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.

  • Letter
  • Published:

Platelet-biased stem cells reside at the apex of the haematopoietic stem-cell hierarchy

Abstract

The blood system is maintained by a small pool of haematopoietic stem cells (HSCs), which are required and sufficient for replenishing all human blood cell lineages at millions of cells per second throughout life. Megakaryocytes in the bone marrow are responsible for the continuous production of platelets in the blood, crucial for preventing bleeding—a common and life-threatening side effect of many cancer therapies—and major efforts are focused at identifying the most suitable cellular and molecular targets to enhance platelet production after bone marrow transplantation or chemotherapy1. Although it has become clear that distinct HSC subsets exist that are stably biased towards the generation of lymphoid or myeloid blood cells2,3,4, we are yet to learn whether other types of lineage-biased HSC exist or understand their inter-relationships and how differently lineage-biased HSCs are generated and maintained. The functional relevance of notable phenotypic and molecular similarities between megakaryocytes and bone marrow cells with an HSC cell-surface phenotype5,6,7,8 remains unclear. Here we identify and prospectively isolate a molecularly and functionally distinct mouse HSC subset primed for platelet-specific gene expression, with enhanced propensity for short- and long-term reconstitution of platelets. Maintenance of platelet-biased HSCs crucially depends on thrombopoietin, the primary extrinsic regulator of platelet development9. Platelet-primed HSCs also frequently have a long-term myeloid lineage bias, can self-renew and give rise to lymphoid-biased HSCs. These findings show that HSC subtypes can be organized into a cellular hierarchy, with platelet-primed HSCs at the apex. They also demonstrate that molecular and functional priming for platelet development initiates already in a distinct HSC population. The identification of a platelet-primed HSC population should enable the rational design of therapies enhancing platelet output.

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: vWF–eGFP-expressing platelet-biased haematopoietic stem cells.
Figure 2: THPO regulates platelet-primed vWF–eGFP+ HSCs.
Figure 3: Hierarchical relationship between vWF+ and vWF HSCs.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

Data deposits

The microarray data can be found in the ArrayExpress database (http://www.ebi.ac.uk/arrayexpress/) with the accession number E-MEXP-3935.

References

  1. Nash, R. A., Gooley, T., Davis, C. & Appelbaum, F. R. The problem of thrombocytopenia after hematopoietic stem cell transplantation. Oncologist 1, 371–380 (1996)

    CAS  PubMed  Google Scholar 

  2. Müller-Sieburg, C. E., Cho, R. H., Thoman, M., Adkins, B. & Sieburg, H. B. Deterministic regulation of hematopoietic stem cell self-renewal and differentiation. Blood 100, 1302–1309 (2002)

    PubMed  Google Scholar 

  3. Dykstra, B. et al. Long-term propagation of distinct hematopoietic differentiation programs in vivo. Cell Stem Cell 1, 218–229 (2007)

    Article  CAS  PubMed  Google Scholar 

  4. Copley, M. R., Beer, P. A. & Eaves, C. J. Hematopoietic stem cell heterogeneity takes center stage. Cell Stem Cell 10, 690–697 (2012)

    Article  CAS  PubMed  Google Scholar 

  5. Akashi, K. et al. Transcriptional accessibility for genes of multiple tissues and hematopoietic lineages is hierarchically controlled during early hematopoiesis. Blood 101, 383–389 (2003)

    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. Månsson, R. et al. Molecular evidence for hierarchical transcriptional lineage priming in fetal and adult stem cells and multipotent progenitors. Immunity 26, 407–419 (2007)

    Article  PubMed  Google Scholar 

  8. Huang, H. & Cantor, A. B. Common features of megakaryocytes and hematopoietic stem cells: what's the connection? J. Cell. Biochem. 107, 857–864 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. de Sauvage, F. J. et al. Physiological regulation of early and late stages of megakaryocytopoiesis by thrombopoietin. J. Exp. Med. 183, 651–656 (1996)

    Article  CAS  PubMed  Google Scholar 

  10. Forsberg, E. C., Bhattacharya, D. & Weissman, I. L. Hematopoietic stem cells: expression profiling and beyond. Stem Cell Rev. 2, 23–30 (2006)

