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:

Independent origins of fetal liver haematopoietic stem and progenitor cells

Abstract

Self-renewal and differentiation are tightly controlled to maintain haematopoietic stem cell (HSC) homeostasis in the adult bone marrow1,2. During fetal development, expansion of HSCs (self-renewal) and production of differentiated haematopoietic cells (differentiation) are both required to sustain the haematopoietic system for body growth3,4. However, it remains unclear how these two seemingly opposing tasks are accomplished within the short embryonic period. Here we used in vivo genetic tracing in mice to analyse the formation of HSCs and progenitors from intra-arterial haematopoietic clusters, which contain HSC precursors and express the transcription factor hepatic leukaemia factor (HLF). Through kinetic study, we observed the simultaneous formation of HSCs and defined progenitors—previously regarded as descendants of HSCs5—from the HLF+ precursor population, followed by prompt formation of the hierarchical haematopoietic population structure in the fetal liver in an HSC-independent manner. The transcription factor EVI1 is heterogeneously expressed within the precursor population, with EVI1hi cells being predominantly localized to intra-embryonic arteries and preferentially giving rise to HSCs. By genetically manipulating EVI1 expression, we were able to alter HSC and progenitor output from precursors in vivo. Using fate tracking, we also demonstrated that fetal HSCs are slowly used to produce short-term HSCs at late gestation. These data suggest that fetal HSCs minimally contribute to the generation of progenitors and functional blood cells before birth. Stem cell-independent pathways during development thus offer a rational strategy for the rapid and simultaneous growth of tissues and stem cell pools.

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: Fate tracing of HCCs reveals HSC-independent progenitor formation from pre-HSPCs.
Fig. 2: Intra-embryonic EVI1hi cells preferentially generate HSCs.
Fig. 3: EVI1 expression level determines the fate of pre-HSPCs.
Fig. 4: Fetal HSCs minimally contribute to progenitors and mature cells.

Similar content being viewed by others

Data availability

All RNA-seq data were deposited in the Gene Expression Omnibus under accessions GSE167932 (E10.5 and E11.5 scRNA-seq), GSE168054 (E10.5 bulk RNA-seq) and GSE190011 (E14.5 bulk RNA-seq). Source data are provided with this paper.

References

  1. Haas, S., Trumpp, A. & Milsom, M. D. Causes and consequences of hematopoietic stem cell heterogeneity. Cell Stem Cell 22, 627–638 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dzierzak, E. & Bigas, A. Blood development: hematopoietic stem cell dependence and independence. Cell Stem Cell 22, 639–651 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Traver, D. et al. Fetal liver myelopoiesis occurs through distinct, prospectively isolatable progenitor subsets. Blood 98, 627–635 (2001).

    Article  CAS  PubMed  Google Scholar 

  6. 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 

  7. 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 

  8. 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 

  9. 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 

  10. Chapple, R. H. et al. Lineage tracing of murine adult hematopoietic stem cells reveals active contribution to steady-state hematopoiesis. Blood Adv. 2, 1220–1228 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sawai, C. M. et al. Hematopoietic stem cells are the major source of multilineage hematopoiesis in adult animals. Immunity 45, 597–609 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sawen, P. et al. Murine HSCs contribute actively to native hematopoiesis but with reduced differentiation capacity upon aging. eLife 7, e41258 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Draper, J. E. et al. A novel prospective isolation of murine fetal liver progenitors to study in utero hematopoietic defects. PLoS Genet. 14, e1007127 (2018).

    Article  PubMed  PubMed Central  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  PubMed  Google Scholar 

  15. Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chen, M. J., Yokomizo, T., Zeigler, B. M., Dzierzak, E. & Speck, N. A. Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter. Nature 457, 887–891 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Boisset, J. C. et al. In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature 464, 116–120 (2010).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Yokomizo, T. & Dzierzak, E. Three-dimensional cartography of hematopoietic clusters in the vasculature of whole mouse embryos. Development 137, 3651–3661 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Taoudi, S. et al. Extensive hematopoietic stem cell generation in the AGM region via maturation of VE-cadherin+CD45+ pre-definitive HSCs. Cell Stem Cell 3, 99–108 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Zhou, F. et al. Tracing haematopoietic stem cell formation at single-cell resolution. Nature 533, 487–492 (2016).

