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:

Population snapshots predict early haematopoietic and erythroid hierarchies

Abstract

The formation of red blood cells begins with the differentiation of multipotent haematopoietic progenitors. Reconstructing the steps of this differentiation represents a general challenge in stem-cell biology. Here we used single-cell transcriptomics, fate assays and a theory that allows the prediction of cell fates from population snapshots to demonstrate that mouse haematopoietic progenitors differentiate through a continuous, hierarchical structure into seven blood lineages. We uncovered coupling between the erythroid and the basophil or mast cell fates, a global haematopoietic response to erythroid stress and novel growth factor receptors that regulate erythropoiesis. We defined a flow cytometry sorting strategy to purify early stages of erythroid differentiation, completely isolating classically defined burst-forming and colony-forming progenitors. We also found that the cell cycle is progressively remodelled during erythroid development and during a sharp transcriptional switch that ends the colony-forming progenitor stage and activates terminal differentiation. Our work showcases the utility of linking transcriptomic data to predictive fate models, and provides insights into lineage development in vivo.

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

Figure 1: The early haematopoietic hierarchy predicted by scRNA-seq.
Figure 2: A novel sorting scheme isolates erythroid progenitors.
Figure 3: Predicted fate couplings confirmed by single-cell fate assays.
Figure 4: Stages of early erythropoiesis and the global erythroid stress response.
Figure 5: Novel growth factor regulators of early erythropoiesis.
Figure 6: Extensive remodelling of the cell cycle during erythroid development.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Fujiwara, Y., Browne, C. P., Cunniff, K., Goff, S. C. & Orkin, S. H. Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1. Proc. Natl Acad. Sci. USA 93, 12355–12358 (1996)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Liu, Y. et al. Suppression of Fas–FasL coexpression by erythropoietin mediates erythroblast expansion during the erythropoietic stress response in vivo. Blood 108, 123–133 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chen, K. et al. Resolving the distinct stages in erythroid differentiation based on dynamic changes in membrane protein expression during erythropoiesis. Proc. Natl Acad. Sci. USA 106, 17413–17418 (2009)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  4. Hara, H. & Ogawa, M. Erythropoietic precursors in mice under erythropoietic stimulation and suppression. Exp. Hematol. 5, 141–148 (1977)

    CAS  PubMed  Google Scholar 

  5. Gregory, C. J., McCulloch, E. A. & Till, J. E. The cellular basis for the defect in haemopoiesis in flexed-tailed mice. III. Restriction of the defect to erythropoietic progenitors capable of transient colony formation in vivo. Br. J. Haematol. 30, 401–410 (1975)

    Article  CAS  PubMed  Google Scholar 

  6. Pronk, C. J. et al. Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy. Cell Stem Cell 1, 428–442 (2007)

    Article  CAS  PubMed  Google Scholar 

  7. Flygare, J., Rayon Estrada, V., Shin, C., Gupta, S. & Lodish, H. F. HIF1α synergizes with glucocorticoids to promote BFU-E progenitor self-renewal. Blood 117, 3435–3444 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Li, J. et al. Isolation and transcriptome analyses of human erythroid progenitors: BFU-E and CFU-E. Blood 124, 3636–3645 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mori, Y., Chen, J. Y., Pluvinage, J. V., Seita, J. & Weissman, I. L. Prospective isolation of human erythroid lineage-committed progenitors. Proc. Natl Acad. Sci. USA 112, 9638–9643 (2015)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Guo, G. et al. Mapping cellular hierarchy by single-cell analysis of the cell surface repertoire. Cell Stem Cell 13, 492–505 (2013)

    Article  CAS  PubMed  Google Scholar 

  11. Sun, J. et al. Clonal dynamics of native haematopoiesis. Nature 514, 322–327 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015)

    Article  CAS  PubMed  Google Scholar 

  13. Busch, K. et al. Fundamental properties of unperturbed haematopoiesis from stem cells in vivo. Nature 518, 542–546 (2015)

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Notta, F. et al. Distinct routes of lineage development reshape the human blood hierarchy across ontogeny. Science 351, aab2116 (2016)

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Nestorowa, S. et al. A single-cell resolution map of mouse hematopoietic stem and progenitor cell differentiation. Blood 128, e20–e31 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Velten, L. et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 19, 271–281 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mercier, F. E. & Scadden, D. T. Not all created equal: lineage hard-wiring in the production of blood. Cell 163, 1568–1570 (2015)

