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.

  • Review Article
  • Published:

A view of human haematopoietic development from the Petri dish

Key Points

  • Human pluripotent stem cells (hPSCs) provide outstanding new opportunities to study human haematopoietic development and blood cell disease processes in vitro. In addition, they represent a novel and potentially unlimited source of haematopoietic stem cells (HSCs) and progenitor cells for the treatment of a range of blood cell disorders.

  • Vertebrate embryonic haematopoietic development is complex and consists of multiple programmes that give rise to distinct subpopulations of haematopoietic progeny, in defined temporal patterns and at different sites within the embryo. HSCs are only generated from the definitive programme.

  • Haematopoietic development from hPSCs in vitro recapitulates many aspects of embryonic haematopoiesis, including the generation of distinct programmes. Although definitive haematopoiesis can be induced in the hPSC differentiation cultures, HSCs have not yet been produced.

  • Analyses of different stages of haematopoietic development in the hPSC differentiation cultures have provided new insights into the specification of the different haematopoietic programmes and into the generation and identity of haemogenic endothelial cells, the precursors to embryonic HSCs.

  • One of the remaining challenges in the field is the generation of hPSC-derived HSCs. Strategies that accurately mimic the processes in the embryo that lead to HSC development will be required to successfully generate these cells in vitro.

Abstract

Human pluripotent stem cells (hPSCs) provide an unparalleled opportunity to establish in vitro differentiation models that will transform our approach to the study of human development. In the case of the blood system, these models will enable investigation of the earliest stages of human embryonic haematopoiesis that was previously not possible. In addition, they will provide platforms for studying the origins of human blood cell diseases and for generating de novo haematopoietic stem and progenitor cell populations for cell-based regenerative therapies.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: A model of mouse development depicting the origin and temporal development of the different embryonic haematopoietic programmes.
Figure 2: Schematic representation of the human embryo during early gastrulation.
Figure 3: A model of human HEC development.
Figure 4: A model of human primitive, EMP and definitive haematopoietic mesoderm development.
Figure 5: A model of human HSC specification from HECs.

Similar content being viewed by others

References

  1. Vo, L. T. & Daley, G. Q. De novo generation of HSCs from somatic and pluripotent stem cell sources. Blood 125, 2641–2648 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Slukvin, I. I. Hematopoietic specification from human pluripotent stem cells: current advances and challenges toward de novo generation of hematopoietic stem cells. Blood 122, 4035–4046 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Clements, W. K. & Traver, D. Signalling pathways that control vertebrate haematopoietic stem cell specification. Nat. Rev. Immunol. 13, 336–348 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dzierzak, E. & Speck, N. A. Of lineage and legacy: the development of mammalian hematopoietic stem cells. Nat. Immunol. 9, 129–136 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dieterlen-Lievre, F. On the origin of haemopoietic stem cells in the avian embryo: an experimental approach. J. Embryol. Exp. Morphol. 33, 607–619 (1975).

    CAS  PubMed  Google Scholar 

  6. Bloom, W. & Bartelmez, G. W. Hematopoiesis in young human embryos. Am. J. Anat. 67, 21–53 (1940).

    Article  Google Scholar 

  7. Palis, J., Robertson, S., Kennedy, M., Wall, C. & Keller, G. Development of erythroid and myeloid progenitors in the yolk sac and embryo proper of the mouse. Development 126, 5073–5084 (1999). Provides a detailed analysis of yolk sac haematopoiesis: demonstrates for the first time that BFU-E progenitors, which are distinct from primitive progenitors, are specified at E8.25 in the yolk sac, providing the first evidence for the presence of EMPs.

    Article  CAS  PubMed  Google Scholar 

  8. Palis, J. Primitive and definitive erythropoiesis in mammals. Front. Physiol. 5, 3 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Naito, M., Yamamura, F., Nishikawa, S. & Takahashi, K. Development, differentiation, and maturation of fetal mouse yolk sac macrophages in cultures. J. Leukoc. Biol. 46, 1–10 (1989).

    Article  CAS  PubMed  Google Scholar 

  10. Takahashi, K., Yamamura, F. & Naito, M. Differentiation, maturation, and proliferation of macrophages in the mouse yolk sac: a light-microscopic, enzyme-cytochemical, immunohistochemical, and ultrastructural study. J. Leukoc. Biol. 45, 87–96 (1989).

