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:

Differentiation of transplanted haematopoietic stem cells tracked by single-cell transcriptomic analysis

Abstract

How transplanted haematopoietic stem cells (HSCs) behave soon after they reside in a preconditioned host has not been studied due to technical limitations. Here, using single-cell RNA sequencing, we first obtained the transcriptome-based classifications of 28 haematopoietic cell types. We then applied them in conjunction with functional assays to track the dynamic changes of immunophenotypically purified HSCs in irradiated recipients within the first week after transplantation. Based on our transcriptional classifications, most homed HSCs in bone marrow and spleen became multipotent progenitors and, occasionally, some HSCs gave rise to megakaryocytic–erythroid or myeloid precursors. Parallel in vitro and in vivo functional experiments supported the paradigm of robust differentiation without substantial HSC expansion during the first week. Therefore, this study uncovers the previously inaccessible kinetics and fate choices of transplanted HSCs in myeloablated recipients at early stage, with implications for clinical applications of HSCs and other stem cells.

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

Access options

Buy this article

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

Fig. 1: Single-cell transcriptomes of iHSCs.
Fig. 2: Single-cell transcriptomes of iMPPs and trajectory analysis of haematopoietic cells.
Fig. 3: Cellular composition of engrafted cells following HSC transplantation.
Fig. 4: Cell division of donor-derived cells after transplantation.
Fig. 5: Temporal fate alteration and differentiation tendency of transplanted HSCs.
Fig. 6: Functional assessments for donor-derived cells after transplantation.
Fig. 7: Emergence of lineage precursors at early phase of HSC transplantation.

Similar content being viewed by others

Data availability

The RNA-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE116530. Source data for Figs. 17 and Extended Data Figs. 35, 8 and 9 are provided with the paper. All other data supporting the findings of this study are available from the corresponding authors upon reasonable request.

Code availability

The computational code used in this study can be obtained by request to P.Z. (zhuping@ihcams.ac.cn).

References

  1. Eaves, C. J. Hematopoietic stem cells: concepts, definitions, and the new reality. Blood 125, 2605–2613 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Laurenti, E. & Gottgens, B. From haematopoietic stem cells to complex differentiation landscapes. Nature 553, 418–426 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chabannon, C. et al. Hematopoietic stem cell transplantation in its 60s: a platform for cellular therapies. Sci. Transl Med. 10, eaap9630 (2018).

    Article  PubMed  CAS  Google Scholar 

  4. Srour, E. F. et al. Homing, cell cycle kinetics and fate of transplanted hematopoietic stem cells. Leukemia 15, 1681–1684 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Shen, H. et al. An acute negative bystander effect of gamma-irradiated recipients on transplanted hematopoietic stem cells. Blood 119, 3629–3637 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mendelson, A. & Frenette, P. S. Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat. Med. 20, 833–846 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Morrison, S. J. & Scadden, D. T. The bone marrow niche for haematopoietic stem cells. Nature 505, 327–334 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Forsberg, E. C., Serwold, T., Kogan, S., Weissman, I. L. & Passegue, E. New evidence supporting megakaryocyte-erythrocyte potential of Fflk2/Flt3+ multipotent hematopoietic progenitors. Cell 126, 415–426 (2006).

    Article  CAS  PubMed  Google Scholar 

  9. Shin, J. Y., Hu, W., Naramura, M. & Park, C. Y. High c-Kit expression identifies hematopoietic stem cells with impaired self-renewal and megakaryocytic bias. J. Exp. Med. 211, 217–231 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, L. D. & Wagers, A. J. Dynamic niches in the origination and differentiation of haematopoietic stem cells. Nat. Rev. Mol. Cell Biol. 12, 643–655 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Abkowitz, J. L., Golinelli, D., Harrison, D. E. & Guttorp, P. In vivo kinetics of murine hemopoietic stem cells. Blood 96, 3399–3405 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Bernitz, J. M., Kim, H. S., MacArthur, B., Sieburg, H. & Moore, K. Hematopoietic stem cells count and remember self-renewal divisions. Cell 167, 1296–1309.e10 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Trumpp, A., Essers, M. & Wilson, A. Awakening dormant haematopoietic stem cells. Nat. Rev. Immunol. 10, 201–209 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Drissen, R. et al. Distinct myeloid progenitor-differentiation pathways identified through single-cell RNA sequencing. Nat. Immunol. 17, 666–676 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  17. Gur-Cohen, S. et al. PAR1 signaling regulates the retention and recruitment of EPCR-expressing bone marrow hematopoietic stem cells. Nat. Med. 21, 1307–1317 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Kent, D. G. et al. Prospective isolation and molecular characterization of hematopoietic stem cells with durable self-renewal potential. Blood 113, 6342–6350 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Yang, L. et al. Identification of LinSca1+kit+CD34+Flt3 short-term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients. Blood 105, 2717–2723 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Yamamoto, R. et al. Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells. Cell 154, 1112–1126 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. 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 