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Kieusseian, A., Brunet de la Grange, P., Burlen-Defranoux, O., Godin, I. & Cumano, A. Immature hematopoietic stem cells undergo maturation in the fetal liver. Development 139, 3521–3530 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Rybtsov, S., Ivanovs, A., Zhao, S. & Medvinsky, A. Concealed expansion of immature precursors underpins acute burst of adult HSC activity in foetal liver. Development 143, 1284–1289 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yokomizo, T. et al. Hlf marks the developmental pathway for hematopoietic stem cells but not for erythro-myeloid progenitors. J. Exp. Med. 216, 1599–1614 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. McGrath, K. E. et al. Distinct sources of hematopoietic progenitors emerge before HSCs and provide functional blood cells in the mammalian embryo. Cell Rep. 11, 1892–1904 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kim, I., He, S., Yilmaz, O. H., Kiel, M. J. & Morrison, S. J. Enhanced purification of fetal liver hematopoietic stem cells using SLAM family receptors. Blood 108, 737–744 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Medvinsky, A. & Dzierzak, E. Definitive hematopoiesis is autonomously initiated by the AGM region. Cell 86, 897–906 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Cumano, A., Dieterlen-Lievre, F. & Godin, I. Lymphoid potential, probed before circulation in mouse, is restricted to caudal intraembryonic splanchnopleura. Cell 86, 907–916 (1996).

    Article  CAS  PubMed  Google Scholar 

  28. Goyama, S. et al. Evi-1 is a critical regulator for hematopoietic stem cells and transformed leukemic cells. Cell Stem Cell 3, 207–220 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Glass, C., Wilson, M., Gonzalez, R., Zhang, Y. & Perkins, A. S. The role of EVI1 in myeloid malignancies. Blood Cells Mol. Dis. 53, 67–76 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Kataoka, K. et al. Evi1 is essential for hematopoietic stem cell self-renewal, and its expression marks hematopoietic cells with long-term multilineage repopulating activity. J. Exp. Med. 208, 2403–2416 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ema, H. & Nakauchi, H. Expansion of hematopoietic stem cells in the developing liver of a mouse embryo. Blood 95, 2284–2288 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Gekas, C., Dieterlen-Lievre, F., Orkin, S. H. & Mikkola, H. K. The placenta is a niche for hematopoietic stem cells. Dev. Cell 8, 365–375 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Khan, J. A. et al. Fetal liver hematopoietic stem cell niches associate with portal vessels. Science 351, 176–180 (2016).

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Dignum, T. et al. Multipotent progenitors and hematopoietic stem cells arise independently from hemogenic endothelium in the mouse embryo. Cell Rep. 36, 109675 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ema, H., Uchinomiya, K., Morita, Y., Suda, T. & Iwasa, Y. Repopulation dynamics of single haematopoietic stem cells in mouse transplantation experiments: Importance of stem cell composition in competitor cells. J. Theor. Biol. 394, 57–67 (2016).

    Article  ADS  MathSciNet  CAS  PubMed  MATH  Google Scholar 

  36. Ulloa, B. A. et al. Definitive hematopoietic stem cells minimally contribute to embryonic hematopoiesis. Cell Rep. 36, 109703 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Doulatov, S. et al. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell 13, 459–470 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Elcheva, I. et al. Direct induction of haematoendothelial programs in human pluripotent stem cells by transcriptional regulators. Nat. Commun. 5, 4372 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  39. Sugimura, R. et al. Haematopoietic stem and progenitor cells from human pluripotent stem cells. Nature 545, 432–438 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tsukada, M. et al. In vivo generation of engraftable murine hematopoietic stem cells by Gfi1b, c-Fos, and Gata2 overexpression within Teratoma. Stem Cell Reports 9, 1024–1033 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Matsuo, J. et al. Identification of stem cells in the epithelium of the stomach corpus and antrum of mice. Gastroenterology 152, 218–231.e14 (2017).