    Article  CAS  PubMed  Google Scholar 

  18. Kondo, M., Weissman, I. L. & Akashi, K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91, 661–672 (1997)

    Article  CAS  PubMed  Google Scholar 

  19. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I. L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000)

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Adolfsson, J. et al. Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 121, 295–306 (2005)

    Article  CAS  PubMed  Google Scholar 

  21. Huang, W., Cao, X., Biase, F. H., Yu, P. & Zhong, S. Time-variant clustering model for understanding cell fate decisions. Proc. Natl Acad. Sci. USA 111, E4797–E4806 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Marco, E. et al. Bifurcation analysis of single-cell gene expression data reveals epigenetic landscape. Proc. Natl Acad. Sci. USA 111, E5643–E5650 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shin, J. et al. Single-cell RNA-seq with waterfall reveals molecular cascades underlying adult neurogenesis. Cell Stem Cell 17, 360–372 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ji, Z. & Ji, H. TSCAN: pseudo-time reconstruction and evaluation in single-cell RNA-seq analysis. Nucleic Acids Res. 44, e117 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Welch, J. D., Hartemink, A. J. & Prins, J. F. SLICER: inferring branched, nonlinear cellular trajectories from single cell RNA-seq data. Genome Biol. 17, 106 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Haghverdi, L., Buttner, 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 

  27. Moignard, V. et al. Decoding the regulatory network of early blood development from single-cell gene expression measurements. Nat. Biotechnol. 33, 269–276 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Khoramian Tusi, B. & Socolovsky, M. High throughput single-cell fate potential assay of murine hematopoietic progenitors in vitro. Ex. Hematol. https://doi.org/10.1016/j.exphem.2018.01.005 (2018)

  29. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Morrison, S. J. & Weissman, I. L. The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype. Immunity 1, 661–673 (1994)

    Article  CAS  PubMed  Google Scholar 

  31. Papayannopoulou, T., Brice, M., Broudy, V. C. & Zsebo, K. M. Isolation of c-kit receptor-expressing cells from bone marrow, peripheral blood, and fetal liver: functional properties and composite antigenic profile. Blood 78, 1403–1412 (1991)

    Article  CAS  PubMed  Google Scholar 

  32. Weinreb, C ., Wolock, S. & Klein, A. SPRING: a kinetic interface for visualizing high dimensional single-cell expression data. Bioinformatics (2017)

  33. Weinreb, C ., Wolock, S ., Khoramian Tusi, B ., Socolovsky, M. & Klein, A. M. Fundamental limits on dynamic inference from single cell snapshots. Proc. Natl Acad. Sci. USA. https://doi.org/10.1073/pnas.1714723115 (2018)

  34. Yanez, A. et al. Granulocyte-monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes. Immunity 47, 890–902.e4 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Magwene, P. M., Lizardi, P. & Kim, J. Reconstructing the temporal ordering of biological samples using microarray data. Bioinformatics 19, 842–850 (2003)

    Article  CAS  PubMed  Google Scholar 

  36. Bendall, S. C. et al. Single-cell trajectory detection uncovers progression and regulatory coordination in human B cell development. Cell 157, 714–725 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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 

  38. Bresnick, E. H., Lee, H.-Y., Fujiwara, T., Johnson, K. D. & Keles, S. GATA switches as developmental drivers. J. Biol. Chem. 285, 31087–31093 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, P. et al. Regulation of bone marrow hematopoietic stem cell is involved in high-altitude erythrocytosis. Exp. Hematol. 39, 37–46 (2011)

    Article  CAS  PubMed  Google Scholar 

  40. Grover, A. et al. Erythropoietin guides multipotent hematopoietic progenitor cells toward an erythroid fate. J. Exp. Med. 211, 181–188 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mancini, E. et al. FOG-1 and GATA-1 act sequentially to specify definitive megakaryocytic and erythroid progenitors. EMBO J. 31, 351–365 (2012)

    Article  CAS  PubMed  Google Scholar 

  42. Koulnis, M ., Porpiglia, E ., Hidalgo, D. & Socolovsky, M. in A Systems Biology Approach to Blood, Vol. 844 (eds Corey, S. J. et al.) Ch. 3, 37–58 (Springer New York, 2014)

