the whiskers extend to ±1.5* IQR. For panels i and j, statistical analyses were performed using the two-sided Student’s t test and p values were shown. k, l, Quantification of proportions of CREST barcodes (k) or InDels (l) with different frequencies in eight biological samples (E9.5–P0). m, Venn diagram showing shared V1 or V2 barcodes and their corresponding cell (UMI) counts in three Nkx2.1-Cre-CREST replicates. 167,763 and 503,571 cells were analyzed for V1 and V2, respectively.

Source data

Extended Data Fig. 3 Marker genes, pseudo-time ranks, and validations of cell types in E11.5 and E15.5 vMB.

a, b, Representative FACS gating strategies for En1-Cre-CREST vMB at E11.5 and E15.5. Cells in Q2 (EGFP+/BFP+) were sorted and collected for scRNA-seq. c, d, Violin plots showing selected marker genes for each cell cluster in E11.5 (c) and E15.5 (d) datasets. Mean expression levels (color shade) in groups are shown. All marker genes used to define cell types are listed in Supplementary Tables 4 and 5. e, Pseudo-time ranks (calculated by Monocle 2) of individual cells are visualized on the merged UMAP space. f, Quantification of mean pseudo-time ranks in each cluster. Earlier state and later state cell types are defined manually based on pseudo-time ranks. Early progenitor types at E11.5 are highlighted in blue. The late neuronal types of E15.5 are highlighted in red. The lower and upper hinges are the first and third quartiles, the central line is the median, and the whiskers extend to ±1.5* IQR (inter-quartile range). Cell number for each cell type is shown in the figure. g, h, In situ hybridization of coronal sections of mice brains E11.5 (g) and E15.5 (h), showing expression of marker genes of different domains and GLU neurons in the midbrain. Data are collected from the Allen Brain Atlas. Scale bar, 100 µm. i, In situ hybridization of sagittal sections of E15.5 mouse brains, showing the expression of marker genes in the mid-hindbrain boundary. Data are collected from Allen Brain Atlas. Scale bar, left 100 µm.

Source data

Extended Data Fig. 4 Cell type comparisons across different samples.

a, Comparison of cell compositions in datasets from this study versus previously published datasets33. Heatmaps showing the distribution of the densities of cells sampled at E11.5 in this study (upper left), at E15.5 in this study (lower left), at E11.5 from the published literature (upper right), and at E15.5 from published literature (lower right) mapped in the UMAP space. b, Cell types in panel a are indicated by canonical markers of neural progenitor (Sox2), neuroblast (Neurod1), and GABAergic neuroblast (Otx2+/Gata3+). c, Co-staining of Nkx6-1 and Nkx6-2 using in situ hybridization (RNAscope) in E11.5 vMB. Representative regions are magnified to show NPBL (Nkx6-1+/Nkx6-2+) and NPBM (Nkx6-1+/Nkx6-2) cells. BL, basal lateral; BM, basal medial; FP, floor plate. Scale bar, left 50 µm, right 10 µm. d, Hierarchical clustering of pairwise correlations between cell types in of E11.5 and E15.5 datasets based on transcriptomic similarities. Neuroblasts captured at both timepoints are highlighted in red. Cell type names are prefixed with ‘E11’ or ‘E15’ to indicate their sources from the E11.5 or E15.5 vMB datasets, respectively. e, Comparison of cell type compositions between En1-Cre-CREST and normally developed (En1-Cre/Ai9) mouse vMB. Two-sided paired t-test was used to examine the correlations between two groups. f, Gene expression correlation of each cell type between En1-Cre-CREST and normally developed (En1-Cre/Ai9) vMB datasets. Each dot represents a gene.

Source data

Extended Data Fig. 5 Additional details of lineage correlation analyses.