    Article  CAS  PubMed  Google Scholar 

  11. Tober, J. et al. The megakaryocyte lineage originates from hemangioblast precursors and is an integral component both of primitive and of definitive hematopoiesis. Blood 109, 1433–1441 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Xu, M. J. et al. Evidence for the presence of murine primitive megakaryocytopoiesis in the early yolk sac. Blood 97, 2016–2022 (2001).

    Article  CAS  Google Scholar 

  13. Medvinsky, A. & Dzierzak, E. Definitive hematopoiesis is autonomously initiated by the AGM region. Cell 86, 897–906 (1996). Demonstrates for the first time that the first definitive HSCs emerge from the AGM region, and not the yolk sac, at E10, and do so through a novel ex vivo organ culture system that is now commonly used in the field.

    Article  CAS  PubMed  Google Scholar 

  14. de Bruijn, M. F. et al. Hematopoietic stem cells localize to the endothelial cell layer in the midgestation mouse aorta. Immunity 16, 673–683 (2002).

    Article  CAS  PubMed  Google Scholar 

  15. de Bruijn, M. F., Speck, N. A., Peeters, M. C. & Dzierzak, E. Definitive hematopoietic stem cells first develop within the major arterial regions of the mouse embryo. EMBO J. 19, 2465–2474 (2000). Demonstrates that HSCs are derived from the aorta and surrounding mesenchyme, but not the gonads or mesonephros of the AGM at E11, providing the first key insights that led to the discovery of haemogenic endothelium.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ivanovs, A. et al. Highly potent human hematopoietic stem cells first emerge in the intraembryonic aorta-gonad-mesonephros region. J. Exp. Med. 208, 2417–2427 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. Li, Z. et al. Mouse embryonic head as a site for hematopoietic stem cell development. Cell Stem Cell 11, 663–675 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Ottersbach, K. & Dzierzak, E. The murine placenta contains hematopoietic stem cells within the vascular labyrinth region. Dev. Cell 8, 377–387 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Bertrand, J. Y. et al. Characterization of purified intraembryonic hematopoietic stem cells as a tool to define their site of origin. Proc. Natl Acad. Sci. USA 102, 134–139 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Taoudi, S. et al. Progressive divergence of definitive haematopoietic stem cells from the endothelial compartment does not depend on contact with the foetal liver. Development 132, 4179–4191 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Ivanovs, A., Rybtsov, S., Anderson, R. A., Turner, M. L. & Medvinsky, A. Identification of the niche and phenotype of the first human hematopoietic stem cells. Stem Cell Rep. 2, 449–456 (2014).

    Article  Google Scholar 

  23. Yoder, M. C. et al. Characterization of definitive lymphohematopoietic stem cells in the day 9 murine yolk sac. Immunity 7, 335–344 (1997).

    Article  CAS  PubMed  Google Scholar 

  24. Yoder, M. C., Hiatt, K. & Mukherjee, P. In vivo repopulating hematopoietic stem cells are present in the murine yolk sac at day 9.0 postcoitus. Proc. Natl Acad. Sci. USA 94, 6776–6780 (1997). Shows that long-term haematopoietic engraftment can be derived from E9 yolk sac cells if engrafted into neonatal mice, thus challenging the concepts of HSC ontogeny and demonstrating the existence of the pre-HSC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rybtsov, S. et al. Tracing the origin of the HSC hierarchy reveals an SCF-dependent, IL-3-independent CD43 embryonic precursor. Stem Cell Rep. 3, 489–501 (2014).

