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:

Decoding human fetal liver haematopoiesis

Abstract

Definitive haematopoiesis in the fetal liver supports self-renewal and differentiation of haematopoietic stem cells and multipotent progenitors (HSC/MPPs) but remains poorly defined in humans. Here, using single-cell transcriptome profiling of approximately 140,000 liver and 74,000 skin, kidney and yolk sac cells, we identify the repertoire of human blood and immune cells during development. We infer differentiation trajectories from HSC/MPPs and evaluate the influence of the tissue microenvironment on blood and immune cell development. We reveal physiological erythropoiesis in fetal skin and the presence of mast cells, natural killer and innate lymphoid cell precursors in the yolk sac. We demonstrate a shift in the haemopoietic composition of fetal liver during gestation away from being predominantly erythroid, accompanied by a parallel change in differentiation potential of HSC/MPPs, which we functionally validate. Our integrated map of fetal liver haematopoiesis provides a blueprint for the study of paediatric blood and immune disorders, and a reference for harnessing the therapeutic potential of HSC/MPPs.

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

Fig. 1: Single-cell transcriptome map of fetal liver.
Fig. 2: Multi-modal and spatial validation of cell types.
Fig. 3: Fetal liver and NLT haematopoiesis.
Fig. 4: Lymphoid lineages in fetal liver and NLT.
Fig. 5: Tissue signatures in developing myeloid cells.
Fig. 6: HSC/MPP differentiation potential by gestation.

Similar content being viewed by others

Data availability

The raw sequencing data, expression count data with cell classifications are deposited at ArrayExpress with accession code E-MTAB-7407.

Code availability

All scripts are available at https://github.com/haniffalab/FCA_liver.

References

  1. Jagannathan-Bogdan, M. & Zon, L. I. Hematopoiesis. Development 140, 2463–2467 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Parekh, C. & Crooks, G. M. Critical differences in hematopoiesis and lymphoid development between humans and mice. J. Clin. Immunol. 33, 711–715 (2013).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  4. Holt, P. G. & Jones, C. A. The development of the immune system during pregnancy and early life. Allergy 55, 688–697 (2000).

    CAS  PubMed  Google Scholar 

  5. Kashem, S. W., Haniffa, M. & Kaplan, D. H. Antigen-presenting cells in the skin. Annu. Rev. Immunol. 35, 469–499 (2017).

    CAS  PubMed  Google Scholar 

  6. Mass, E. et al. Specification of tissue-resident macrophages during organogenesis. Science 353, aaf4238 (2016).

    PubMed  PubMed Central  Google Scholar 

  7. Korsunsky, I. et al. Fast, sensitive, and accurate integration of single cell data with Harmony. Preprint at bioRxiv https://doi.org/10.1101/461954 (2018).

  8. Ohls, R. K. et al. Neutrophil pool sizes and granulocyte colony-stimulating factor production in human mid-trimester fetuses. Pediatr. Res. 37, 806–811 (1995).

    CAS  PubMed  Google Scholar 

  9. Picard, C. et al. International Union of Immunological Societies: 2017 Primary Immunodeficiency Diseases Committee Report on Inborn Errors of Immunity. J. Clin. Immunol. 38, 96–128 (2018).

    PubMed  Google Scholar 

  10. Gentek, R. et al. Hemogenic endothelial fate mapping reveals dual developmental origin of mast cells. Immunity 48, 1160–1171 (2018).

    CAS  PubMed  Google Scholar 

  11. Iskander, D. et al. Elucidation of the EP defect in Diamond–Blackfan anemia by characterization and prospective isolation of human EPs. Blood 125, 2553–2557 (2015).

    CAS  PubMed  Google Scholar 

  12. MacParland, S. A. et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 9, 4383 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  13. An, X. et al. Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood 123, 3466–3477 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Gautier, E.-F. et al. Comprehensive proteomic analysis of human erythropoiesis. Cell Rep. 16, 1470–1484 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Dedhia, P., Kambayashi, T. & Pear, W. S. Notch2 paves the way to mast cells by Hes1 and Gata3. Proc. Natl Acad. Sci. USA 105, 7629–7630 (2008).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Okada, Y. et al. Homeodomain proteins MEIS1 and PBXs regulate the lineage-specific transcription of the platelet factor 4 gene. Blood 101, 4748–4756 (2003).