    Google Scholar 

  43. Agosti, V., Karur, V., Sathyanarayana, P., Besmer, P. & Wojchowski, D. M. A KIT juxtamembrane PY567-directed pathway provides nonredundant signals for erythroid progenitor cell development and stress erythropoiesis. Exp. Hematol. 37, 159–171 (2009)

    Article  CAS  PubMed  Google Scholar 

  44. Koury, M. J. & Bondurant, M. C. Erythropoietin retards DNA breakdown and prevents programmed death in erythroid progenitor cells. Science 248, 378–381 (1990)

    Article  ADS  CAS  PubMed  Google Scholar 

  45. Yee, K., Bishop, T. R., Mather, C. & Zon, L. I. Isolation of a novel receptor tyrosine kinase cDNA expressed by developing erythroid progenitors. Blood 82, 1335–1343 (1993)

    Article  CAS  PubMed  Google Scholar 

  46. van den Akker, E. et al. Tyrosine kinase receptor RON functions downstream of the erythropoietin receptor to induce expansion of erythroid progenitors. Blood 103, 4457–4465 (2004)

    Article  CAS  PubMed  Google Scholar 

  47. Pop, R. et al. A key commitment step in erythropoiesis is synchronized with the cell cycle clock through mutual inhibition between PU.1 and S-phase progression. PLoS Biol. 8, e1000484 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hwang, Y. et al. Global increase in replication fork speed during a p57KIP2-regulated erythroid cell fate switch. Sci. Adv. 3, e1700298 (2017)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  49. Shearstone, J. R. et al. Global DNA demethylation during mouse erythropoiesis in vivo. Science 334, 799–802 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Nemeth, E. & Ganz, T. Anemia of inflammation. Hematol. Oncol. Clin. North Am. 28, 671–681 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  51. Liang, R. et al. A systems approach identifies essential FOXO3 functions at key steps of terminal erythropoiesis. PLoS Genet. 11, e1005526 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Whitfield, M. L. et al. Identification of genes periodically expressed in the human cell cycle and their expression in tumors. Mol. Biol. Cell 13, 1977–2000 (2002)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zilionis, R. et al. Single-cell barcoding and sequencing using droplet microfluidics. Nat. Protocols 12, 44–73 (2017)

    Article  CAS  PubMed  Google Scholar 

  54. Ester, M ., Kriegel, H ., Sander, J. & Xu, X. A density-based algorithm for discovering clusters in large spatial databases with noise. In Proc. 2nd International Conference on Knowledge Discovery and Data Mining (Eds Simoudis, E . et al.) 226–231 (AAAI, 1996)

  55. Daszykowski, M., Walczak, B. & Massart, D. L. Looking for natural patterns in data: Part 1. Density-based approach. Chemomtr. Intell. Lab. Syst. 56, 83–92 (2001)

    Article  CAS  Google Scholar 

  56. van der Maaten, L. Accelerating t-SNE using tree-based algorithms. J. Mach. Learn. Res. 15, 3221–3245 (2014)

    MathSciNet  MATH  Google Scholar 

  57. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Weinreb, C., Wolock, S. & Klein, A. SPRING: a kinetic interface for visualizing high dimensional single-cell expression data. Bioinformatics https://doi.org/10.1093/bioinformatics/btx792 (2017)

  59. Vandin, F., Upfal, E. & Raphael, B. J. Algorithms for detecting significantly mutated pathways in cancer. J. Comput. Biol. 18, 507–522 (2011)

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  60. Heng, T. S. et al. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008)

    Article  CAS  PubMed  Google Scholar 

  61. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lachmann, A. et al. ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments. Bioinformatics 26, 2438–2444 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Scialdone, A. et al. Computational assignment of cell-cycle stage from single-cell transcriptome data. Methods 85, 54–61 (2015)

    Article  CAS  PubMed  Google Scholar 

  64. Santos, A., Wernersson, R. & Jensen, L. J. Cyclebase 3.0: a multi-organism database on cell-cycle regulation and phenotypes. Nucleic Acids Res. 43, D1140–D1144 (2015)

    Article  CAS  PubMed  Google Scholar 

  65. Shekhar, K. et al. Comprehensive classification of retinal bipolar neurons by single-cell transcriptomics. Cell 166, 1308–1323.e1330 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995)