    Article  CAS  Google Scholar 

  26. Rybtsov, S. et al. Hierarchical organization and early hematopoietic specification of the developing HSC lineage in the AGM region. J. Exp. Med. 208, 1305–1315 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Hadland, B. K. et al. Endothelium and NOTCH specify and amplify aorta-gonad-mesonephros-derived hematopoietic stem cells. J. Clin. Invest. 125, 2032–2045 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Lux, C. T. et al. All primitive and definitive hematopoietic progenitor cells emerging before E10 in the mouse embryo are products of the yolk sac. Blood 111, 3435–3438 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rhodes, K. E. et al. The emergence of hematopoietic stem cells is initiated in the placental vasculature in the absence of circulation. Cell Stem Cell 2, 252–263 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bertrand, J. Y. et al. Three pathways to mature macrophages in the early mouse yolk sac. Blood 106, 3004–3011 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. McGrath, K. E. et al. A transient definitive erythroid lineage with unique regulation of the β-globin locus in the mammalian embryo. Blood 117, 4600–4608 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. Chen, M. J. et al. Erythroid/myeloid progenitors and hematopoietic stem cells originate from distinct populations of endothelial cells. Cell Stem Cell 9, 541–552 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yoshimoto, M. et al. Embryonic day 9 yolk sac and intra-embryonic hemogenic endothelium independently generate a B-1 and marginal zone progenitor lacking B-2 potential. Proc. Natl Acad. Sci. USA 108, 1468–1473 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yoshimoto, M. et al. Autonomous murine T-cell progenitor production in the extra-embryonic yolk sac before HSC emergence. Blood 119, 5706–5714 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Boiers, C. et al. Lymphomyeloid contribution of an immune-restricted progenitor emerging prior to definitive hematopoietic stem cells. Cell Stem Cell 13, 535–548 (2013). Reports the identification of a yolk sac-derived HSC-independent RAG1+IL7Rα+ LMPP, providing evidence for a distinct LMPP programme and challenging the notion that lymphoid potential is always associated with definitive haematopoiesis.

    Article  PubMed  CAS  Google Scholar 

  38. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518, 547–551 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86–90 (2012). Tracks the fates of Myb−/− embryonic macrophages, showing that they are the source of the 'tissue-resident' macrophages, including Kupffer, Langerhans and microglial cells, thus providing direct evidence for a yolk sac origin of these populations.

    CAS  PubMed  Google Scholar 

  40. Hoeffel, G. et al. C-Myb+ erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 42, 665–678 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hao, Z. & Rajewsky, K. Homeostasis of peripheral B cells in the absence of B cell influx from the bone marrow. J. Exp. Med. 194, 1151–1164 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Havran, W. L. & Allison, J. P. Developmentally ordered appearance of thymocytes expressing different T-cell antigen receptors. Nature 335, 443–445 (1988).

    Article  CAS  PubMed  Google Scholar 

  43. Amit, I., Winter, D. R. & Jung, S. The role of the local environment and epigenetics in shaping macrophage identity and their effect on tissue homeostasis. Nat. Immunol. 17, 18–25 (2015).

    Article  CAS  Google Scholar 

  44. Medvinsky, A., Rybtsov, S. & Taoudi, S. Embryonic origin of the adult hematopoietic system: advances and questions. Development 138, 1017–1031 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Murray, P. D. F. The development in vitro of blood of the early chick embryo. Proc. R. Soc. Lond. B 111, 497–521 (1932).

    Article  CAS  Google Scholar 

  46. Sabin, F. Studies on the origin of blood vessels and of red blood corpuscles as seen in the living blastoderm of chicks during the second day of incubation. Contrib. Embryol. 9, 214–262 (1920).

    Google Scholar 

  47. Choi, K., Kennedy, M., Kazarov, A., Papadimitriou, J. C. & Keller, G. A common precursor for hematopoietic and endothelial cells. Development 125, 725–732 (1998). The first demonstration of the existence of the haematopoietic and endothelial bi-potential haemangioblast using the in vitro mouse ES cell differentiation model.

    Article  CAS  PubMed  Google Scholar 

  48. Huber, T. L., Kouskoff, V., Fehling, H. J., Palis, J. & Keller, G. Haemangioblast commitment is initiated in the primitive streak of the mouse embryo. Nature 432, 625–630 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Lugus, J. J., Park, C., Ma, Y. D. & Choi, K. Both primitive and definitive blood cells are derived from Flk-1+ mesoderm. Blood 113, 563–566 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zovein, A. C. et al. Fate tracing reveals the endothelial origin of hematopoietic stem cells. Cell Stem Cell 3, 625–636 (2008). The first formal demonstration that HSCs originate from an endothelial intermediate.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Jaffredo, T., Gautier, R., Brajeul, V. & Dieterlen-Lievre, F. Tracing the progeny of the aortic hemangioblast in the avian embryo. Dev. Biol. 224, 204–214 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Jaffredo, T., Gautier, R., Eichmann, A. & Dieterlen-Lievre, F. Intraaortic hemopoietic cells are derived from endothelial cells during ontogeny. Development 125, 4575–4583 (1998).