    Article  PubMed  Google Scholar 

  42. Okabe, K. et al. Neurons limit angiogenesis by titrating VEGF in retina. Cell 159, 584–596 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. 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 

  44. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003).

    Article  PubMed  Google Scholar 

  46. Kisanuki, Y. Y. et al. Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo. Dev. Biol. 230, 230–242 (2001).

    Article  CAS  PubMed  Google Scholar 

  47. Ishikawa, E. et al. Protein kinase D regulates positive selection of CD4+ thymocytes through phosphorylation of SHP-1. Nat. Commun. 7, 12756 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yokomizo, T. et al. Whole-mount three-dimensional imaging of internally localized immunostained cells within mouse embryos. Nat. Protoc. 7, 421–431 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, J. et al. An in situ atlas of mitochondrial DNA in mammalian tissues reveals high content in stem and proliferative compartments. Am. J. Pathol. 190, 1565–1579 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ransick, A. et al. Single-cell profiling reveals sex, lineage, and regional diversity in the mouse kidney. Dev. Cell 51, 399–413.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mercer, T. R. et al. The human mitochondrial transcriptome. Cell 146, 645–658 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Warnes, G. R. et al. gplots: various R programming tools for plotting data. R package version 3.1.1 (2020).

  55. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Vink, C. S. et al. Iterative single-cell analyses define the transcriptome of the first functional hematopoietic stem cells. Cell Rep. 31, 107627 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Fadlullah, M. Z. H. et al. Murine AGM single-cell profiling identifies a continuum of hemogenic endothelium differentiation marked by ACE. Blood 139, 343–356 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hayashi, T. et al. Single-cell full-length total RNA sequencing uncovers dynamics of recursive splicing and enhancer RNAs. Nat. Commun. 9, 619 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  59. Gazit, R. et al. Transcriptome analysis identifies regulators of hematopoietic stem and progenitor cells. Stem Cell Rep. 1, 266–280 (2013).

    Article  CAS  Google Scholar 

  60. Perdiguero, E. G. et al. The origin of tissue-resident macrophages: when an erythro-myeloid progenitor is an erythro-myeloid progenitor. Immunity 43, 1023–1024 (2015).

    Article  PubMed  Google Scholar 

  61. Soares-da-Silva, F. et al. Yolk sac, but not hematopoietic stem cell-derived progenitors, sustain erythropoiesis throughout murine embryonic life. J. Exp. Med. 218, e20201729 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Dzierzak for critical comments on this manuscript; and T. Umemoto, M. Kataoka and R. Koitabashi for assistance with RNA sequencing. This work was supported by JSPS Kakenhi Grant (20K08758, JP16H0627 (AdAMS) to T.Y. and 26221309 to T. Suda), SENSHIN Medical Research Foundation (to T.Y.), Takeda Science Foundation (to T.Y.), Japanese Society of Hematology (to T.Y.), the Sumitomo Foundation (to T.Y.), the National Medical Research Council grant of Singapore Translational Research Investigator Award (NMRC/STaR/0019/2014 to T. Suda) and the programme of the Inter-University Research Network for High Depth Omics, Institute of Molecular Embryology and Genetics, Kumamoto University (to S.M.-K. and M. Ogawa).

Author information

Authors and Affiliations

Authors

Contributions

T.Y. conceived the project, designed and performed the research, and wrote the manuscript. T.I. made the targeting constructs and performed whole-mount immunostaining. S.M.-K. and M. Ogawa performed the scRNA-seq analysis. C.Y.T. and M. Osato performed the bioinformatic analysis. N.T. and K.A. generated mouse lines. T. Sato, Y.K., M.K. and N.K. contributed analytical tools. T.Y., M. Osato and T. Suda participated in project planning.