    CAS  PubMed  Google Scholar 

  17. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal–fetal interface in humans. Nature 563, 347–353 (2018).

    ADS  CAS  PubMed  Google Scholar 

  18. Klei, T. R. L., Meinderts, S. M., van den Berg, T. K. & van Bruggen, R. From the cradle to the grave: the role of macrophages in erythropoiesis and erythrophagocytosis. Front. Immunol. 8, 73 (2017).

    PubMed  PubMed Central  Google Scholar 

  19. Kessel, K. U. et al. Emergence of CD43-expressing hematopoietic progenitors from human induced pluripotent stem cells. Transfus. Med. Hemother. 44, 143–150 (2017).

    PubMed  PubMed Central  Google Scholar 

  20. Li, W. et al. Identification and transcriptome analysis of erythroblastic island macrophages. Blood 134, 480–491 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Ebert, B. L. et al. An erythroid differentiation signature predicts response to lenalidomide in myelodysplastic syndrome. PLoS Med. 5, e35 (2008).

    PubMed  PubMed Central  Google Scholar 

  22. Gale, R. P. in Fetal Liver Transplantation (eds Touraine, J.-L., Gale, R. P. & Kochupillai, V.) 45–56 (Springer, 1987).

  23. Phillips, J. H. et al. Ontogeny of human natural killer (NK) cells: fetal NK cells mediate cytolytic function and express cytoplasmic CD3ε,δ proteins. J. Exp. Med. 175, 1055–1066 (1992).

    CAS  PubMed  Google Scholar 

  24. Forkel, M. et al. Composition and functionality of the intrahepatic innate lymphoid cell-compartment in human nonfibrotic and fibrotic livers. Eur. J. Immunol. 47, 1280–1294 (2017).

    CAS  PubMed  Google Scholar 

  25. Haynes, B. F. & Heinly, C. S. Early human T cell development: analysis of the human thymus at the time of initial entry of hematopoietic stem cells into the fetal thymic microenvironment. J. Exp. Med. 181, 1445–1458 (1995).

    CAS  PubMed  Google Scholar 

  26. Sánchez, M. J., Spits, H., Lanier, L. L. & Phillips, J. H. Human natural killer cell committed thymocytes and their relation to the T cell lineage. J. Exp. Med. 178, 1857–1866 (1993).

    PubMed  Google Scholar 

  27. Darrasse-Jèze, G., Marodon, G., Salomon, B. L., Catala, M. & Klatzmann, D. Ontogeny of CD4+CD25+ regulatory/suppressor T cells in human fetuses. Blood 105, 4715–4721 (2005).

    PubMed  Google Scholar 

  28. Wucherpfennig, K. W. et al. Structural requirements for binding of an immunodominant myelin basic protein peptide to DR2 isotypes and for its recognition by human T cell clones. J. Exp. Med. 179, 279–290 (1994).

    CAS  PubMed  Google Scholar 

  29. Spits, H. et al. Innate lymphoid cells—a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    CAS  PubMed  Google Scholar 

  30. Chen, L. et al. CD56 expression marks human group 2 innate lymphoid cell divergence from a shared NK cell and group 3 innate lymphoid cell developmental pathway. Immunity 49, 464–476 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Roy, A. et al. Perturbation of fetal liver hematopoietic stem and progenitor cell development by trisomy 21. Proc. Natl Acad. Sci. USA 109, 17579–17584 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Almaden, J. V. et al. B-cell survival and development controlled by the coordination of NF-κB family members RelB and cRel. Blood 127, 1276–1286 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Stremmel, C. et al. Yolk sac macrophage progenitors traffic to the embryo during defined stages of development. Nat. Commun. 9, 75 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).

    CAS  PubMed  Google Scholar 

  35. Murphy, T. L. et al. Transcriptional control of dendritic cell development. Annu. Rev. Immunol. 34, 93–119 (2016).

    CAS  PubMed  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rodrigues, P. F. et al. Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells. Nat. Immunol. 19, 711–722 (2018).