    Article  CAS  PubMed  Google Scholar 

  53. Bertrand, J. Y. et al. Haematopoietic stem cells derive directly from aortic endothelium during development. Nature 464, 108–111 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kissa, K. & Herbomel, P. Blood stem cells emerge from aortic endothelium by a novel type of cell transition. Nature 464, 112–115 (2010).

    Article  CAS  PubMed  Google Scholar 

  55. Boisset, J. C. et al. In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature 464, 116–120 (2010). References 53–55 all contributed to our understanding of the haemogenic endothelial origin of haematopoietic progenitors through the demonstration of the EHT in the ventral aspect of the dorsal aorta.

    Article  CAS  PubMed  Google Scholar 

  56. North, T. et al. Cbfa2 is required for the formation of intra-aortic hematopoietic clusters. Development 126, 2563–2575 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. North, T. E. et al. Runx1 expression marks long-term repopulating hematopoietic stem cells in the midgestation mouse embryo. Immunity 16, 661–672 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Sanchez, M. J., Holmes, A., Miles, C. & Dzierzak, E. Characterization of the first definitive hematopoietic stem cells in the AGM and liver of the mouse embryo. Immunity 5, 513–525 (1996).

    Article  CAS  PubMed  Google Scholar 

  59. Ling, K. W. et al. GATA-2 plays two functionally distinct roles during the ontogeny of hematopoietic stem cells. J. Exp. Med. 200, 871–882 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Minegishi, N. et al. The mouse GATA-2 gene is expressed in the para-aortic splanchnopleura and aorta-gonads and mesonephros region. Blood 93, 4196–4207 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Swiers, G. et al. Early dynamic fate changes in haemogenic endothelium characterized at the single-cell level. Nat. Commun. 4, 2924 (2013). Provides compelling evidence that HECs in the mouse embryo are not bi-potent progenitors.

    Article  PubMed  CAS  Google Scholar 

  62. Ciau-Uitz, A., Walmsley, M. & Patient, R. Distinct origins of adult and embryonic blood in Xenopus. Cell 102, 787–796 (2000).

    Article  CAS  PubMed  Google Scholar 

  63. Dieterlen-Lievre, F. & Martin, C. Diffuse intraembryonic hemopoiesis in normal and chimeric avian development. Dev. Biol. 88, 180–191 (1981).

    Article  CAS  PubMed  Google Scholar 

  64. Garcia-Porrero, J. A., Godin, I. E. & Dieterlen-Lievre, F. Potential intraembryonic hemogenic sites at pre-liver stages in the mouse. Anat. Embryol. 192, 425–435 (1995).

    Article  CAS  Google Scholar 

  65. Pardanaud, L., Yassine, F. & Dieterlen-Lievre, F. Relationship between vasculogenesis, angiogenesis and haemopoiesis during avian ontogeny. Development 105, 473–485 (1989).

    Article  CAS  PubMed  Google Scholar 

  66. Tavian, M. et al. Aorta-associated CD34+ hematopoietic cells in the early human embryo. Blood 87, 67–72 (1996).

    Article  CAS  PubMed  Google Scholar 

  67. Wood, H. B., May, G., Healy, L., Enver, T. & Morriss-Kay, G. M. CD34 expression patterns during early mouse development are related to modes of blood vessel formation and reveal additional sites of hematopoiesis. Blood 90, 2300–2311 (1997).

    Article  CAS  PubMed  Google Scholar 

  68. Padron-Barthe, L. et al. Clonal analysis identifies hemogenic endothelium as the source of the blood−endothelial common lineage in the mouse embryo. Blood 124, 2523–2532 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lancrin, C. et al. The haemangioblast generates haematopoietic cells through a haemogenic endothelium stage. Nature 457, 892–895 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Keller, G. Embryonic stem cell differentiation: emergence of a new era in biology and medicine. Genes Dev. 19, 1129–1155 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Ackermann, M., Liebhaber, S., Klusmann, J. H. & Lachmann, N. Lost in translation: pluripotent stem cell-derived hematopoiesis. EMBO Mol. Med. 7, 1388–1402 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kennedy, M., D'Souza, S. L., Lynch-Kattman, M., Schwantz, S. & Keller, G. Development of the hemangioblast defines the onset of hematopoiesis in human ES cell differentiation cultures. Blood 109, 2679–2687 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Zambidis, E. T. et al. Expression of angiotensin-converting enzyme (CD143) identifies and regulates primitive hemangioblasts derived from human pluripotent stem cells. Blood 112, 3601–3614 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chadwick, K. et al. Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood 102, 906–915 (2003).