    CAS  PubMed  Google Scholar 

  11. Denis, C. et al. A mouse model of severe von Willebrand disease: defects in hemostasis and thrombosis. Proc. Natl Acad. Sci. USA 95, 9524–9529 (1998)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kiel, M. J. et al. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121, 1109–1121 (2005)

    Article  CAS  PubMed  Google Scholar 

  13. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–245 (1996)

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Benveniste, P. et al. Intermediate-term hematopoietic stem cells with extended but time-limited reconstitution potential. Cell Stem Cell 6, 48–58 (2010)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  16. Morita, Y., Ema, H. & Nakauchi, H. Heterogeneity and hierarchy within the most primitive hematopoietic stem cell compartment. J. Exp. Med. 207, 1173–1182 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gekas, C. & Graf, T. CD41 expression marks myeloid-biased adult hematopoietic stem cells and increases with age. Blood 121, 4463–4472 (2013)

    Article  CAS  PubMed  Google Scholar 

  18. Shimazu, T. et al. CD86 is expressed on murine hematopoietic stem cells and denotes lymphopoietic potential. Blood 119, 4889–4897 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Goodell, M. A., Brose, K., Paradis, G., Conner, A. S. & Mulligan, R. C. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J. Exp. Med. 183, 1797–1806 (1996)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  21. Yoshihara, H. et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell 1, 685–697 (2007)

    Article  CAS  PubMed  Google Scholar 

  22. Qian, H. et al. Critical role of thrombopoietin in maintaining adult quiescent hematopoietic stem cells. Cell Stem Cell 1, 671–684 (2007)

    Article  CAS  PubMed  Google Scholar 

  23. Moreira, P. N., Pozueta, J., Giraldo, P., Gutierrez-Adan, A. & Montoliu, L. Generation of yeast artificial chromosome transgenic mice by intracytoplasmic sperm injection. Methods Mol. Biol. 349, 151–161 (2006)

    PubMed  Google Scholar 

  24. Bereshchenko, O. et al. Hematopoietic stem cell expansion precedes the generation of committed myeloid leukemia-initiating cells in C/EBPα mutant AML. Cancer Cell 16, 390–400 (2009)

    Article  CAS  PubMed  Google Scholar 

  25. Irizarry, R. A. et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003)

    Article  PubMed  Google Scholar 

  26. Smyth, G. K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, Article3 (2004)

    Article  MathSciNet  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Luc, S. et al. The earliest thymic T cell progenitors sustain B cell and myeloid lineage potential. Nature Immunol. 13, 412–419 (2012)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank the Biomedical Services at Oxford University, B. Wu and H. Ferry for technical assistance, and M. José Fernandez Nestosa, S. Luc and N. Buza-Vidas for discussions and experimental contributions. This work was supported by the Medical Research Council, UK Grant H4RPLK0 (to S.E.W.J.) and an MRC Program Grant and Strategic Award (to C.N.), by the European Commission FP7 CardioCell (to C.N.) and EuroSyStem (to S.E.W.J.) projects, and by the Association for International Cancer Research (to C.N.). A.S.-P. was supported by a postdoctoral BEOI fellowship from the Spanish Ministry of Education and Science.

Author information

Authors and Affiliations

Authors

Contributions

S.E.W.J., C.N. and A.S.-P. designed the project, analysed data and wrote the manuscript. A.S.-P., I.C.M. and C.T.J. organized, performed and analysed most experiments. P.S.W., T.C.L. and A.M. participated in experimental design, statistical analysis and performed/analysed experiments. S.Mo. and S.T. performed analysis of gene expression data. C.C. contributed with cloning of targeting construct for the BAC transgenic mouse model. T.B.J. helped with peripheral blood reconstitution analysis in transplantation studies. O.C. performed stem-cell transplantation experiments. M.L. performed in vitro megakaryocyte culture experiments and helped with transplantations. S.Ma. performed PCR analysis of endothelial and osteoblastic cells. L.S., J.C.A.G., R.F., H.B., A.Gr., A.Ga., J.C. and P.T. were involved in reconstitution analysis. D.A. helped with single-cell qPCR experiments. S.-A.C. performed cell sorting. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Claus Nerlov or Sten Eirik W. Jacobsen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 vWF–eGFP expression in endothelial cells and mature blood cell lineages.