    CAS  PubMed  Google Scholar 

  38. Seu, K. G. et al. Unraveling macrophage heterogeneity in erythroblastic islands. Front. Immunol. 8, 1140 (2017).

    PubMed  PubMed Central  Google Scholar 

  39. McGovern, N. et al. Human dermal CD14+ cells are a transient population of monocyte-derived macrophages. Immunity 41, 465–477 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sun, J.-B. et al. Deficiency in calcium-binding protein S100A4 impairs the adjuvant action of cholera toxin. Front. Immunol. 8, 1119 (2017).

    PubMed  PubMed Central  Google Scholar 

  41. Janelsins, B. M., Lu, M. & Datta, S. K. Altered inactivation of commensal LPS due to acyloxyacyl hydrolase deficiency in colonic dendritic cells impairs mucosal Th17 immunity. Proc. Natl Acad. Sci. USA 111, 373–378 (2014).

    ADS  CAS  PubMed  Google Scholar 

  42. McGovern, N. et al. Human fetal dendritic cells promote prenatal T-cell immune suppression through arginase-2. Nature 546, 662–666 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Grün, D. et al. De novo prediction of stem cell identity using single-cell transcriptome data. Cell Stem Cell 19, 266–277 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Belluschi, S. et al. Myelo-lymphoid lineage restriction occurs in the human haematopoietic stem cell compartment before lymphoid-primed multipotent progenitors. Nat. Commun. 9, 4100 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  46. Cabezas-Wallscheid, N. et al. Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell 15, 507–522 (2014).

    CAS  PubMed  Google Scholar 

  47. Pietras, E. M. et al. Functionally distinct subsets of lineage-biased multipotent progenitors control blood production in normal and regenerative conditions. Cell Stem Cell 17, 35–46 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. 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).

    CAS  PubMed  Google Scholar 

  49. Laurenti, E. et al. The transcriptional architecture of early human hematopoiesis identifies multilevel control of lymphoid commitment. Nat. Immunol. 14, 756 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Morrison, S. J., Hemmati, H. D., Wandycz, A. M. & Weissman, I. L. The purification and characterization of fetal liver hematopoietic stem cells. Proc. Natl Acad. Sci. USA 92, 10302–10306 (1995).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Bowie, M. B. et al. Hematopoietic stem cells proliferate until after birth and show a reversible phase-specific engraftment defect. J. Clin. Invest. 116, 2808–2816 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. 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).

    CAS  PubMed  Google Scholar 

  53. Gerrelli, D., Lisgo, S., Copp, A. J. & Lindsay, S. Enabling research with human embryonic and fetal tissue resources. Development 142, 3073–3076 (2015).

    CAS  PubMed  Google Scholar 

  54. Bullen, P. & Wilson, D. in Molecular Genetics of Early Human Development (eds Strachan, T. et al.) 27–35 (Bios Scientific, 1997).

  55. Hern, W. M. Correlation of fetal age and measurements between 10 and 26 weeks of gestation. Obstet. Gynecol. 63, 26–32 (1984).

    CAS  PubMed  Google Scholar 

  56. Roy, A. et al. High resolution IgH repertoire analysis reveals fetal liver as the likely origin of life-long, innate B lymphopoiesis in humans. Clin. Immunol. 183, 8–16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Villani, A.-C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

    PubMed  PubMed Central  Google Scholar 

  58. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. 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).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hu, H. et al. AnimalTFDB 3.0: a comprehensive resource for annotation and prediction of animal transcription factors. Nucleic Acids Res. 47, D33–D38 (2019).

    CAS  PubMed  Google Scholar 

  62. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB v2.0: Inferring cell-cell communication from combined expression of multi-subunit receptor-ligand complexes. Preprint at bioRxiv https://doi.org/10.1101/680926 (2019).