    Article  CAS  PubMed  Google Scholar 

  75. Pick, M., Azzola, L., Mossman, A., Stanley, E. G. & Elefanty, A. G. Differentiation of human embryonic stem cells in serum-free medium reveals distinct roles for bone morphogenetic protein 4, vascular endothelial growth factor, stem cell factor, and fibroblast growth factor 2 in hematopoiesis. Stem Cells 25, 2206–2214 (2007).

    Article  CAS  PubMed  Google Scholar 

  76. Wang, Y. & Nakayama, N. WNT and BMP signaling are both required for hematopoietic cell development from human ES cells. Stem Cell Res. 3, 113–125 (2009).

    Article  PubMed  Google Scholar 

  77. Choi, K. D., Vodyanik, M. A. & Slukvin, I. I. Generation of mature human myelomonocytic cells through expansion and differentiation of pluripotent stem cell-derived lin-CD34+CD43+CD45+ progenitors. J. Clin. Invest. 119, 2818–2829 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zambidis, E. T., Peault, B., Park, T. S., Bunz, F. & Civin, C. I. Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development. Blood 106, 860–870 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Choi, K. D. et al. Identification of the hemogenic endothelial progenitor and its direct precursor in human pluripotent stem cell differentiation cultures. Cell Rep. 2, 553–567 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kotini, A. G. et al. Functional analysis of a chromosomal deletion associated with myelodysplastic syndromes using isogenic human induced pluripotent stem cells. Nat. Biotechnol. 33, 646–655 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Peschle, C. et al. Embryonic fetal Hb switch in humans: studies on erythroid bursts generated by embryonic progenitors from yolk sac and liver. Proc. Natl Acad. Sci. USA 81, 2416–2420 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Kennedy, M. et al. T lymphocyte potential marks the emergence of definitive hematopoietic progenitors in human pluripotent stem cell differentiation cultures. Cell Rep. 2, 1722–1735 (2012).

    Article  CAS  PubMed  Google Scholar 

  83. Timmermans, F. et al. Generation of T cells from human embryonic stem cell-derived hematopoietic zones. J. Immunol. 182, 6879–6888 (2009).

    Article  CAS  PubMed  Google Scholar 

  84. Uenishi, G. et al. Tenascin C promotes hematoendothelial development and T lymphoid commitment from human pluripotent stem cells in chemically defined conditions. Stem Cell Rep. 3, 1073–1084 (2014).

    Article  CAS  Google Scholar 

  85. Sturgeon, C. M., Ditadi, A., Awong, G., Kennedy, M. & Keller, G. Wnt signaling controls the specification of definitive and primitive hematopoiesis from human pluripotent stem cells. Nat. Biotechnol. 32, 554–561 (2014). Challenges the dogma that the definitive haematopoietic programme is specified following primitive haematopoiesis and provides evidence that they are both induced early, probably as independent mesoderm populations from the primitive streak.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Nostro, M. C., Cheng, X., Keller, G. M. & Gadue, P. Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood. Cell Stem Cell 2, 60–71 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sadler, T. W. & Langman, J. Langman's Medical Embryology 9th edn (Lippincott Williams and Wilkins, 2004).

    Google Scholar 

  88. Marvin, M. J., Di Rocco, G., Gardiner, A., Bush, S. M. & Lassar, A. B. Inhibition of Wnt activity induces heart formation from posterior mesoderm. Genes Dev. 15, 316–327 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Kinder, S. J. et al. The orderly allocation of mesodermal cells to the extraembryonic structures and the anteroposterior axis during gastrulation of the mouse embryo. Development 126, 4691–4701 (1999).

    Article  CAS  PubMed  Google Scholar 

  90. Schoenwolf, G. C., Garcia-Martinez, V. & Dias, M. S. Mesoderm movement and fate during avian gastrulation and neurulation. Dev. Dyn. 193, 235–248 (1992).