    MathSciNet  MATH  Google Scholar 

  67. Tusi, B. K. & Socolovsky, M. Novel FACS strategy for identification of early hematopoietic progenitors including BFU-e, CFU-e and erythroid-biased MPPs Protoc. Exch. https://doi.org/10.1038/protex.2018.031 (2018)

  68. El Malki, K. et al. An alternative pathway of imiquimod-induced psoriasis-like skin inflammation in the absence of interleukin-17 receptor a signaling. J. Invest. Dermatol. 133, 441–451 (2013)

    Article  CAS  PubMed  Google Scholar 

  69. Porpiglia, E., Hidalgo, D., Koulnis, M., Tzafriri, A. R. & Socolovsky, M. Stat5 signaling specifies basal versus stress erythropoietic responses through distinct binary and graded dynamic modalities. PLoS Biol. 10, e1001383 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was funded by a Leukemia and Lymphoma Society Scholar award (1728-13) and R01DK100915 and R01099281 (M.S.). A.M.K. is supported by a BW Fund CASI award and an Edward J Mallinckrodt Foundation Grant. S.L.W. and C.W. are supported by National Institutes of Health (NIH) training grant 5T32GM080177-07.

Author information

Authors and Affiliations

Authors

Contributions

M.S. and A.M.K. designed the experiments and supervised the project. B.K.T., S.L.W., Y.H., D.H. and R.Z. performed experiments including inDrops (B.K.T., R.Z., S.L.W.), FACS and antibody screening (B.K.T., D.H.), single-cell fate assays and cell cycle analysis (B.K.T.), western blotting (Y.H.), qRT–PCR (B.T.K.), pSTAT3/5 (Y.H., D.H.) and colony assays for novel growth factors (Y.H.). S.L.W. and C.W. performed single-cell data analysis, informatics and PBA modelling. A.W. and J.R.H. provided Il17ra−/− mice. B.K.T., S.L.W., C.W., Y.H., D.H., A.M.K. and M.S. prepared figures and wrote the manuscript.

Corresponding authors

Correspondence to Allon M. Klein or Merav Socolovsky.

Ethics declarations

Competing interests

A.M.K. is a co-founder of 1Cell-Bio.

Additional information

Reviewer Information Nature thanks B. Göttgens, F. Hamey and the other anonymous reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Figure 1 scRNA-seq of Kit+ haematopoietic progenitors for prediction of the early haematopoietic hierarchy.

a, Top, SPRING plot of 7,959 human peripheral blood mononuclear cells (PBMCs) from 10X Genomics (https://support.10xgenomics.com/single-cell-gene-expression/datasets/2.0.1/pbmc8k). Clusters were generated by performing spectral clustering on the underlying kNN graph and annotated on the basis of marker genes. NK, natural killer. Random walks over kNN graphs for the PBMC (middle) and Kit+ bone marrow (bottom) datasets. Each plot shows the fraction of nodes (cells) visited for 1,000 simulated random walks. b, Top, SPRING plot of 2,855 LinKit+SCA1 mouse HPCs from a previously published dataset12. Bottom, SPRING plot of 1,656 cells from three mouse haematopoietic progenitor populations (LinKit+Sca1, LinKit+SCA1+, and LinKit+SCA1+FLK2CD34+) from a previously published dataset15. Coloured (non-grey) cells indicate expression of lineage-specific genes (see Supplementary Table 7). c, Schematic of the population balance law, which relates the dynamic velocities of cells to the distribution of states they are in at a moment in time. The law states that in steady state, after accounting for cell division and loss, the flux of cells entering any region of gene expression space equals the flux out of that region. d, Flow diagram of the inputs and outputs of the PBA algorithm. The population balance law is applied to inputs that include single-cell expression data and estimates of cell proliferation and loss rates at each point in gene expression space; inferred outputs include cell dynamics such as fate probabilities and pseudo-temporal ordering. e, SPRING plot of bone marrow Kit+ cells (Fig. 1) constructed using only the PBA-predicted fate probabilities and differentiation ordering as inputs (n = 4,763 cells from one inDrops experiment). Coloured cells indicate expression of lineage-specific genes as in Fig. 1b. f, SPRING plot of bone marrow Kit+ cells (Fig. 1), with cells coloured by library preparation batch.

Extended Data Figure 2 Predicting key regulators at haematopoietic choice points.