  63. The 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Google Scholar 

  64. Belle, M. et al. Tridimensional visualization and analysis of early human development. Cell 169, 161–173 (2017).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This publication is part of the Human Cell Atlas (www.humancellatlas.org/publications). We thank the Newcastle University Flow Cytometry Core Facility, Bioimaging Core Facility, Genomics Facility, NUIT for technical assistance, School of Computing for access to the High-Performance Computing Cluster and Newcastle Molecular Pathology Node Proximity Laboratory; A. Farnworth for clinical liaison, S. Hambleton for primary immunodeficiency expertise, H. Chen for immunohistochemistry assistance and M. Belle and S. Fouquet for light-sheet fluorescence microscopy assistance. The human embryonic and fetal material was provided by the Joint MRC–Wellcome (MR/R006237/1) Human Developmental Biology Resource (www.hdbr.org). We acknowledge funding from the Wellcome Human Cell Atlas Strategic Science Support (WT211276/Z/18/Z); M.H. is funded by Wellcome (WT107931/Z/15/Z), The Lister Institute for Preventive Medicine and NIHR and Newcastle Biomedical Research Centre; S.A.T. is funded by Wellcome (WT206194), ERC Consolidator and EU MRG-Grammar awards; S.B. is funded by Wellcome (WT110104/Z/15/Z) and St. Baldrick’s Foundation; E.L. is funded by a Wellcome Sir Henry Dale and Royal Society Fellowship, European Haematology Association, Wellcome and MRC to the Wellcome–MRC Cambridge Stem Cell Institute and BBSRC; A. Regev is funded by the Manton Foundation and Klarman Cell Observatory.

Author information

Authors and Affiliations

Authors

Contributions

M.H., S.A.T. and E.L. conceived and directed the study. M.H., S.A.T., E.L. and R.A.B. designed the experiments. Samples were collected by S. Lisgo and S. Lindsay, isolated by R.A.B. and libraries prepared by E.S., L.M., D.-M.P., R.V.-T., J.-E.P. and J.F. Flow cytometry and FACS experiments were performed by R.A.B., E.F.C., L.J., D. Maunder. and A. Filby. Imaging mass cytometry experiments were performed by M.A., B.M., B.I., D. McDonald and A. Fuller. Cytospins were performed by D.D. and J.F. and in vitro culture differentiation experiments were performed by L.J., D. Maunder. and E.F.C. Immunohistochemistry was performed by B.I., M.A., F.G. and C.M.; P.C. and M.A. interpreted immunohistochemistry and developmental pathology sections. Y.G. and A.C. performed and interpreted light-sheet fluorescence microscopy experiments. M.S.K., B.L., O.A., M.T., D.D., T.L.T., M.S., O.R.-R. and A. Regev. generated adult and cord blood scRNA-seq datasets. D.-M.P., K.G., K.P., S.W., I.G., M.E., P.V., M.D.Y., Z.M. and J.B performed the computational analysis. M.H., D.-M.P., R.A.B., B.G., E.L, I.R., A. Roy, E.F.C., L.J., A.-C.V., R.R., E.P., M.M., J.-E.P., G.R., K.B.M., M.J.T.S., A. Filby., K.G., S.W., I.G., S.B. and J.B. interpreted the data. M.H., L.J., R.A.B., E.S., D.-M.P., B.G., E.L., I.R., K.G., S.W., I.G., A.-C.V. and A. Roy. wrote the manuscript. All authors read and accepted the manuscript.

Corresponding authors

Correspondence to Sam Behjati, Elisa Laurenti, Sarah A. Teichmann or Muzlifah Haniffa.

Ethics declarations

Competing interests

A. Regev is a founder and equity holder of Celsius Therapeutics and an SAB member of Neogene Therapeutics, ThermoFisher Scientific and Syros Pharmaceuticals.

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 Single-cell transcriptome map of fetal liver.

a, Fetal skin and kidney haematopoietic cells visualized by UMAP. Colours indicate cell state. Inset, colours indicate tissue type. b, UMAP visualization of yolk sac haematopoietic cells. Colours indicate cell state. Inset, colours indicate location within yolk sac. c, UMAP visualization of 3′ liver 10x cells after batch correction, coloured by sample. d, UMAP visualization (top) of 3′ 10x liver sample sex mixing grouped by developmental stage, and violin plots (bottom) showing ln-normalized median expression of XIST (green) and RSP4Y1 (purple), which mark female and male samples, respectively. e, UMAP visualization of fetal liver composition by developmental stage. Colours indicate cell state. f, UMAP visualization of fetal liver cells profiled using Smart-seq2. Colours indicate cell states as shown in e. g, Frequency (mean ± s.e.m.) of B cells in the CD34 cells detected in 6–19 PCW fetal livers by flow cytometry (*P < 0.05, ***P = 0.003 and ****P < 0.001).