    Article  CAS  PubMed  Google Scholar 

  91. Ditadi, A. et al. Human definitive haemogenic endothelium and arterial vascular endothelium represent distinct lineages. Nat. Cell Biol. 17, 580–591 (2015). Provides compelling evidence, using the hPSC-differentiation model, that HECs are haematopoietic-restricted and distinct from vascular endothelium.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Bertrand, J. Y., Cisson, J. L., Stachura, D. L. & Traver, D. Notch signaling distinguishes 2 waves of definitive hematopoiesis in the zebrafish embryo. Blood 115, 2777–2783 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Hadland, B. K. et al. A requirement for Notch1 distinguishes 2 phases of definitive hematopoiesis during development. Blood 104, 3097–3105 (2004). The first formal demonstration that Notch signalling is required cell autonomously for HSC specification.

    Article  CAS  PubMed  Google Scholar 

  94. Gama-Norton, L. et al. Notch signal strength controls cell fate in the haemogenic endothelium. Nat. Commun. 6, 8510 (2015).

    Article  CAS  PubMed  Google Scholar 

  95. Kim, A. D. et al. Discrete Notch signaling requirements in the specification of hematopoietic stem cells. EMBO J. 33, 2363–2373 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Clements, W. K. et al. A somitic Wnt16/Notch pathway specifies haematopoietic stem cells. Nature 474, 220–224 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Kobayashi, I. et al. Jam1a–Jam2a interactions regulate haematopoietic stem cell fate through Notch signalling. Nature 512, 319–323 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Nguyen, P. D. et al. Haematopoietic stem cell induction by somite-derived endothelial cells controlled by meox1. Nature 512, 314–318 (2014).

    Article  CAS  PubMed  Google Scholar 

  99. Samokhvalov, I. M., Samokhvalova, N. I. & Nishikawa, S. Cell tracing shows the contribution of the yolk sac to adult haematopoiesis. Nature 446, 1056–1061 (2007). Challenges the dogma that the definitive haematopoietic programme is specified after primitive haematopoiesis.

    Article  CAS  PubMed  Google Scholar 

  100. Tanaka, Y. et al. Early ontogenic origin of the hematopoietic stem cell lineage. Proc. Natl Acad. Sci. USA 109, 4515–4520 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Tanaka, Y. et al. Circulation-independent differentiation pathway from extraembryonic mesoderm toward hematopoietic stem cells via hemogenic angioblasts. Cell Rep. 8, 31–39 (2014).

    Article  CAS  PubMed  Google Scholar 

  102. Lacaud, G., Kouskoff, V., Trumble, A., Schwantz, S. & Keller, G. Haploinsufficiency of Runx1 results in the acceleration of mesodermal development and hemangioblast specification upon in vitro differentiation of ES cells. Blood 103, 886–889 (2004).

    Article  CAS  PubMed  Google Scholar 

  103. Gori, J. L. et al. Vascular niche promotes hematopoietic multipotent progenitor formation from pluripotent stem cells. J. Clin. Invest. 125, 1243–1254 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  104. Ledran, M. H. et al. Efficient hematopoietic differentiation of human embryonic stem cells on stromal cells derived from hematopoietic niches. Cell Stem Cell 3, 85–98 (2008).

    Article  CAS  PubMed  Google Scholar 

  105. Lu, M., Kardel, M. D., O'Connor, M. D. & Eaves, C. J. Enhanced generation of hematopoietic cells from human hepatocarcinoma cell-stimulated human embryonic and induced pluripotent stem cells. Exp. Hematol. 37, 924–936 (2009).

    Article  CAS  PubMed  Google Scholar 

  106. Wang, L. et al. Generation of hematopoietic repopulating cells from human embryonic stem cells independent of ectopic HOXB4 expression. J. Exp. Med. 201, 1603–1614 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Amabile, G. et al. In vivo generation of transplantable human hematopoietic cells from induced pluripotent stem cells. Blood 121, 1255–1264 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Suzuki, N. et al. Generation of engraftable hematopoietic stem cells from induced pluripotent stem cells by way of teratoma formation. Mol. Ther. 21, 1424–1431 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Kyba, M., Perlingeiro, R. C. & Daley, G. Q. HoxB4 confers definitive lymphoid-myeloid engraftment potential on embryonic stem cell and yolk sac hematopoietic progenitors. Cell 109, 29–37 (2002).