  23. 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  CAS  PubMed  Google Scholar 

  24. 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 

  25. Chambers, S. M. et al. Hematopoietic fingerprints: an expression database of stem cells and their progeny. Cell Stem Cell 1, 578–591 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. Venezia, T. A. et al. Molecular signatures of proliferation and quiescence in hematopoietic stem cells. PLoS Biol. 2, e301 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Cheng, H. et al. Leukemic marrow infiltration reveals a novel role for Egr3 as a potent inhibitor of normal hematopoietic stem cell proliferation. Blood 126, 1302–1313 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kubota, Y., Osawa, M., Jakt, L. M., Yoshikawa, K. & Nishikawa, S. Necdin restricts proliferation of hematopoietic stem cells during hematopoietic regeneration. Blood 114, 4383–4392 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Dong, F. et al. Mouse acute leukemia develops independent of self-renewal and differentiation potentials in hematopoietic stem and progenitor cells. Blood Adv. 3, 419–431 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. 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 

  32. Min, I. M. et al. The transcription factor EGR1 controls both the proliferation and localization of hematopoietic stem cells. Cell Stem Cell 2, 380–391 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Copley, M. R. et al. The Lin28b–let-7–Hmga2 axis determines the higher self-renewal potential of fetal haematopoietic stem cells. Nat. Cell Biol. 15, 916–925 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Masumi, A. et al. Interferon regulatory factor-2 induces megakaryopoiesis in mouse bone marrow hematopoietic cells. FEBS Lett. 583, 3493–3500 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Haug, J. S. et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell 2, 367–379 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Wilkinson, A. C. et al. Long-term ex vivo haematopoietic-stem-cell expansion allows nonconditioned transplantation. Nature 571, 117–121 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Grinenko, T. et al. Hematopoietic stem cells can differentiate into restricted myeloid progenitors before cell division in mice. Nat. Commun. 9, 1898 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. 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 

  39. Tusi, B. K. et al. Population snapshots predict early haematopoietic and erythroid hierarchies. Nature 555, 54–60 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Weinreb, C., Wolock, S., Tusi, B. K., Socolovsky, M. & Klein, A. M. Fundamental limits on dynamic inference from single-cell snapshots. Proc. Natl Acad. Sci. USA 115, E2467–E2476 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Plett, P. A., Frankovitz, S. M. & Orschell-Traycoff, C. M. In vivo trafficking, cell cycle activity, and engraftment potential of phenotypically defined primitive hematopoietic cells after transplantation into irradiated or nonirradiated recipients. Blood 100, 3545–3552 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Hu, L. et al. Antioxidant N-acetyl-l-cysteine increases engraftment of human hematopoietic stem cells in immune-deficient mice. Blood 124, e45–e48 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ema, H. et al. Adult mouse hematopoietic stem cells: purification and single-cell assays. Nat. Protoc. 1, 2979–2987 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  46. McCarthy, D. J., Campbell, K. R., Lun, A. T. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lun, A. T., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Research 5, 2122 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. Kowalczyk, M. S. et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 25, 1860–1872 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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 

  50. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. 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  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zhang, B. et al. Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia. Cancer Cell 21, 577–592 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Green for constructive suggestions on our data and A. Wilkinson for sharing the PVA in vitro culture protocol. We are grateful to our lab members and collaborators for their insightful discussions during the course of this work and in the preparation of this manuscript. This work was supported by the grants from the National Key R&D Program of China (2016YFA0100600, 2017YFA0103400, 2017YFA0104900, 2019YFA0110203, 2018YFA0107801 and 2015CB964400), the National Natural Science Foundation of China (81421002, 81730006, 81922002, 81890990, 81861148029, 81670105 and 81870086), the CAMS Initiative for Innovative Medicine (2016-I2M-1-017, 2017-I2M-1–015, 2017-I2M-3-009 and 2019-I2M-1-006), Distinguished Young Scholars of Tianjin (19JCJQJC63400), CAMS Fundamental Research Funds for Central Research Institutes (2019RC310003), and the Atlas of Blood Cell Alliance. F.K.H. was funded by a MRC studentship (MR/K500975/1), and F.K.H. and B.G. were supported by infrastructure funding from the Wellcome and MRC to the Wellcome & MRC Cambridge Stem Cell Institute (203151/Z/16/Z, MC_PC_12009).