a, vWF–eGFP targeting construct. be, eGFP expression in adult mice in peripheral blood (n = 1) T cells (CD4+ CD8+), B cells (CD19+) and myeloid cells (Mac1+ NK1.1) (b); CD4+and CD8+ single-positive and CD4+ CD8+ double-positive thymocytes (n = 1) (c); platelets (n = 7) and erythrocytes (n = 7) (d); and bone marrow endothelial Ter119CD45CD31+ cells (n = 4) (e). Mean percentages of parental gates are shown. f, g, Endothelial (f) and osteoblastic (g) gene expression in FACS-sorted Ter119CD45CD31+ vWF+ endothelial cells (mean ± s.d., n = 3) and Ter119CD45CD31ALCAM+ Sca-1 osteoblasts (mean ± s.d., n = 3). Hprt was used as a housekeeping gene.

Extended Data Figure 2 vWF–eGFP expression in adult bone marrow precursors and progenitors.

a, b, Gating strategy for adult bone marrow mature B cells (B220+ CD19+), T cells (CD3+) and myeloid cells (Gr-1+ Mac-1+) (a) for analysis of vWF–eGFP expression (n = 3) (b). c, vWF–eGFP expression in erythroid cells (I: CD71highTer119medium; II: CD71high Ter119high; III: CD71medium Ter119high; IV: CD71low Ter119high) (n = 3). d, vWF–eGFP expression in bone marrow progenitors (MKP, preCFU-E, preGM and preMegE in Vwf-eGFPtg/+) (n = 3). e, vWF–eGFP expression in adult LSK CD150Flt3hi lymphoid-primed multipotent progenitors (n = 7).

Extended Data Figure 3 vWF–eGFP mice report Vwf expression.

a, Expression of Vwf messenger RNA relative to Hprt in sorted vWF–eGFP+ and vWF–eGFP LSK CD150+ CD48 CD34 cells isolated from 8–10-week-old Vwf-eGFP reporter mice. Mean ± s.e.m. results from five biological replicates are shown. ***P < 0.0001 (Student’s t-test). b, qPCR analysis showing Vwf mRNA levels relative to Gapdh in FACS-sorted LSK CD150+ CD34CD48 cells isolated from 8-week-old wild-type (WT; n = 2) and transgenic (TG; n = 3) Vwf-eGFP littermates. Each bar shows one biological replicate determination. Gapdh gene expression was used for normalization. c, d, Co-expression on HSCs of vWF–eGFP with CD49b and CD201 (n = 4) (c) and CD41 and CD86 (n = 3) (d). Mean frequencies of parental gate. e, Mean percentages of gated vWF+ and vWF HSCs with side population (SP) phenotype (n = 2). f, Cell cycle analysis of vWF+ and vWF HSCs and CD34+ multipotent progenitors (mean percentages, n = 2, pool of 7 mice each). DAPI, 4′,6-diamidino-2-phenylindole. g, Left, mean (± s.e.m.) total colony formation (white bars) and colonies containing megakaryocytes (grey bars) from single vWF–eGFP+ and vWF–eGFP LSK CD150+ CD48 HSCs (n = 3). Right, typical multilineage colony with megakaryocyte and granulocyte/macrophage lineages.