a, Workflow of single-cell lineage analysis adapted to the CREST system. b, Proportions of cell barcode overlap across three CREST barcode amplicons for each scRNA-seq sample. For E11.5 (n = 6), 82.7 ± 2.3% and 72.8 ± 2.7% of cell barcodes were shared across three amplicons for V1 and V2, respectively; the numbers were 78.3 ± 0.9% and 81.5 ± 3.6% for E15.5 (n = 3); 76.4 ± 0.5% and 80.3 ± 0.9% for organoids (n = 3). c, Barcode recovery among samples. 77.3 ± 1.2% E11.5 cells (45.7 ± 8.1% for V1, 74.1 ± 0.9% for V2, and 42.5 ± 8.0% for both V1 and V2; n = 6), 70.9 ± 3.4% E15.5 cells (32.0 ± 4.5% for V1, 69.3 ± 3.6% for V2, and 30.4 ± 4.6% for both V1 and V2; n = 3), and 56.6 ± 4.1% organoid cells (31.2 ± 9.8% for V1, 52.5 ± 5.5% for V2, and 27.1 ± 11.2% for both V1 and V2; n = 3) were recovered with at least one CERST barcodes. d, e, Bar plots showing the proportions of cells recovered with only V1 barcodes, only V2 barcodes, both V1 and V2 barcodes, or no CREST barcode in each cell type in six E11.5 (d) and three E15.5 (e) datasets. f, Pairwise lineage correlation similarities among replicates. Each dot represents the lineage correlation (calculated as the absolute Pearson correlation based on V1 + V2) of a pair of cell types. g, Summary of the correlation coefficient in panel f. The p values were calculated using the two-sided ANOVA test. h, Pairwise lineage correlation similarities across lineage relationships obtained based on V1 + V2 CREST barcodes, V1 CREST barcodes, and V2 CREST barcodes (see Methods). Each dot represents the pairwise correlation calculated from the combined dataset of all three replicates for each group of experiments. i, Summary of the correlation coefficient in panel h. In panels b, c, g, data are presented as mean +/− SEM. In panels f and h, the p values were calculated using the two-sided Pearson’s correlation test.

Source data

Extended Data Fig. 6 Benchmarking, lineage validation, and details of clonal properties in vivo.

a, Workflow for benchmarking CRISPR barcoding lineage analysis methods. Phylogenic trees were first divided into several given numbers (leaf resolution) of subtrees (upper). These subtrees were then taken as leaves (that is, clone groups) to calculate the pairwise lineage correlations. The correlations among the pairwise lineages of three replicates were calculated for each leaf resolution. b, Benchmarking results of different lineage reconstruction methods, including two top scoring phylogenic tree reconstruction methods in the DREAM challenge (Cassiopeia and DCLEAR)36, one traditional phylogenic tree reconstruction method based on Hamming distance and the clonal analysis method. The consistency of lineage results across replicates peaks at single leaf resolution (that is, clonal analysis). For each method at each resolution, three snapCREST E15.5 replicate datasets were used (n = 3 for each box). c, Schematics of fate mapping of floor plate (Lmx1a+) cells. Lmx1a-iCreER mice were crossed with Ai9 (Rosa26-LSL-tdT) reporter mice. Tamoxifen was administrated at E10.5. Embryos were collected at E15.5. d, Fate mapping results of floor plate cells. Representative images are magnified to show the floor plate-derived DA neurons and GLU neurons (GLUFP). Scale bars, 100 µm (upper left) and 10 µm (upper right and lower right). e, Boxplots showing the size distributions of all clones in the E11.5 and E15.5 datasets. f, g, Boxplots showing the size distributions of all clones in each domain. Clones in the E11.5 (f) and E15.5 (g) datasets are classified and grouped based on the dominant regional identities of the clonal cells. h-m, Statistical analysis of InDel number vs. regional identity at E11.5 (h) and E15.5 (i), vs. proliferative potential at E11.5 (j) and E15.5 (k), and vs. progenitor type at E11.5 (l) and E15.5 (m). Statistics: two-sided Student’s t for each indicated pair in panels e-g, l-m; two-sided ANOVA test for panels h-i; two-sided Mann–Kendall trend test for panels f-g, j-k. For each boxplot in this figure, the lower and upper hinges are the first and third quartiles, the central line is the median, and the whiskers extend to ±1.5* IQR.

Source data

Extended Data Fig. 7 Additional data of regional analyses and clustering of clones at E11.5.

a, Visualization of dispersion behaviors of clones along the dorsal-ventral axis in the E11.5 dataset, where circles, sectors, and boxes are colored by regional identities as described in Fig. 4b. b, Distribution of dispersion scores in E11.5 clones. c, Relationships between dispersion score and maximum percentage (calculated as [number of cells with the most dominant regional identity in each clone] / [total number of cells in each clone (referred to as clone size)]); between clone size and cell type complexity (calculated as the Shannon-Wiener Index based on the clonal cell type compositions); and between clone size and dispersion score of clones in E11.5 (upper) and E15.5 (lower) datasets. d, e, Clustering of pairwise lineage correlations as determined by CREST barcodes shared between midbrain domains in the E11.5 dataset (d) and the E15.5 dataset (e). Cell types with a same regional identity in each dataset (as described in Fig. 2f–j) were merged into a domain cluster. For example, NbFP and Rgl1 in the E11.5 dataset were merged as the E11.5 floor plate (FP) cluster. Cell types with undefined regional identities were discarded. f, Quantification of the number of clonal cell types in clones from the E11.5 (left) and E15.5 (right) datasets. Multipotent progenitors (producing at least two distinct cell types) were predominant, giving rise to 85.8% and 88.8% of individual clones at E11.5 and E15.5, respectively. g, Clones in the E11.5 dataset are clustered based on the cell type composition matrix. h, Visualization of E11.5 clone clusters mapped on the UMAP space. The UMAP reference is colored based on spatial identities.