Author information

Authors and Affiliations

Authors

Contributions

F.D., S.H., S.Z., C.Z. and H.C. designed and performed the experiments, analysed the data and wrote the manuscript. X.W., A.G. and F.W. helped with the mouse experiments and flow cytometry. Y.G., Z.Y. and C.W. helped with single-cell sequencing. F.K.H. and J.D. helped with the scRNA-seq data analysis and assisted with the manuscript. J.W., B.L., Y.L., H.E. and F.T. assisted with the manuscript. B.G. and P.Z. performed the bioinformatics analysis and assisted with the manuscript. T.C. conceived the study, designed the experiments, interpreted the results, wrote the paper and oversaw the research project.

Corresponding authors

Correspondence to Berthold Göttgens, Ping Zhu or Tao Cheng.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Flow cytometry plots of 28 cell populations isolated from bone marrow (BM) or spleen (SP) of mice for scRNA-seq.

Highly purified 28 populations including: a-c, 5 haematopoietic stem cell (HSC) populations (BM): HSCLT, Fraction I, Fraction III, ESLAM, ESLAMSK. a, c, d, 9 multipotent progenitor (MPP) populations (BM): HSCST, LMPP, MPP1, MPP2, MPP3, MPP4, Fraction II, HPC2 and HPC3. e, h, 4 committed progenitor populations (BM): Common Myeloid Progenitor (CMP), Granulocyte Macrophage Progenitor (GMP), Megakaryocyte Erythrocyte Progenitor (MEP), and Common Lymphoid Progenitor (CLP). f, 2 erythroid populations (BM): Erythrocyte A and Erythrocyte B (EryA and EryB). g, 3 mature myeloid cell populations (BM): Granulocyte, Monocyte and Macrophage, 1 Megakaryocyte population (BM): Megakaryocytes (Mk). i, j, k, 4 mature lymphoid cell populations (SP): B cell, CD4+ T cell, CD8+ T cell and Natural Killer (NK) cell. 2 independent experiments for cell sorting.

Extended Data Fig. 2 The comparisons of scRNA-seq data with published results in the same immunophenotypical cell populations.

a, Diffusion map comparisons were performed on HSCLT, LMPP, MEP and GMP cells in Sonia Nestorowa’s paper (PMID:27365425) and this study. b, Visualization of cell types defined in Sonia Nestorowa’s paper by force-directed graph with broad gating (LSK cells) and each highlighted with red colour. c, Immunophenotypical HSCs and MPPs sorted by FACS in this study were projected onto Sonia Nestorowa’s data, and each cell population was found in the k-nearest neighbours of cell types defined in Sonia Nestorowa’s data22. Cells were coloured by log-transformed score.

Extended Data Fig. 3 Clustering of committed progenitor cells and lineage cells according to the transcriptome profiling.

a, Heatmap and unsupervised hierarchical clustering of diversely expressed genes in 4 immunophenotypical committed progenitors (CPs) (CMP, GMP, MEP and CLP). Representative GO enrichments of specifically expressed genes for each cluster were listed on the right. 3 types of CPs (tCP1, 2 and 3) were thus grouped according to transcriptome profiling. b, Compositions of tCPs by immunophenotypical CPs. Cell numbers were n=39/26/30 for tCP1/2/3. c, Heatmap and unsupervised hierarchical clustering of diversely expressed genes in 3 immunophenotypical megakaryocytes and erythrocytes (MEs). Representative GO enrichments of specifically expressed genes for each cluster were listed on the right respectively. 3 types of MEs (tME1, 2 and 3) were thus grouped according to transcriptome profiling. d, Compositions of tMEs by immunophenotypical MEs. Cell numbers were n=41/29/19 for tME1/2/3. e, Heatmap and unsupervised hierarchical clustering of diversely expressed genes in 3 immunophenotypical granulocytes and macrophages/monocytes (GMs). Representative GO enrichments of specifically expressed genes for each cluster were listed on the right respectively. 3 types of GMs (tGM1,2 and 3) were thus grouped according to transcriptome profiling. f, Compositions of tGMs by immunophenotypical GMs. Cell numbers were n=46/24/36 for tGM1/2/3. g, Heatmap and unsupervised hierarchical clustering of diversely expressed genes in 4 immunophenotypical lymphocytes including B, CD4+ T, CD8+ T and NK cells (Lyms). Representative GO enrichments of specifically expressed genes for each cluster were listed on the right respectively. 4 types of Lyms (tLym1, 2, 3 and 4) were thus grouped according to transcriptome profiling. h, Compositions of tLyms by immunophenotypical Lyms (1-4). Cell numbers were n=48/42/41/36 for tLym1/2/3/4.