Extended Data Figure 4 Reconstitution experiments.

a, Experimental outline to evaluate multi-lineage reconstitution and self-renewal potential of LSK CD34CD150+ CD48 eGFP and eGFP+ HSCs. vWF+ and vWF HSCs (CD45.2), either single-cell-sorted by FACS-based automated cell deposition unit or 50 cells bulk sorted, were competitively transplanted along with 1 × 106 w41/w41 unfractionated bone marrow (BM) cells (CD45.1) or 2 × 105 wild-type unfractionated bone marrow cells (CD45.1). Cells were injected into lethally irradiated CD45.1 recipients. Peripheral blood (PB) reconstitution analysis was determined at indicated time points. The HSC compartment was analysed at 32 weeks after transplantation (TP) and bone marrow cells were retrieved from 50-cell recipients and re-transplanted into secondary hosts. b, Sorting of eGFP and eGFP+ LinSca-1+ c-Kit+ CD150+ CD48CD34 cells was performed on c-Kit-enriched bone marrow cells and used for functional (competitive transplantation and hierarchical transplantation experiments) and molecular (global gene profiling and multiplex qPCR) assays. c, Mean (± s.e.m.) contribution of 50 vWF+ and vWF LSK CD34CD150+ CD48 HSCs to peripheral blood platelets, myeloid cells, B cells and T cells 12 weeks after secondary transplantation (n = 12 mice). **P < 0.01 (Student’s t-test).

Extended Data Figure 5 Gating strategy for peripheral blood reconstitution analysis.

a, b, White blood cell and platelet reconstitution analysis in mice 16 weeks after competitive transplantation of CD45.2 vWF+ HSCs (a) and vWF HSCs (b). Donor platelets were identified based on scatter properties (SSC, side scatter) CD150, CD41 and eGFP expression as indicated. Peripheral blood leukocytes were gated as B cells (CD19+), T cells (CD4/CD8+) and myeloid cells (Mac1+ NK1.1); and the percentage donor/test-cell (CD45.2) contribution to each lineage was calculated from these gates.

Extended Data Figure 6 Lineage-biased reconstitution patterns in mice competitively transplanted with limiting numbers of vWF and vWF+ HSCs.

a, Gating strategy to analyse plasma-derived platelets (CD41+ CD150+) and myeloid (Mac1+ NK1.1), B (CD19+) and T (CD4+ CD8+) cells in peripheral blood from competitively transplanted mice. b, Representative FACS plots showing minimal background staining of peripheral blood from CD45.1 mouse stained with anti-CD45.2 antibody. c, Representative FACS plots showing lymphoid-biased reconstitution pattern in peripheral blood of mice transplanted with 10 vWF LSK CD34CD150+CD48 cells at 16 weeks (mouse 1) or 27 weeks (mouse 2) after transplantation. Platelet or platelet/myeloid reconstitution patterns were not observed in any vWF HSC recipients. Also shown are representative 16-week platelet-biased (mice 3–4) and lymphoid-biased (mouse 5) reconstitution patterns in mice transplanted with 10 vWF+ LSK CD34CD150+ CD48 cells. Percentage test-cell (eGFP+ platelets or CD45.2+) contribution to the different lineages is indicated. d, Platelet/myeloid-biased peripheral blood reconstitution in mice transplanted with 5 vWF+ LSK CD34CD150+ CD48 cells as analysed at 24 (mice 8 and 9) or 31–33 (mice 6 and 7) weeks after transplantation. e, FACS plots showing donor contribution to platelets (eGFP+) and white blood cells (CD45.2+) in mice reconstituted with a single vWF+ LSK CD34CD150+CD48 HSC in Fig. 1l.

Extended Data Figure 7 Test-cell contribution to HSC and progenitor compartments in recipients of limiting numbers of vWF and vWF+ HSCs.

a, Gating strategy to analyse HSC and progenitors in bone marrow. b, Representative FACS plots showing staining background in CD45.1 bone marrow stained with anti-CD45.2 antibody. c, Donor contribution to HSC compartment, eGFP expression in donor-derived HSCs and donor contribution to bone marrow progenitor compartments at 29 weeks in recipients, with identity numbers corresponding to peripheral blood analysis in Fig. 1. d, Donor contribution to HSC compartment, eGFP expression in donor-derived HSCs and donor contribution to bone marrow progenitors at 33 weeks in recipients of 5 vWF+ HSCs with identity numbers corresponding to peripheral blood analysis in Fig. 1.