Corresponding authors

Correspondence to Tomomasa Yokomizo or Toshio Suda.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Anna Bigas, Georges Lacaud and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Hematopoietic stem/progenitor cells in the late gestational fetal liver.

a, Disappearance of EMPs at mid-late gestation. Since Hlf is not expressed in EMPs (red rectangle), EMPs and other progenitors can be distinguished by HlftdTomato reporter23. Top right, Frequency of Hlf-tdTomato+ cells in c-Kithi cells (E11.5, n = 6; E14.5, n = 6; E18.5, n = 5; adult, n = 3). b, Waves of hematopoietic progenitors in the fetal liver. Schematic is drawn based on previous reports24,60,61 and our data (a). At E14.5, most c-Kit+ progenitors in the fetal liver are classical HSC-dependent progenitors. c, Representative flow cytometry plots and hematopoietic stem/progenitor cell hierarchy in the bone marrow. Circle size represents the population size of the defined fractions. d, Representative flow cytometry plots and hematopoietic stem/progenitor cell hierarchy in the fetal liver (E14.5). Relative population size of progenitor fractions is similar to that of bone marrow progenitors. The hematopoietic stem/progenitor cell hierarchy in the late gestational fetal liver is believed to be established through the differentiation of fetal HSCs (arrows in the hierarchy) in the standard model. e, Transplantation experiments. Irradiated mice were transplanted with 100 donor cells (HSC, ST-HSC, or MPP) sorted from E14.5 fetal liver. n = 10.

Source data

Extended Data Fig. 2 Generation and characterization of HlfCreERT2 mice.

a, Experimental design for precursor labeling by CreERT2 lines. 1; Labeling of endothelial cells (e.g. Runx1-CreERT2 and VEcad-CreERT2). Labeled endothelial cells may transform into hematopoietic cells at a later time, leading to the mixture of cells at various stage of maturation. 2; Labeling of hematopoietic clusters. EHT, endothelial-to-hematopoietic transition. b, Whole-mount immunostaining analysis of E10.5 (35 sp) HlftdTomato embryos for tdTomato (red), c-Kit (green) and CD31 (white). Scale bar, 100 μm. c, Whole-mount immunostaining analysis of E10.5 (33 sp) HlftdTomato yolk sac for tdTomato (red), c-Kit (green) and CD31 (white). Scale bar, 50 μm. d, Targeting strategy of the HlfCreERT2 mouse. e, White blood cell (WBC), red blood cell (RBC), and platelet (PLT) fractions in the blood of adult Hlf+/+ and HlfCreERT2/+ mice (Hlf+/+ mice, n = 7; HlfCreERT2/+ mice, n = 9). f, Bone marrow (BM) cellularity (Hlf+/+ mice, n = 5; HlfCreERT2/+ mice, n = 6). g, HSC and progenitor fractions in HlfCreERT2/+ mice (Hlf+/+ mice, n = 4; HlfCreERT2/+ mice, n = 5). h, Whole-mount immunostaining analysis of E10.75 (tamoxifen at E10.0) HlfCreERT2ROSAtdTomato embryos. Scale bars, 50 μm. i, Frequency of tdTomato+ cells in c-Kit+ cells (Dorsal aorta, n = 6; Yolk sac, n = 3; Vitelline artery, n = 6; Umbilical artery, n = 6). j, HSC analysis. HlfCreERT2ROSAtdTomato embryos were administered with tamoxifen at E10.5 and were analyzed at E14.5. k, EMP analysis. HlfCreERT2ROSAtdTomato embryos were administered with tamoxifen at E8.0 and were analyzed at E10.5. l, Contribution of HlfCreEERT2-labeled cells to adult hematopoiesis. HlfCreERT2ROSAtdTomato embryos were administered with tamoxifen at E9.75. n = 5. m, Lympho-myeloid progenitor (LMP) analysis. HlfCreERT2ROSAtdTomato embryos were administered with tamoxifen at E9.75 and were analyzed at E11.5. n = 8. All error bars represent means ± SD. Statistical analysis was performed using two-sided unpaired Student’s t-test (eg).