    Article  CAS  PubMed  Google Scholar 

  110. Lengerke, C. et al. The cdx-hox pathway in hematopoietic stem cell formation from embryonic stem cells. Ann. NY Acad. Sci. 1106, 197–208 (2007).

    Article  CAS  PubMed  Google Scholar 

  111. Pearson, S., Cuvertino, S., Fleury, M., Lacaud, G. & Kouskoff, V. In vivo repopulating activity emerges at the onset of hematopoietic specification during embryonic stem cell differentiation. Stem Cell Rep. 4, 431–444 (2015).

    Article  CAS  Google Scholar 

  112. Sandler, V. M. et al. Reprogramming human endothelial cells to haematopoietic cells requires vascular induction. Nature 511, 312–318 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Riddell, J. et al. Reprogramming committed murine blood cells to induced hematopoietic stem cells with defined factors. Cell 157, 549–564 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. 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 

  115. McKinney-Freeman, S. et al. The transcriptional landscape of hematopoietic stem cell ontogeny. Cell Stem Cell 11, 701–714 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Lizama, C. O. et al. Repression of arterial genes in hemogenic endothelium is sufficient for haematopoietic fate acquisition. Nat. Commun. 6, 7739 (2015).

    Article  PubMed  Google Scholar 

  117. Thambyrajah, R. et al. GFI1 proteins orchestrate the emergence of haematopoietic stem cells through recruitment of LSD1. Nat. Cell Biol. 18, 21–32 (2015).

    Article  PubMed  CAS  Google Scholar 

  118. 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 

  119. Solaimani Kartalaei, P. et al. Whole-transcriptome analysis of endothelial to hematopoietic stem cell transition reveals a requirement for Gpr56 in HSC generation. J. Exp. Med. 212, 93–106 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Souilhol, C. et al. Inductive interactions mediated by interplay of asymmetric signalling underlie development of adult haematopoietic stem cells. Nat. Commun. 7, 10784 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Ruiz-Herguido, C. et al. Hematopoietic stem cell development requires transient Wnt/β-catenin activity. J. Exp. Med. 209, 1457–1468 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Goessling, W. et al. Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell 136, 1136–1147 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Chanda, B., Ditadi, A., Iscove, N. N. & Keller, G. Retinoic acid signaling is essential for embryonic hematopoietic stem cell development. Cell 155, 215–227 (2013).

    Article  CAS  PubMed  Google Scholar 

  124. Boitano, A. E. et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science 329, 1345–1348 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Fares, I. et al. Cord blood expansion. Pyrimidoindole derivatives are agonists of human hematopoietic stem cell self-renewal. Science 345, 1509–1512 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Genovese, P. et al. Targeted genome editing in human repopulating haematopoietic stem cells. Nature 510, 235–240 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Huang, J., Nguyen-McCarty, M., Hexner, E. O., Danet-Desnoyers, G. & Klein, P. S. Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nat. Med. 18, 1778–1785 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Mantel, C. R. et al. Enhancing hematopoietic stem cell transplantation efficacy by mitigating oxygen shock. Cell 161, 1553–1565 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. North, T. E. et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 447, 1007–1011 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Wong, P. M., Chung, S. W., Reicheld, S. M. & Chui, D. H. Hemoglobin switching during murine embryonic development: evidence for two populations of embryonic erythropoietic progenitor cells. Blood 67, 716–721 (1986).

    Article  CAS  PubMed  Google Scholar 

  131. Yi, Z. et al. Sox6 directly silences epsilon globin expression in definitive erythropoiesis. PLoS Genet. 2, e14 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Wood, W. G. Haemoglobin synthesis during human fetal development. Br. Med. Bull. 32, 282–287 (1976).

    Article  CAS  PubMed  Google Scholar 

  133. Robert-Moreno, A., Espinosa, L., de la Pompa, J. L. & Bigas, A. RBPjκ-dependent Notch function regulates Gata2 and is essential for the formation of intra-embryonic hematopoietic cells. Development 132, 1117–1126 (2005).

    Article  CAS  PubMed  Google Scholar 

  134. Lacaud, G. et al. Runx1 is essential for hematopoietic commitment at the hemangioblast stage of development in vitro. Blood 100, 458–466 (2002).