Extended Data Figure 8 Acute depletion of platelets induces rapid cell cycle entry of platelet-primed LSK CD150+ CD48vWF+ HSCs.

a, Outline of experimental design. b, Platelet levels in PBS and anti-CD42b-treated mice 24 h after treatment (n = 6). c, Plasma THPO levels in PBS (n = 2) and anti-CD42b-treated mice (n = 2) 24 h after treatment. Each bar shows one biological replicate determination. d, Representative cell cycle analysis of vWF and vWF+ HSCs in control and anti-CD42b-treated mice 24 h after treatment. Percentages are mean values of 7 mice for control and 8 mice for anti-CD42b-treated mice, from three separate experiments.

Extended Data Figure 9 Expression of lineage-affiliated genes in HSCs in Thpo−/− mice.

a, qPCR analysis of expression of myeloid, erythroid and lymphoid genes in 9-week-old Thpo+/+ (black) and Thpo−/− (white) LSK CD34Flt3 bone marrow cells relative to Hprt. Plots show mean (± s.e.m.) expression values of 6 replicate determinations. **P < 0.01, ***P < 0.001 (Student’s t-test). b, c, Mean contribution of transplanted Thpo−/−vWFtg/+ bone marrow cells (CD45.1) 18 weeks after transplantation to B-cell progenitors (n = 9) (b) and the HSC compartment (n = 4) (c).

Extended Data Figure 10 vWF+ and vWF HSC contribution to B- and T-cell progenitors in secondary recipients.

a, Outline of experiments to establish hierarchical relationship between vWF–eGFP+ and vWF–eGFP HSCs. b, Gating strategy for sorting of CD45.2+ LinvWF–eGFP and CD45.2+LinvWF–eGFP+ cells for transplantation. Cells were sorted from primary recipients competitively transplanted with 150 vWF–eGFPor vWF–eGFP+ CD45.2+ LSK CD150+ CD48CD34 cells 6 weeks earlier and subsequently transplanted into secondary recipients in the hierarchical experiment outlined in a. Bottom panels show purity analysis for sorted populations; the percentage purity of each population is presented as the mean value from three individual experiments. c, Mean peripheral blood lineage contribution 16 weeks after secondary transplantation of CD45.2+ LineGFP or eGFP+ bone marrow cells. eGFP status of cells transplanted into primary and secondary recipients is indicated (n = 8 mice/group from 3 experiments). **P < 0.01, ***P < 0.001 (ANOVA). dg, Gating strategies and test-cell contribution to CD4+ CD8+ thymocytes and B220+ CD19+ CD43+ bone marrow B-cell progenitors 18–20 weeks after secondary transplantation (n = 5) in hierarchical experiments. Representative plots from eGFP+/eGFP+ and eGFP/eGFP recipients are shown, including percentages of CD45.1 and CD45.2 contribution to each cell type. Reconstituted recipients/groups are indicated.

Supplementary information

Supplementary Table 1

This file contains configuration of FACS instruments. (XLS 21 kb)

Supplementary Table 2

This file contains antibodies, viability dyes and reagents used for FACS staining. (XLS 32 kb)

Supplementary Table 3

This file contains UPL assays used for Q-PCR analysis. (XLS 21 kb)

Supplementary Table 4

This file contains Taqman assays used for Q-PCR analysis. (XLS 25 kb)

Supplementary Table 5

This file contains custom prepared gene sets representing signatures of lineage restricted progenitors used for GSEA analysis. (XLSX 31 kb)

Supplementary Table 6

This file contains differentially regulated genes in Vwf- and Vwf+ HSCs by global gene expression profiling. Genes differentially expressed (adjusted P-value <0.05) between Vwf+ and Vwf– HSCs were identified using LIMMA. For these genes the Affymetrix probe set ID, gene symbol, P-value, adjusted P-value, and Vwf+/Vwf– ratio (log2 and linear scale) are shown. Normalized intensity values from individual arrays are also provided. (XLSX 65 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sanjuan-Pla, A., Macaulay, I., Jensen, C. et al. Platelet-biased stem cells reside at the apex of the haematopoietic stem-cell hierarchy. Nature 502, 232–236 (2013). https://doi.org/10.1038/nature12495

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12495

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