Source data

Extended Data Fig. 3 Fate tracing of Hlf+ cells.

a, Phenotypic comparison between E10.5 Hlf+c-Kit+ hematopoietic cluster cells and HlfCreERT2 -labeled cells in E11.5 fetal livers (tamoxifen at E9.75). b, Representative flow cytometry plots of HlfCreERT2-labeled cells in the AGM and yolk sac (red dots). c, Representative flow cytometry plots of tdTomato fraction in the fetal liver and AGM of HlfCreERT2ROSAtdTomato embryos. d, Top, Schematic of tamoxifen treatment and analysis. Middle, Lineage tracing of Hlf+ cells. HlfCreERT2ROSAtdTomato embryos were administered with tamoxifen at various stages (E9.0–11.5) and were analyzed at E14.5. Each graph represents data from one litter. The highest frequency population is shaded (KL, gray; MPP, blue; HSC, orange). Bottom, Proportion of dominant-cell types.

Source data

Extended Data Fig. 4 Heterogeneity within pre-HSPCs.

a, Schematic of single-cell (sc)RNA-seq analysis. Middle segment of the dorsal aorta region and a lobe of the fetal liver were dissected to generate single cells from E10.5 and E11.5 embryos. b, Uniform manifold approximation and projection (UMAP) visualization of 13,193 cells isolated from the dorsal aorta and its surrounding tissues, including the fetal liver. Combined data from E10.5 (6,608 cells) and E11.5 (6,585 cells) are plotted. PGC, primordial germ cell; SMC, smooth muscle cell; Im, intermediate mesoderm. c, UMAP visualization of hematopoietic/endothelial clusters colored by representative lineage-specific genes (EC, endothelial cells; Ery, erythroid lineage; Meg, megakaryocytic lineage; My, myeloid lineage). d, Identification of Hlfhic-Kithi pre-HSPC population. e, UMAP visualization of stem-associated genes. f, UMAP visualization of committed hematopoietic marker genes. g, Hierarchical clustering of the Hlfhic-Kithi pre-HSPC population from E10.5 and E11.5 (marked by black circles) embryos. h, Identification of pre-HSPCs (Hlf+c-Kit+) in CD31+c-KithiGata2med AGM single cells. SPRING visualization of selected genes from the interactive website (https://gottgens-lab.stemcells.cam.ac.uk/DZIERZAK/). i, HSC score. j, SPRING visualization of stem-associated genes. k, SPRING visualization of committed hematopoietic marker genes. l, Hierarchical clustering of the pre-HSPC population. Hlf+c-Kit+ cells (n = 52) were extracted from the Vink et al. dataset (GSE143637). m, Hierarchical clustering of the pre-HSPC population. Hlf+c-Kit+ cells (n = 27) were extracted from the Fadlullah et al. dataset (GSE150412).

Source data

Extended Data Fig. 5 Evi1 expression in E10.5-11.5 embryos.

a, b, Whole-mount immunostaining analysis of E10.5 (36 sp) Evi1GFP embryos for GFP (green) and CD31 (white). a, Representative immunofluorescent images of the embryo proper and yolk sac. White arrows indicate the entrance of the yolk sac artery. GFPlo cells are detected around this region. b, Representative immunofluorescent images of the umbilical artery, vitelline artery, and umbilical vein. Orange arrows indicate hematopoietic clusters. White arrows indicate the walls of the umbilical vein. Scale bars, 50 μm. DA, dorsal aorta; VA, vitelline artery; UA, umbilical artery; UV, umbilical vein. c, Flow cytometry analysis of Evi1-GFP expression in Evi1GFPHlftdTomato embryos. Left, Representative flow cytometry plots of the E11.5 Evi1GFP/+HlftdTomato/+ AGM region. Right, Mean fluorescence intensity (MFI) of Evi1-GFP in the HCC fraction of the AGM region (E10.5, n = 3; E11.5, n = 4). HCC, hematopoietic cluster cell. All error bars represent means ± SD. Statistical analysis was performed using two-sided unpaired Student’s t-test (c).