Candidate regulators of fate choice, identified by ranking transcription factors and transmembrane receptors by their correlation with PBA-predicted fate probabilities at key choice points in haematopoiesis. Top-ranked genes are shown; these include many canonical regulators. Candidate genes that have not been previously reported are marked with asterisks. Several candidates participate in more than one fate choice. Insets show SPRING plots coloured by expression of representative genes. Fog1 is also known as Zfpm1, B-myb is also known as Mybl2, PU.1 is also known as Spi1 and Oct2 is also known as Slc22a2.

Source data

Extended Data Figure 3 Mapping HPC subsets P1–P5 to the Kit+ SPRING plot using qRT–PCR and scRNA-seq.

a, Subpopulations P1–P5 map onto specific regions of the SPRING plot. SPRING plot heat maps for a panel of marker genes (left) and corresponding measured expression for each of the marker genes by qRT–PCR (middle), performed on sorted cell subsets P1–P5, and on EryA (cells undergoing ETD2). Probable mapping of each of the P1–P5 subpopulations on the basis of qRT–PCR is shown on the SPRING plot (right). Bars represent the mean of two independent experiments (circles, triangles or squares). Expression is shown normalized to Actb mRNA. b, c, SPRING plot of single-cell transcriptomes from freshly sorted P1–P5 subsets (Fig. 2a, b). Cells are coloured on the basis of sorted subpopulation (b) or the expression of lineage-specific marker genes (c) (Supplementary Table 7). d, e, SPRING plots of P1–P5 subpopulation cells, coloured on the basis of expression of basophil (d) and mast cell (e) marker genes. The larger number of cells in the P3 region of the graph resolves a split between the two lineages that was not observable in the original Kit+ dataset.

Source data

Extended Data Figure 4 Validation of PBA predictions.

a, Megakaryocytic colonies from sorted subsets P1–P5 and from Kit+CD55 cells, stained for the megakaryocytic marker acetylcholinesterase. Duplicate cultures are shown; representative of two independent experiments. b, Representative flow cytometry plots to assay fate output of single cells in liquid culture (see Fig. 3, including Fig. 3a for experimental design). Each row corresponds to a single clone, with the left column indicating the source subset (P1–P5, CD55) of the clone and the cell type(s) produced, as inferred from the FACS plots in the remaining columns. These data are representative of 1,158 single-cell clonal cultures, pooled from three independent sorting experiments (complete dataset is shown in Fig. 3b). c, Bulk liquid cultures of freshly sorted P1–P5 subsets and Kit+CD55 cells in the presence of EPO and a mixture of cytokines supporting myeloid progenitors. On the indicated days, cells were labelled with antibodies as indicated and analysed by flow cytometry.

Source data

Extended Data Figure 5 The early erythroid trajectory.

a, qRT–PCR for expression of established erythroid regulators in sorted P1–P5 subsets. Expression of each gene is normalized to Actb. Bars represent the mean of two independent experiments (circles). b, Left, representative western blot of GATA1 expression in sorted P1 (subdivided into CD71medium and CD71high subsets), P2 and EryA (CD71+TER119+FSChigh cells, representative of ETD) cells. 3T3-GATA1, positive control 3T3 cells virally transduced with a GATA1 expression vector; untransduced 3T3 cells were used as negative control. Right, quantification of GATA1 expression (mean) by densitometry. Data points are from two independent western blots. For gel source data, see Supplementary Fig. 1. c, Density of FACS subsets P1–P5 along the erythroid trajectory. Single-cell transcriptomes from each subset were mapped to their most similar counterparts in the Kit+ data (Fig. 2a, b; cell numbers analysed for each subset are indicated in Fig. 2b). Shown here is the fraction of mapped cells after smoothing with a Gaussian kernel. Also included are CD71high P1 cells, constituting cells with the 30% highest CD71 expression in that subset (n = 752 cells post-filter). d, Distribution of CD71 expression in P1 (top) and P2 (bottom) cells immediately after sorting (grey) and after 24 h of in vitro differentiation (lavender). Data are representative of two independent experiments. e, Dynamically varying genes along the MPP-to-erythroid axis were clustered on the basis of their behaviour across three transition points. At each transition, gene expression is increased, decreased or unchanged, giving a total of 27 potential dynamic patterns across all three transitions, shown in red. The number of genes corresponding to each dynamic pattern is noted, and z-score-normalized expression traces for each individual gene are shown in black. Selected clusters were further analysed in Extended Data Fig. 6 and are marked with an asterisk and a representative gene.