Source data

Extended Data Fig. 2 Transcriptome validation of fetal liver cells.

a, Assessment of 48 genes from the 4,471 highly variable genes by using a random forest classifier to assign cell labels, where ‘true cell label’ indicates the manual annotation based on the full list of variable genes. b, Comparison of representative mini bulk RNA-seq data (in coloured triangles) and liver EI populations (early, mid and late erythroids, VCAM1+ EI macrophages), Kupffer cells and endothelium validated by Smart-seq2 (SS2) (in colour) overlaid on whole liver SS2 populations (grey). c, Dot plot showing representative median-scaled ln-normalized gene expression of 100 FACS-isolated liver cells based on marker gene expression in Fig. 2a. Gene expression indicated by spot size and colour intensity. d, Dot plot showing median-scaled ln-normalized gene expression of FACS-sorted single cells from liver EI populations (early, mid and late erythroids, VCAM1+ EI macrophages), Kupffer cells and endothelium shown as coloured dots in b based on marker gene expression in Fig. 2a. Gene-expression frequency (per cent of cells within cell type expressing the gene) indicated by spot size and expression level by colour intensity. e, Overlay pseudo-colour Hyperion representative images for 8 PCW and 15 PCW fetal liver. Far left images are shown at 5× magnification with zoom of insets on right at 20× magnification (1 μm per pixel). Asterisks indicate bile ducts.

Extended Data Fig. 3 Fetal liver and NLT haematopoiesis.

a, PAGA analysis of fetal liver HSC/MPP, erythroid, megakaryocyte and mast cell lineages from Fig. 3a. Lines show connections; line thickness corresponds to the level of connectivity (low (thin) to high (thick) PAGA connectivity). b, Heat map showing min − max normalized expression of statistically significant (P < 0.001), dynamically variable genes from pseudotime analysis for erythroid, megakaryocyte and mast cell inferred trajectories. Transcription factors in bold, asterisks mark genes not previously implicated for the respective lineages. c, FDG visualization of fetal liver, skin and kidney HSC/MPP, MEMP, erythroid, megakaryocyte and mast cell lineages. d, Heat map showing the scaled ln-expression of selected marker genes in fetal liver, NLT and yolk sac subsets. e, PAGA connectivity scores of HSC/MPP, erythroid, megakaryocyte and mast cell lineages between fetal liver, skin, kidney (K) and yolk sac. f, Representative immunohistochemical staining of sequential sections of 8 PCW fetal skin for endothelium (CD34+) and erythroblasts (nucleated and GYPA+), nuclei stained with blue alkaline phosphatase. Zoom in of insets (right) bordered with black (top) indicate nucleated cells stained positive for GYPA within CD34+ blood vessels, and those bordered with red (bottom) indicate nucleated GYPA+ cells outside CD34+ blood vessels. Scale bars, 100 μm. g, Representative confocal fluorescence microscopy of embryo (5 PCW) hand skin. Scale bar, 10 μm; red, TO-PRO-3 nuclei; green, GYPA (see also Supplementary Video 2 showing light-sheet fluorescence microscopy). The arrowhead indicates extravascular nucleated erythroid cells. h, Stacked bar plots (left) showing percentage (mean ± s.d.) of fetal liver (red), skin (blue) and kidney (green) HSC/MPP, MEMP, erythroid, megakaryocyte and mast cells in each stage of the cell cycle (G1 (navy), G2M (blue) and S (white) phase), and ln-normalized median expression of MKI67 transcript (right) in corresponding liver vs NLT cell types (total percent of MKI67-expressing cells shown above plots; each dot represents a single cell). *P < 0.05, **P < 0.01 and ***P < 0.005.