Source data

Extended Data Fig. 6 Fate tracing of Evi1hi cells.

a, Targeting strategy of the Evi1CreERT2 mouse. b, Fetal liver (FL) cellularity (Evi1+/+ mice, n = 7; Evi1CreERT2/+ mice, n = 10). c, HSC and progenitor fractions in Evi1CreERT2/+ (Evi1+/+ mice, n = 7; Evi1CreERT2/+ mice, n = 10; Evi1GFP/+ mice, n = 5). See Supplementary Discussion for HSC reduction in Evi1CreERT2 and Evi1GFP embryos. d, Labeling of Evi1-GFPhi cells by Evi1CreERT2. Evi1CreERT2 mice were crossed with Evi1GFPROSAtdTomato mice to obtain Evi1CreERT2/GFPROSAtdTomato embryos (tamoxifen at E10.5). n = 6. e, Whole-mount immunostaining analysis of E11.25 (tamoxifen at E10.5) Evi1CreERT2ROSAtdTomato embryos. Labeled endothelial cells and hematopoietic clusters are observed in the dorsal aorta. Scale bars, 100 μm. f, Whole-mount immunostaining analysis of E11.25 (tamoxifen at E10.5) Evi1CreERT2ROSAtdTomato embryos. In contrast to the vitelline and umbilical arteries, tdTomato+c-Kit+ cells are rarely observed in the yolk sac or fetal liver (orange arrows). Scale bars, 100 μm. g, Lineage tracing of Evi1+ cells. Evi1CreERT2ROSAtdTomato embryos were administered with tamoxifen at various stages (E8.5 and E9.5) and were analyzed at E14.5 (E8.5, n = 4; E9.5, n = 4). h, Whole-mount immunostaining analysis of E9.5 (tamoxifen at E8.5) Evi1CreERT2ROSAtdTomato embryo. Flat-shaped endothelial cells are labeled. Scale bar, 200 μm. All error bars represent means ± SD. Statistical analysis was performed two-sided unpaired Student’s t-test (b and c).

Source data

Extended Data Fig. 7 Evi1extreme hi cells preferentially generate HSCs.

a, Gene set enrichment analysis (GSEA) of an Evi1+ subset (Hlf+c-Kit+CD45 cells from caudal half region) compared with an Evi1lo/− subset (Hlf+c-Kit+CD45 cells from yolk sac) for HSC, MPP, and HSPC signatures. NES, normalized enrichment score; FDR, false discovery rate. b, Experimental design to trace the fate of Evi1extreme hi cells and Evi1hi cells within the same mouse (Evi1CreERT2ROSAtdTomato/YFP dual-reporter embryo). Given that the recombination rate of the ROSA26 locus is correlated with the expression level of Evi1 (Extended Data Fig. 6d), it can be inferred that tdTomato+YFP+ cells (recombination of two ROSA26 loci) originate from extreme Evi1 expressors. c, Lineage tracing of Evi1extreme hi (tdTomato+YFP+) and Evi1hi (tdTomato+YFP or tdTomatoYFP+) cells. Evi1CreERT2ROSAtdTomato/YFP embryos were administered with tamoxifen at E10.5 and were analyzed at E14.5. Top, Representative flow cytometry plots of E14.5 Evi1CreERT2ROSAtdTomato/YFP fetal liver. Bottom, Comparison of cell labeling between tdTomato+YFP+ cells (Evi1extreme hi-derived) and tdTomato+YFP or tdTomatoYFP+ cells (Evi1hi-derived). n = 7. d, Models and expected outputs of tracing experiments. In the standard model, label frequencies of progenitors would be similar to that of HSCs. In the HSC-independent model, label frequencies of progenitors would be significantly lower than that of HSCs. e, Number of Hlf+c-Kit+ cells in E10.5 (32–36 sp) HlftdTomato embryos (AGM, n = 3; yolk sac, n = 4). Number was calculated from flow cytometry data. f, Number of KSL (HSCs, ST-HSCs, MPPs) cells and KL (CMPs, GMPs, MEPs) cells in E14.5 fetal liver. n = 8. Number was calculated from flow cytometry data. g, Heterogeneity of Hlf+ hematopoietic clusters in the embryo. Whole-mount immunostaining analysis of E10.5 (32 sp) Evi1GFPHlftdTomato embryo for tdTomato (red), GFP (green) and CD31 (white). Scale bar, 50 μm. All error bars represent means ± SD. Statistical analysis was performed two-sided unpaired Student’s t-test (c).