Source data

Extended Data Figure 6 Gene set enrichment on dynamic gene clusters in early erythroid differentiation.

ad, Nine key dynamic gene clusters along the MPP-to-erythroid progression (Extended Data Fig. 5e) are analysed further for Gene Ontology terms. Each cluster is identified by its dynamic pattern with a cartoon of nodes and edges. Each node represents a progenitor stage (in order, MPP, EBMP/EEP, CEP and ETD), connected to the next stage by an edge that either goes up (for increased expression) or down (for decreased expression) (see Extended Data Fig. 5e). a, Number and identity of transcription factors (TFs) whose targets are enriched in the dynamic clusters, as predicted by chromatin immunoprecipitation methods (ChIP-X) experiments62. b, Significance of enrichment for signalling pathways and Gene Ontology gene sets in the dynamic gene clusters (hypergeometric test with Benjamini–Hochberg correction for multiple hypothesis testing). Rep., representative. c, d, Enrichment of transcription factor targets. c, Heat map (−log10 of P value of hypergeometric test with Benjamini–Hochberg correction for multiple hypothesis testing) of target gene enrichment for transcription factors (rows) with targets significantly enriched (P < 0.05) in at least one of the nine dynamic clusters (columns, labelled on top) highlighted in Extended Data Fig. 5e. Note that the transcription factor targets shown are based on previous ChIP-X experiments62 and it is possible that unappreciated transcription factor targets occur in early erythropoiesis. d, Gene expression traces over the erythroid trajectory for the transcription factors from c. Rows match those in c.

Source data

Extended Data Figure 7 Quantification of absolute Kit+ cell number in bone marrow after in vivo administration of EPO.

a, b, Eight-week-old female mice were injected with either EPO (100 U per 25 g body weight) or saline (basal), once per day for two days. Bone marrow was collected at 48 h. Viable (trypan blue-negative) cells were counted using a TC20 automated cell counter (BIORAD) and stained for Kit, TER119 and CD71 and lineage markers. Data are from two independent experiments, with five mice analysed individually for each group (basal or EPO) in each experiment. a, Representative flow cytometric analysis of either basal or EPO-stimulated bone marrow, gating on Kit+Lin cells (left) or on proerythroblasts (ProE) and TER119high cells (right; ProE and TER119high cells are sequential stages of ETD). b, Data summary (mean ± s.d.) for all mice (ten in each group). Top, the fraction of all bone marrow cells for each of the flow cytometric gates defined in a. Bottom, the absolute cell count in adult bone marrow for subsets defined in each flow cytometric gate, or for the total number of bone marrow cells. Significant (P < 0.05) P values are shown (two-tailed t-test, unequal variances).

Source data

Extended Data Figure 8 Identification of stage-specific differential gene expression during the erythroid stress response.

a, Identification of genes that are differentially expressed in EEP cells of either EPO-stimulated bone marrow (eBM, left) or fetal liver (FL, right) samples, compared with basal bone marrow (bBM). One single-cell inDrop experiment per condition. P values were calculated using a binomial test for differential expression (see Methods) and measure the significance of the expression difference. The specific enrichment score (see Methods) measures the degree to which the differential expression is specific to this region of interest (EEPs); positive scores correspond to region-specific upregulation, and negative to region-specific downregulation. Selected genes are highlighted. b, The analysis in a applied to the CEP stage. One single-cell inDrop experiment per condition. c, Stage-specific differential gene expression during stress, comparing EPO-stimulated and fetal liver samples. The heat map shows the number of differentially expressed genes at each stage that show similar or different patterns of upregulation and downregulation in fetal liver and EPO-stimulated bone marrow samples. Representative gene traces are shown on the right.