Source data

Extended Data Fig. 4 Enriched GO terms from each classified cell population and validation of cell type classifications and annotations.

a, Enriched GO terms from each cell type based on specifically expressed genes. Top five GO terms enriched for each gene sets specifically expressed in each cluster under homeostasis. Dot color indicates the logarithmic transformed adjusted P value. Dot size indicates enrichment score estimated by Enrichr. Cell numbers were n=189/93/23 for tHSC1/2/3, n=100/145/95/93/75 for tMPP1/2/3/4/5/, n=39/26/30 for tCP1/2/3, n=41/29/19 for tME1/2/3, n=46/24/36 for tGM1/2/3, n=48/42/41/36 for tLym1/2/3/4 (logarithmic transformed adjusted P values with Benjamini-Hochberg correction). b, Heatmap showed the prediction results of cell type determination by the classifier build on the unique gene expressions of haematopoietic cells under homeostasis.

Source data

Extended Data Fig. 5 Cell cycle analysis of different cell types based on transcriptome profiling and characteristics comparison among subtypes of tHSCs and tMPPs.

a, Cell cycle distributions (G0, G1, S and G2/M) of cell populations based on expression levels of known genes related to cell cycle. b, GSEA results of HSC state (HSC signature and quiescence) and lineage differentiation (Megakaryocytic, erythroid, myeloid and lymphoid) associated gene sets among tHSC1, 2, and 3 under homeostasis. Cell numbers were n=189/93/23 for tHSC1/2/3 (Permutation test, two-side). c, Box plots showing the expression level (mean normalized expressed of each gene sets) of HSC signature, proliferation, CLP, pre-megakaryocyte-erythrocyte, megakaryocyte progenitor, and pre-granulocyte/monocyte commitment related genes in all single cells of tMPP populations under homeostasis Box represents the first and the third quartile of the expression levels and whiskers above and below the box show the locations of the minimum and maximum values except outliers. The line in the box represents the median value and the black circles represent outliers. Cell numbers were n=100/145/95/93/75 for tMPP1/2/3/4/5 (unpaired t-test, two-side).

Source data

Extended Data Fig. 6 Specifically expressed genes on HSPC cell populations.

a, The expression of specially expressed genes in HSPCs (Procr, Esam, Mmrn1, Sult1a1) were displayed using published data (Nestorowa’s paper, PMID:27365425, n=1656). Colors indicate the gene expression levels. b, Expression levels of genes (Procr, Esam, Mmrn1, Sult1a1) and other representative genes (Necdin, Dst, and Tgm2) were shown on the t-SNE map in this study (n=1270). Colors indicate the gene expression levels. Top-left panel showed the cell type annotations with indicated colors.

Extended Data Fig. 7 Differentially expressed genes and transcription factor regulation of engrafted cells.

a, Differentially expressed genes in transplanted ESLAMSK cells compared with donor-derived GFP+ cells at indicated time points. b, Enriched transcription factor regulation in donor-derived GFP+ cells at indicated time points. Representative genes for myeloid, erythroid and self-renewal signatures were listed on the right with dynamic changes.

Extended Data Fig. 8 Validation of cell type classification after transplantation and comparisons of apoptosis and autophagy related gene sets in tHSC1/2 after transplantation.

a–d, Heatmap showed the consistent expression patterns of cell type specifically expressed genes under homeostasis (Ctrl) and after transplantation (Tx) for each cell type, indicating high accuracy of cell classification of single cells after transplantation. a, tHSCs, b, tMPPs, c, tMEs, d, tGMs. Colors from purpose to red indicate low to high gene expression levels. e, Additional 137 donor-derived cells were collected from BM and SP of 3 individual recipient mice at day 1 after transplantation and single-cell RNA-seq was performed on these cells. f. GSEA results of apoptosis and autophagy related gene sets in Tx tHSC1/2 comparing with ESLAMSK tHSC1/2 (n=16/15 for Tx tHSC1/2, n=34/22 for ESLAMSK tHSC1/2, Permutation test, two-side).