Source data

Extended Data Fig. 4 Investigation of interactions between fetal liver macrophages and erythroid cells.

a, Representative immunohistochemical staining of fetal liver for erythroblasts and macrophages with GYPA and CD68, respectively. Scale bar, 50 μm. Statistically significantly (P < 0.05) enriched receptor–ligand interactions from CellPhoneDB between VCAM1+ EI macrophages (purple) and two erythroid populations (early and mid; red) (n = 14 biologically independent samples). Asterisks indicate protein complexes. Violin plots show ln-normalized median gene-expression value of VCAM1 and ITGA4 in cells analysed by CellPhoneDB (indicated by # in dot plot). b, Representative immunohistochemical staining of sequential sections of 8 PCW fetal liver for VCAM1+ EI macrophages (VCAM1+) and CD49d+GYPA+ cells with nuclei stained using blue alkaline phosphatase. Right, zoom in of insets, with coloured arrows indicating erythroblast (yellow) and VCAM1+ EI macrophage (purple). Scale bars, 100 μm. c, Representative gating strategy used to visualize fetal liver erythroid cells, VCAM1+ EI macrophages, Kupffer cells, monocyte-macrophages and mast cells. d, Bright field (BF), VCAM1 (CD106), CD34, CD45, KIT (CD117), GYPA, CD14 and HLA-DR images for each cell type within gates shown in c. e, Representative bright-field images of cells found within the single-cell and doublet gates. f, Bar plots showing the proportion of each cell type within the single-cell gate (white) or doublet gate (grey) (mean ± s.d.); *P = 0.0194. g, Comparison of macrophage and erythroid gene expression in mouse macrophages (red) and EI macrophages (blue), n = 3 from ref. 20.

Source data

Extended Data Fig 5 Lymphoid lineages in fetal liver and NLT.

a, PAGA analysis of fetal liver HSC/MPP and lymphoid cell types from Fig. 1b showing changes over four developmental stages. Lines symbolize connection; line thickness corresponds to the level of connectivity (low (thin) to high (thick) PAGA connectivity). b, c, Feature plots (b) and violin plots (c) showing ln-normalized median expression of selected known NK, ILC and T cell genes over gestation for early lymphoid/T lymphocyte cluster; **P < 0.001, ***P < 0.005 and ****P < 0.0001. d, Dot plot showing median-scaled ln-normalized median expression of V(D)J transcripts in fetal liver lymphoid cell types. Gene expression indicated by spot size and colour intensity. e, Heat map showing normalized expression of statistically significant, dynamically variable genes from pseudotime analysis for B cell lineage inferred trajectory (likelihood ratio test). Transcription factors are in bold. Morphology of liver pro-B and pre-B cells and B cells by Giemsa stain after cytospin. f, ln-normalized expression (mean ± s.e.m.) of TNFSF13B in Kupffer cells and NFKBIA in HSC/MPPs and cells in the B cell lineage across four developmental stages spanning 6–17 PCW; trend lines showing linear regression. g, PAGA connectivity scores of HSC/MPP and lymphoid cells from fetal liver, skin, kidney and yolk sac. h, ln-normalized median expression of selected known NK (top) and ILC precursor (bottom) marker genes and selected DEGs between liver (red), skin (blue) and kidney (green) visualized by violin plots (***P < 0.005 and ****P < 0.001). i, Violin plots showing ln-normalized median expression of selected known ILC and NK cell genes expressed in ILC precursors from fetal liver, skin and kidney.

Source data

Extended Data Fig. 6 Tissue signatures in developing myeloid cells.

a, Diffusion map of fetal liver HSC/MPP, progenitors and precursors from Fig. 1b. b, Heat map showing min − max normalized expression (P < 0.001) of dynamically variable genes from pseudotime analysis for monocyte, DC1 and DC2 inferred trajectories. Transcription factors in bold, asterisks mark genes not previously implicated for the respective lineages. c, Heat map visualization comparing scaled expression of the top marker genes of decidua–placenta (red), fetal liver (black) and yolk sac (purple) progenitor and myeloid populations. d, PAGA connectivity scores of HSC/MPP and myeloid cells from fetal liver, skin and kidney. e, ln-normalized median expression of three known marker genes and three DEGs in corresponding myeloid populations across fetal liver, skin and kidney visualized by violin plots (*P < 0.05, ***P < 0.005 and ****P < 0.0001).