Extended Data Figure 9 Localized gene expression and functional response of the erythroid lineage to stimulation of MST1, RYK and IL-17RA.

a, b, Predicted expression pattern (a) and confirmation by qRT–PCR (b) for Mst1r, Ryk and Il17ra in basal bone marrow. In a, traces show the smoothed scRNA-seq gene expression of cells from the basal bone marrow (bBM), fetal liver (FL) and EPO-stimulated bone marrow (eBM) samples, arranged along the erythroid trajectory. qRT–PCR data represent the mean (bars) of two independent experiments (circles). c, Complete results for CFU-e and BFU-e colony-formation assays in methylcellulose, supporting the data shown in Fig. 5. Curves show colony numbers in the presence of increasing concentrations of either EPO, or EPO with a ligand (MSP, WNT5A or IL-17A). Error bars show s.d. of two independent experiments, with four replicates per experiment. Where appropriate, data were fitted to a dose–response curve with a Hill coefficient of one. d, Western blot analysis shows that IL-17RA expression peaks in EEP P2 cells, dropping in CEP P1 cells and in the granulocytic branch (which contributes most of the CD55 cells), consistent with the SPRING plots in Fig. 5a. The western blot is representative of two independent experiments. For gel source images, see Supplementary Fig. 1.

Source data

Extended Data Figure 10 Cell ordering independently of cell cycle genes, and evidence of an S-phase-dependent CEP-to-ETD transition in bone marrow erythropoiesis.

a, The computational ordering of cells from MPP to ETD is not sensitive to the inclusion or exclusion of annotated cell cycle genes (cell ordering correlation is R = 0.97). b, c, Reproduction of Fig. 6b, c after the exclusion of cell cycle genes shows that the computationally inferred expression dynamics of cell cycle genes during EEP to CEP differentiation are not sensitive to the inclusion or exclusion of annotated cell cycle genes when ordering cells. d–f, Activation of ETD is dependent on the S phase. d, Schematic of experiments shown in e and f that test the link between S-phase progression and the CEP-to-ETD transition. Kit+LinCD71 bone marrow cells were cultured in the presence of EPO for 28 h, with or without the presence of the DNA polymerase inhibitor aphidicolin (Aphi) for the first 10 h. e, Kit+LinCD71 bone marrow cells require the S phase to upregulate CD71, an early event in ETD. Left, CD71high cells fail to appear in the first 10 h if cells are exposed to aphidicolin; they appear as soon as aphidicolin is removed from the medium. Right, cell cycle analysis of the same cells shows that aphidicolin prevented S-phase progression; aphidicolin removal was followed by a full recovery of S-phase progression, with a high fraction of CD71high cells in S phase. Data are representative of three independent experiments. f, Aphidicolin exposure for 10 h delays induction of β-globin (Hbb-b1) by 10 h. Data are representative of two independent experiments. g, CD71 expression (top), cell cycle phase distribution (middle), and intra-S-phase DNA synthesis rate (bottom), for consecutive FACS gates of increasing CD71 in the early stages of erythropoiesis from the fetal liver (left, representative of four independent experiments) and EPO-simulated bone marrow (right, representative of two independent experiments) samples. See Fig. 6e, f for similar analysis of basal bone marrow samples. h, Western blots (top) and quantification by densitometry (bottom) showing an increase in S-phase proteins during progression from EEP (P2) to early CEP (P1-CD71low) and late CEP (P1-CD71high). Control 3T3 cells were either cycling or contact-inhibited (non-cycling), as indicated. Western blots are representative of three independent experiments. For gel source images, see Supplementary Fig. 1.

Source data

Supplementary information

Life Sciences Reporting Summary (PDF 159 kb)

Supplementary Information

This file contains full legends for Supplementary Tables 1-7, gel source data and Supplementary Tables 1, 2 and 7. (PDF 5505 kb)

Supplementary Table 3

This table contains a list of significantly varying genes in the basal BM dataset - see Supplementary Information document for the full description. (XLSX 391 kb)

Supplementary Table 4

This table shows gene set enrichment analysis (GSEA) of dynamic gene clusters - see Supplementary Information document for the full description. (XLSX 230 kb)

Supplementary Table 5

This table shows genes differentially expressed in stress - see Supplementary Information document for the full description. (XLSX 443 kb)

Supplementary Table 6

This table shows genes correlated with progression through the CEP stage - see Supplementary Information document for the full description. (XLSX 265 kb)

Supplementary Data

This zipped file contains the input data files and code for running Population Balance Analysis on the Bone Marrow and Fetal Liver data sets. (ZIP 1146 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tusi, B., Wolock, S., Weinreb, C. et al. Population snapshots predict early haematopoietic and erythroid hierarchies. Nature 555, 54–60 (2018). https://doi.org/10.1038/nature25741

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature25741

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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