Source data

Extended Data Fig. 8 Characterization of Evi1+/− embryos.

a, Analysis of E12.5 Evi1+/ embryos. Left, Representative images of E12.5 embryos. Scale bars, 200 μm. Right, Quantitation of c-Kit+ cells in the fetal liver (Evi1+/+ mice, n = 5 ; Evi1+/ mice, n = 7). b, Normal hematopoietic cluster formation in Evi1+/ embryos. Left, Whole-mount immunostaining of Evi1+/+ (34 sp) and Evi1+/ (33 sp) embryos for c-Kit (green) and CD31 (magenta) expression. Scale bars, 100 μm. Right, Number of c-Kit+ cells localized in the middle segment of the dorsal aorta (DA). Middle segment is 7 somite-lengths18. Evi1+/+ (n = 3, 33–35 sp). Evi1+/ (n = 3, 33 and 34 sp). c, Specific decrease in stem cell fraction in Evi1 heterozygous embryos at E14.5. Left, Representative flow cytometry plots. Right, Frequency of HSC and progenitor fractions (Evi1+/+ mice, n = 8 ; Evi1+/ mice, n = 6). Similar to the results at the E12.5 stage (Fig. 3b), severe defects were observed in the HSC fractions (15-fold decrease) in Evi1+/ embryos at this time point. d, Kinetic analysis of HSC and progenitor formation from HlfCreERT2-labeled cells in Evi1+/ embryos. Top left, Schematic of tamoxifen treatment and analysis. Bottom left, Representative flow cytometry plots of HlfCreERT2-labeled cells (red dots). Right, Quantitation of tdTomato+ cells in the fetal liver (E11.5 Evi1+/+, n = 6; E12.5, Evi1+/+ n = 5; E11.5, Evi1+/ n = 5; E12.5 Evi1+/, n = 5). All error bars represent means ± SD. Statistical analysis was performed two-sided unpaired Student’s t-test (ac).

Source data

Extended Data Fig. 9 Ectopic expression of Evi1.