Source data

Extended Data Fig. 8 Additional data for analysis and validation of floor plate lineages.

a, Visualization of fate-biased NbFP cells on the UMAP space. b, Loadings of principal component 1 (PC1) and principal component 2 (PC2). c, Design of sgRNAs targeting Pbx1 and primers for genotyping, also see Methods. d, Representative TA cloning results of Pbx1-KO embryos. Each panel shows an allele of the Pbx1 gene. The upper sequence in each panel shows the sequence of exon I of Pbx1 in a wild-type mouse. Embryos with both alleles being out-of-frame mutated were used in Pbx1-KO group. e, f, Statistical analysis of total DA cell (TH+) counts and total GLU cell (THVglut2+) counts in control (n = 6) and Pbx1-KO (n = 6) embryos. E14.5 mouse embryos were analyzed. For each boxplot, p values were calculated using two-sided Student’s t test. Lower and upper hinges are the first and third quartiles, the central line is the median, and the whiskers extend to the minima and maxima.

Source data

Extended Data Fig. 9 Transcriptional similarities between sister subclones split from the E11.5 dataset and between cell types in the organoid and E15.5 vMB.

a, b, Cells from split clone pairs (that is, sister subclones) are transcriptionally similar. The replicate 1 of snapCREST-E11.5 dataset was randomly divided for analyses. Variations in integrated clonal transcriptomes (a) and integrated clonal progenitor transcriptomes (b) between sister subclones were visualized and analyzed using: 1) UMAP clustering of integrated transcriptomes; 2) typical examples of sister subclones/progenitors pairs, colored by the clone cluster defined in Extended Data Fig. 7g, and sister subclones/progenitors are linked by colored lines; 3) distributions of UMAP distances between sister and random subclone pairs; and 4) quantification of pairwise Pearson’s correlations of integrated transcriptomes between random, nearest neighbors in UMAP spaces, and sister subclone pairs. See also Methods. For each boxplot, the lower and upper hinges are the first and third quartiles, the central line is the median, and the whiskers extend to ±1.5* IQR; p values were calculated using two-sided Student’s t test and were shown in the figure. c, d, Sister subclones (c) and sister progenitors (d) prefer to localize in the same or nearby transcriptome-based clusters. Left, cumulative probability that subclone pairs will lie within the same cluster or across N nearest clusters of panel a; right, heatmaps showing the probability that a sister subclone pair lies in which two clusters of panel a. See also Methods. e, Heatmaps showing cell distribution of the densities in the organoid and E15.5 datasets on the integrated UMAP space. f, Hierarchical clustering of pairwise correlations between cell types in E15.5 and organoid datasets based on transcriptomes. The names of cell types are prefixed with ‘E15’ or ‘OGN’ to indicate their sources. g, Cell types in (e) are indicated by canonical markers of Rgl (Slc1a3), DA (Pitx3), GABA (Gad1), GLU (Slc17a6), BM (Pitx2), AL/BM (Pou4f1), AL (Lhx2), and MHB (Pax5). h, Gene expression correlation of each cell type between ex vivo organoids and in vivo E15.5 vMB datasets.

Source data

Extended Data Fig. 10 Lineage similarities between in vivo and ex vivo datasets and fate maps for E11.5 progenitors.

a, PCA analysis of clonal cell-type compositions for in vivo and ex vivo clones. b, Pairwise lineage correlation (calculated as absolute Pearson correlation) of cell types captured both in vivo and ex vivo. The upper right shows the lineage relationship in vivo. The lower left shows the lineage relationship ex vivo. c, Examples of the fate of individual NPBM-restricted clones in Fig. 6f are shown on UMAP spaces of the E11.5 and OGN datasets. d, Fate maps for individual NPBL-restricted clones. An example of each fate is shown on the UMAP spaces of the E11.5 and OGN datasets.

Source data

Supplementary information

Supplementary Information

Supplementary Table 1, Discussion and references.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Tables 2–8.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Extended Data Fig. 1

Unprocessed Southern blots and gels for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

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

Xie, L., Liu, H., You, Z. et al. Comprehensive spatiotemporal mapping of single-cell lineages in developing mouse brain by CRISPR-based barcoding. Nat Methods 20, 1244–1255 (2023). https://doi.org/10.1038/s41592-023-01947-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-01947-3

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