Source data

Extended Data Fig. 9 Characteristics of tMPPs after transplantation.

a, Cell cycle analysis (S/G2/M) of Tx tMPPs compared with the counterparts under homeostasis. b, GSEA of indicated gene signatures (HSC signature, proliferation, megakaryocytic, erythroid, myeloid and lymphoid) comparing Tx tMPPs with tMPPs under homeostasis respectively. Red squares indicate positive enrichment of Tx tMPPs and blue squares indicate negative enrichment of Tx tMPPs (Permutation test, two-side). c, The expression of lineage specific transcription factors or marker genes on tMPP populations after transplantation (Tx) and the counterparts under homeostasis (Ctrl). Boxplot displaying distribution of normalized expressed of each gene within cluster. Box represents the first and the third quartile of the expression levels and whiskers below and above the box show the locations of the minimum and maximum values except outliers. The line in the box represents the median value and the black circles represent outliers. *, p<0.05, **, p<0.01, ***, p<0.001 (unpaired two-sided t-test). Cell numbers (for b and c) were n=169/348/114/56/166 for Tx, and n=100/145/95/93/75 for tMPP1/2/3/4/5 for Ctrl.

Source data

Extended Data Fig. 10 Differentially expressed genes (DEGs) and dynamic transcriptome changes of tMPPs after transplantation.

a, Volcano plots showed the DEGs between tMPP1/2/3/4/5 under homeostasis and after transplantation. Each dot represents a gene. The red dots represent high expressed genes after transplantation and the blue dots represent low expressed genes under homeostasis. The black dots represent no significant difference in expression. Cell numbers were n=100/145/95/93/75 for tMPP1/2/3/4/5 under homeostasis, and n=169/348/114/56/166 for tMPP1/2/3/4/5 after transplantation (P value were estimated by two-side t-test and corrected by Benjamini-Hochberg method). b, Heatmaps showed the mean expressed, transiently down-regulated genes (estimated by ImpulseDE2 package) in single cells under homeostasis (Ctrl) and after transplantation (D1/3/5/7) of tMPP1/2/3/4/5. Each row represents a gene and each column represents a time point of tMPPs. The table on the right of each heatmap showed the top 10 KEGG pathways enrichment of corresponding genes, RC* means endoplasmic reticulum, * in heatmap means transient, (number) means the number of genes changed expressions. Cell numbers were n=100/145/95/93/75 for MPP1/2/3/4/5 in Ctrl, n=32/53/68/16 for tMPP1 D1/3/5/7, n=28/53/170/97 for tMPP2 D1/3/5/7, n=6/107 for tMPP3 D5/7, n=6/4/20/26 for tMPP4 D1/3/5/7, and n=10/17/76/63 for tMPP5 D1/3/5/7 after transplantation (P values were estimated by two-side hypergeometric test). c, The normalized expression value of Ifitm1 in different cell populations after transplantation (Tx) compared with the counterparts under homeostasis (Ctrl). Box represents the first and the third quartile of the expression levels and whiskers below and above the box show the locations of the minimum and maximum values except outliers. The line in the box represents the median value and the black circles represent outliers. The red circles show the expression of individual cells within the cell type with less than 10 cells. Cell numbers were n=189/93/23 for tHSC1/2/3, n=100/145/95/93/75 for tMPP1/2/3/4/5/, n=39/26/30 for tCP1/2/3, n=41/29/19 for tME1/2/3, n=46/24/36 for tGM1/2/3, n=48/42/41/36 for tLym1/2/3/4 in Ctrl group. Cell numbers were n=16/15/2 for tHSC1/2/3, n=169/348/114/56/166 for tMPP1/2/3/4/5/, n=6/1 for tCP1/3, n=50/2 for tME1/2/3, n=68/8 for tGM1/3, n=3/3/3/1 for tLym1/2/3/4 in Tx group (Two-side Wilcoxon test). d, Schematic model of blood cell production under homeostasis and regeneration condition.

Supplementary information

Supplementary Tables 1–4

Supplementary Table 1: Immunophenotypes of haematopoietic cells for scRNA-seq. Supplementary Table 2: The estimated HSC frequencies in cells collected at different time points through limiting dilution analysis. Supplementary Table 3: The catalogue numbers for Taqman probes. Supplementary Table 4: The catalogue numbers for antibodies.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical surce data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dong, F., Hao, S., Zhang, S. et al. Differentiation of transplanted haematopoietic stem cells tracked by single-cell transcriptomic analysis. Nat Cell Biol 22, 630–639 (2020). https://doi.org/10.1038/s41556-020-0512-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0512-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research