a, Targeting strategy of the ROSAEvi1-IRES-GFP mouse. b, Schematic of tamoxifen treatment and analysis. c, Ectopic expression of Evi1 in VE-cadherin+ cells. Left, Representative flow cytometry plots. Right, Frequency of HSC and progenitor fractions (control, VEcad-CreERT2 embryos, n = 8; VEcad-CreERT2::ROSAEvi1-IRES-GFP embryos, n = 7). d, Fate tracing of Evi1-induced VE-cadherin+ cells. Left, Representative flow cytometry plots of E14.5 fetal liver cells from a VEcad-CreERT2::ROSAtdTomato embryo (control) and a VEcad-CreERT2::ROSAEvi1-IRES-GFP embryo. Right, Frequency of HSC and KSL in E14.5 fetal livers (VEcad-CreERT2::ROSAtdTomato embryos, n = 7; VEcad-CreERT2::ROSAEvi1-IRES-GFP embryos, n = 7). eh, Characterization of HSCs in Tie2-Cre::ROSAEvi1-IRES-GFP mice. e, Flow cytometry analysis of EPCR and CD86 expression. f, Transcriptome analysis. Left, Principal component analysis (PCA). Right, Expression of HSC-related genes, presented as transcripts per kilobase million (TPM). g, Transplantation experiments. Irradiated mice were transplanted with 100 GFP+ HSCs isolated from E14.5 Tie2-Cre::ROSAEvi1-IRES-GFP fetal liver (n = 6). Donor chimerism of myeloid, B cells, and T cells in peripheral blood was analyzed by the frequency of GFP+ cells. Chimerism of CD45+ cells is shown in Fig. 3e. h, Contribution of Evi1-induced cells (GFP+ cells) to adult hematopoiesis. Although the majority (about 70%) of Tie2-Cre::ROSAEvi1-IRES-GFP mice died soon after birth for unknown reasons, the peripheral blood from the five surviving mice was analyzed (6-month-old, n = 2; 8-month old, n = 2; 1-year-old, n = 1). i, Targeting strategy of the ROSACAG-Evi1-IRES-GFP mouse. j, Frequency of GFP+ cells in fetal liver cells from HlfCreERT2ROSAEvi1-IRES-GFP (n = 8) and HlfCreERT2ROSACAG-Evi1-IRES-GFP (n = 5) embryos. Embryos were administered with tamoxifen at E9.75 and were analyzed at E14.5. All error bars represent means ± SD. Statistical analysis was performed two-sided unpaired Student’s t-test (c,d and j) and one-way ANOVA with Tukey-Kramer test (e and f).

Source data

Extended Data Fig. 10 Decrease in ST-HSC at late gestation.

a, Analysis of the KSL fraction. Top, Representative flow cytometry plots of the KSL fraction in E14.5 fetal livers (FL), E18.5 FL and 1-week bone marrow (BM). Middle, Comparison of HSPC fractions between E14.5 and E18.5. Number of HSCs, ST-HSCs, MPP, and c-Kit+Lin cells. HSPC, hematopoietic stem and progenitor cells; KSL, c-Kit+Sca-1+Lineage. Bottom, Frequency of HSC and progenitors (E14.5 embryos, n = 6; E18.5 embryos, n = 5; 1w mice, n = 6). b, Representative images of E18.5 embryos. Scale bars, 300 μm. All error bars represent means ± SD. Statistical analysis was performed two-sided unpaired Student’s t-test (a).

Source data

Supplementary information

Supplementary Discussion

Discussion on Evi1creERT2 tracing.

Reporting Summary

Supplementary Figure 1

Gating strategies to sort nascent haematopoietic cluster cells and GFP+ HSCs. a, b, Gating strategy to sort nascent haematopoietic cluster cells (Hlf+c-Kit+CD45- cells) from E10.5 HlftdTomato embryo proper and yolk sac presented on Fig. 2a. c, Gating strategy to sort GFP+ HSCs (GFP+CD150+CD48-LSK cells) from E14.5 Tie2-Cre::ROSAEvi1-IRES-GFP embryos presented on Fig. 3e.

Peer Review File

Supplementary Table 1

Markers for cell type identification in scRNA-seq (Extended Data Fig. 4b). Cell type-specific marker genes (2–5 genes) were obtained from literatures.

Supplementary Table 2

Sample size information for whole-mount immunostaining. Representative micrographs from at least two biological replicates are shown in Figures and Extended Data Figures.

Source data

Rights and permissions

Springer Nature or its licensor 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

Yokomizo, T., Ideue, T., Morino-Koga, S. et al. Independent origins of fetal liver haematopoietic stem and progenitor cells. Nature 609, 779–784 (2022). https://doi.org/10.1038/s41586-022-05203-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05203-0

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