Extended Data Fig. 7 HSC/MPP differentiation potential by gestation.

a, Experimental design for single-cell transcriptome and culture of fetal liver cells from the representative FACS gates illustrated. b, Alignment of 349 scRNA-seq-profiled cells from FACS gates in a with 10x-profiled HSC/MPPs and early progenitors visualized using FDG; point shape corresponds to sequencing type (triangle, SS2 plate data; circle, 10x data). c, Stacked bar plot of all different types of colonies generated by single HSC pool gate cells (gate defined in a). d, Stacked bar plot of all different types of colonies generated by single HSC pool gate cells without MS5 stroma layer (gate defined in a) by stage (left) and in individual samples (right), *P < 0.05. e, Percentage of colonies generated by single HSC pool cells without MS5 stroma layer containing erythroid cells (sum of ery, ery–meg, ery–meg–my and ery–my colonies shown in c); **P < 0.01. f, Percentage of colonies from single-cell culture (shown in Fig. 6c) that differentiated along three lineage (defined as sum of ery, NK and my, and ery, meg and my colonies) branches (***P < 0.005). g, Percentage of colonies containing NK cells following B/NK optimized culture of ten cells from the HSC pool gate (*P < 0.05 and **P < 0.01). h, Percentage (mean ± s.e.m.) of HSC/MPPs and early progenitors in fetal liver, yolk sac, cord blood and adult bone marrow expressing MKI67 (*P < 0.05, **P < 0.01 and ****P < 0.001). i, Percentage (mean ± s.e.m.) of CD34+CD38 and CD34+CD38+cells in the indicated cell cycle phases (right) as determined by flow cytometry analysis (left; representative plot of n = 8 biologically independent samples) (G0, Ki67DAPI; G1, Ki67+DAPI; S–G2–M, Ki67+DAPI+ (left)).

Source data

Extended Data Fig. 8 Expression of known PID-linked genes in fetal liver.

Dot plots showing relative expression of genes known to be associated with major PID disease categories in fetal liver cell types from Fig. 1b. Gene-expression frequency (per cent of cells within the cell type expressing the gene) is indicated by spot size and expression level is indicated by colour intensity.

Extended Data Fig. 9 FACS gating strategy for scRNA-seq analysis.

a, Gating strategy used to isolate cells for droplet (10x) and plate-based scRNA-seq (Smart-seq2) for samples F2–F17. b, Gating strategy used to isolate cells for cytospins, scRNA-seq (Smart-seq2) and 100-cell RNA-seq. c, Flow cytometry gating strategy used to identify the colonies cultured in vitro from single cells as shown in Fig. 6c. d, Flow cytometry gating strategy used to identify B and NK colonies cultured in vitro from 10 cells as shown in Fig. 6e.

Supplementary information

Supplementary Figures

Giemsa-stained cytospin images of fetal liver cell states. Raw Giemsa-stained cytospin images as shown in Fig. 2b and Extended Data 5e, where left and right corresponds to their position in Fig. 2b. a, NK cells (left and right). b, pDC precursor (left and right). c, DC1 (left and right). d, DC2. e, Monocytes (left and right). f, Mono-Mac (left and right). g, Kupffer cells (left and right). h, Mast cells (left and right). i, Early erythroid cells (left and right). j, Mid erythroid cells. k, Late erythroid cells (left and right). l, Endothelial cells. m, Fibroblasts (left and right). n, Hepatocytes. o, pre/pro B cells. p, B cells (left and right). All images for a cell state were taken from the same slide at 100x magnification. Scale bar, 10μm.

Reporting Summary

Supplementary Tables

This file contains Supplementary Tables 1-13 with a guide.

Supplementary Tables

This file contains Supplementary Tables 14-22 with a guide.

Supplementary Video 1

Animated force-directed graph of all liver cell states. Animated FDG visualization of fetal liver cell states shown in Fig. 1b. Video can also be found on the web portal.

Supplementary Video 2

3D analysis of fetal skin erythropoiesis. Animation of 3D light-sheet fluorescence microscopy analysis corresponding to Extended Data 3g. Parallel movies of maximum intensity projection rendering (left) and additional shading processes to emphasize 3D rendering (right). < indicates extravascular nucleated erythroid cells.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Popescu, DM., Botting, R.A., Stephenson, E. et al. Decoding human fetal liver haematopoiesis. Nature 574, 365–371 (2019). https://doi.org/10.1038/s41586-019-1652-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1652-y

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