    Article  CAS  PubMed  Google Scholar 

  135. Mucenski, M. L. et al. A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis. Cell 65, 677–689 (1991).

    Article  CAS  PubMed  Google Scholar 

  136. Wang, Q. et al. Disruption of the Cbfa2 gene causes necrosis and hemorrhaging in the central nervous system and blocks definitive hematopoiesis. Proc. Natl Acad. Sci. USA 93, 3444–3449 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Bee, T. et al. Alternative Runx1 promoter usage in mouse developmental hematopoiesis. Blood Cells Mol. Dis. 43, 35–42 (2009).

    Article  CAS  PubMed  Google Scholar 

  138. Bee, T. et al. Nonredundant roles for Runx1 alternative promoters reflect their activity at discrete stages of developmental hematopoiesis. Blood 115, 3042–3050 (2010).

    Article  CAS  PubMed  Google Scholar 

  139. Clarke, D. et al. In vitro differentiation of c-myb−/− ES cells reveals that the colony forming capacity of unilineage macrophage precursors and myeloid progenitor commitment are c-Myb independent. Oncogene 19, 3343–3351 (2000).

    Article  CAS  PubMed  Google Scholar 

  140. Soza-Ried, C., Hess, I., Netuschil, N., Schorpp, M. & Boehm, T. Essential role of c-myb in definitive hematopoiesis is evolutionarily conserved. Proc. Natl Acad. Sci. USA 107, 17304–17308 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Tober, J., McGrath, K. E. & Palis, J. Primitive erythropoiesis and megakaryopoiesis in the yolk sac are independent of c-myb. Blood 111, 2636–2639 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The referenced work from the Keller lab was supported by the US National Institutes of Health grants U01 HL100395 and 5R01HL091724 and by the Canadian Institutes of Health Research grant MOP126117.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gordon Keller.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Induced pluripotent stem cells

(iPSCs). Pluripotent stem cells generated from adult somatic cells through genetic reprogramming with transcription factors. Similar to human embryonic stem cells, iPSCs can be propagated indefinitely in culture while retaining the capacity to differentiate and give rise to virtually all cell types found in the human body. iPSCs should theoretically be an identical immunological match to the patient they are derived from.

CRISPR–Cas9 technology

The enzyme Cas9 causes a DNA double-strand break at a site (or sites) within the genome that harbours complementarity to homologous short RNA sequences that can be easily synthesized and delivered to cells in vitro. This permits easy, cost-effective generation of in-frame deletions, or insertional mutagenesis, within human pluripotent stem cells.

Yolk sac

An extra-embryonic developmental tissue that harbours the first site of haematopoietic development.

Embryo proper

The anatomical structure that ultimately gives rise to the mature embryo and all tissues found in fetal and adult life. It does not include the extra-embryonic annexes.

Aorta–gonad–mesonephros

(AGM). A region in the caudal portion of the embryo containing the developing aorta, genital ridges and mesonephros that gives rise to haematopoietic stem cells.

OP9 stromal cells

A stromal cell line derived from the op/op mouse that does not produce macrophage colony-stimulating factor (M-CSF). These cells are commonly used to support differentiation and expansion of mouse and human haematopoietic progenitor populations.

Ontogeny

The development of a specific cell type within an organism.

B-1 cells

A subclass of B lymphocytes that are not part of the adaptive immune system and lack the ability to form memory B cells.

γδ T cells

A subset of T cells that have a distinct T cell receptor composed of a γ- and δ-chain.

Primitive streak

A dorsally located structure that forms in the epiblast of the early vertebrate embryo. The primitive streak establishes bilateral symmetry and is commonly associated with the onset of gastrulation and the generation of the three germ layers. Undifferentiated epiblast cells adopt either a mesoderm or an endoderm fate as they move through the primitive streak.

Viral marking

The use of a virus that inserts randomly into the genome as a means of tracking the progression of a cell within a population of other cells.

Somites

An embryonic tissue of paraxial mesoderm found in segments along the head-to-tail axis of the developing embryo. Somites ultimately give rise to the vertebrae, rib cage and occipital bone, as well as to cartilage, tendons and muscle.

Teratoma

A tumour that possesses tissue components derived from more than one germ layer.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ditadi, A., Sturgeon, C. & Keller, G. A view of human haematopoietic development from the Petri dish. Nat Rev Mol Cell Biol 18, 56–67 (2017). https://doi.org/10.1038/nrm.2016.127

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm.2016.127

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing