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:

The transcription factor Foxm1 is essential for the quiescence and maintenance of hematopoietic stem cells

Abstract

Foxm1 is known as a typical proliferation-associated transcription factor. Here we found that Foxm1 was essential for maintenance of the quiescence and self-renewal capacity of hematopoietic stem cells (HSCs) in vivo in mice. Reducing expression of FOXM1 also decreased the quiescence of human CD34+ HSCs and progenitor cells, and its downregulation was associated with a subset of myelodysplastic syndrome (MDS). Mechanistically, Foxm1 directly bound to the promoter region of the gene encoding the receptor Nurr1 (Nr4a2; called 'Nurr1' here), inducing transcription, while forced expression of Nurr1 reversed the loss of quiescence observed in Foxm1-deficient cells in vivo. Thus, our studies reveal a previously unrecognized role for Foxm1 as a critical regulator of the quiescence and self-renewal of HSCs mediated at least in part by control of Nurr1 expression.

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

Access options

Buy this article

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

Figure 1: Foxm1 loss leads to abnormal hematopoiesis.
Figure 2: Marked decrease in the size of HSC and HPC pools in Foxm1-deficient mice.
Figure 3: Depletion of Foxm1 decreases the self-renewal capacity of HSCs.
Figure 4: Foxm1-deficient HSCs have a significantly diminished repopulation capacity.
Figure 5: Deletion of Foxm1 leads to accumulation of cells in the S and G2-M phases of the cell cycle and loss of quiescence in HSCs and/or HPCs.
Figure 6: Genes and molecular pathways dysregulated in Foxm1-deficient HSCs.
Figure 7: Forced expression of Nurr1 'rescues' Foxm1 deletion–induced loss of quiescence in vivo.
Figure 8: Downregulation of FOXM1 is associated with a subset of patients with MDS with deletion of chromosome 5q, and reduced expression of FOXM1 in human primitive hematopoietic cells decreases quiescence.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Reya, T., Morrison, S.J., Clarke, M.F. & Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Cheung, T.H. & Rando, T.A. Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Pietras, E.M., Warr, M.R. & Passegue, E. Cell cycle regulation in hematopoietic stem cells. J. Cell Biol. 195, 709–720 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kalin, T.V., Ustiyan, V. & Kalinichenko, V.V. Multiple faces of FoxM1 transcription factor: lessons from transgenic mouse models. Cell Cycle 10, 396–405 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Costa, R.H. FoxM1 dances with mitosis. Nat. Cell Biol. 7, 108–110 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Laoukili, J. et al. FoxM1 is required for execution of the mitotic programme and chromosome stability. Nat. Cell Biol. 7, 126–136 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Ye, H. et al. Hepatocyte nuclear factor 3/fork head homolog 11 is expressed in proliferating epithelial and mesenchymal cells of embryonic and adult tissues. Mol. Cell. Biol. 17, 1626–1641 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kalin, T.V. et al. Forkhead Box m1 transcription factor is required for perinatal lung function. Proc. Natl. Acad. Sci. USA 105, 19330–19335 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ustiyan, V. et al. Forkhead box M1 transcriptional factor is required for smooth muscle cells during embryonic development of blood vessels and esophagus. Dev. Biol. 336, 266–279 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, X., Kiyokawa, H., Dennewitz, M.B. & Costa, R.H. The Forkhead Box m1b transcription factor is essential for hepatocyte DNA replication and mitosis during mouse liver regeneration. Proc. Natl. Acad. Sci. USA 99, 16881–16886 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ackermann Misfeldt, A., Costa, R.H. & Gannon, M. Beta-cell proliferation, but not neogenesis, following 60% partial pancreatectomy is impaired in the absence of FoxM1. Diabetes 57, 3069–3077 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ye, H., Holterman, A.X., Yoo, K.W., Franks, R.R. & Costa, R.H. Premature expression of the winged helix transcription factor HFH-11B in regenerating mouse liver accelerates hepatocyte entry into S phase. Mol. Cell. Biol. 19, 8570–8580 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Koo, C.Y., Muir, K.W. & Lam, E.W. FOXM1: from cancer initiation to progression and treatment. Biochim. Biophys. Acta 1819, 28–37 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Xue, L., Chiang, L., He, B., Zhao, Y.Y. & Winoto, A. FoxM1, a forkhead transcription factor is a master cell cycle regulator for mouse mature T cells but not double positive thymocytes. PLoS ONE 5, e9229 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ren, X. et al. Forkhead box M1 transcription factor is required for macrophage recruitment during liver repair. Mol. Cell. Biol. 30, 5381–5393 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sirin, O., Lukov, G.L., Mao, R., Conneely, O.M. & Goodell, M.A. The orphan nuclear receptor Nurr1 restricts the proliferation of haematopoietic stem cells. Nat. Cell Biol. 12, 1213–1219 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. Ficara, F., Murphy, M.J., Lin, M. & Cleary, M.L. Pbx1 regulates self-renewal of long-term hematopoietic stem cells by maintaining their quiescence. Cell Stem Cell 2, 484–496 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Socolovsky, M. et al. Ineffective erythropoiesis in Stat5a−/−5b−/− mice due to decreased survival of early erythroblasts. Blood 98, 3261–3273 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I.L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. 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 

  23. Ding, L., Saunders, T.L., Enikolopov, G. & Morrison, S.J. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 481, 457–462 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Butler, J.M. et al. Endothelial cells are essential for the self-renewal and repopulation of Notch-dependent hematopoietic stem cells. Cell Stem Cell 6, 251–264 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kobayashi, H. et al. Angiocrine factors from Akt-activated endothelial cells balance self-renewal and differentiation of haematopoietic stem cells. Nat. Cell Biol. 12, 1046–1056 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kühn, R., Schwenk, F., Aguet, M. & Rajewsky, K. Inducible gene targeting in mice. Science 269, 1427–1429 (1995).

    Article  PubMed  Google Scholar 

  27. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804–1808 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Ma, R.Y. et al. Raf/MEK/MAPK signaling stimulates the nuclear translocation and transactivating activity of FOXM1c. J. Cell Sci. 118, 795–806 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Wang, I.C. et al. Forkhead box M1 regulates the transcriptional network of genes essential for mitotic progression and genes encoding the SCF (Skp2-Cks1) ubiquitin ligase. Mol. Cell. Biol. 25, 10875–10894 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang, I.C. et al. FoxM1 regulates transcription of JNK1 to promote the G1/S transition and tumor cell invasiveness. J. Biol. Chem. 283, 20770–20778 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yilmaz, O.H. et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 441, 475–482 (2006).

    CAS  PubMed  Google Scholar 

  32. Qian, Z., Chen, L., Fernald, A.A., Williams, B.O. & Le Beau, M.M. A critical role for Apc in hematopoietic stem and progenitor cell survival. J. Exp. Med. 205, 2163–2175 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Harrison, D.E., Stone, M. & Astle, C.M. Effects of transplantation on the primitive immunohematopoietic stem cell. J. Exp. Med. 172, 431–437 (1990).

    Article  CAS  PubMed  Google Scholar 

  34. Xue, S. et al. RNA regulons in Hox 5′ UTRs confer ribosome specificity to gene regulation. Nature 517, 33–38 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Signer, R.A., Magee, J.A., Salic, A. & Morrison, S.J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 509, 49–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Helin, K. Regulation of cell proliferation by the E2F transcription factors. Curr. Opin. Genet. Dev. 8, 28–35 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Wierstra, I. & Alves, J. Despite its strong transactivation domain, transcription factor FOXM1c is kept almost inactive by two different inhibitory domains. Biol. Chem. 387, 963–976 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Gossen, M. et al. Transcriptional activation by tetracyclines in mammalian cells. Science 268, 1766–1769 (1995).

    Article  CAS  PubMed  Google Scholar 

  40. Lacorazza, H.D. et al. The transcription factor MEF/ELF4 regulates the quiescence of primitive hematopoietic cells. Cancer Cell 9, 175–187 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Nimer, S.D. MDS: a stem cell disorder–but what exactly is wrong with the primitive hematopoietic cells in this disease? Hematology 43–51 (2008).

  42. Pellagatti, A. et al. Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells. Leukemia 24, 756–764 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Xie, Z. et al. Foxm1 transcription factor is required for maintenance of pluripotency of P19 embryonal carcinoma cells. Nucleic Acids Res. 38, 8027–8038 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wang, Z. et al. FoxM1 in tumorigenicity of the neuroblastoma cells and renewal of the neural progenitors. Cancer Res. 71, 4292–4302 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhang, N. et al. FoxM1 promotes β-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell 20, 427–442 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wierstra, I. The transcription factor FOXM1 (Forkhead box M1): proliferation-specific expression, transcription factor function, target genes, mouse models, and normal biological roles. Adv. Cancer Res. 118, 97–398 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Qian, Z., Chen, L., Fernald, A.A., Williams, B.O. & Le Beau, M.M. A critical role for Apc in hematopoietic stem and progenitor cell survival. J. Exp. Med. 205, 2163–2175 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hou, Y. et al. FHL2 regulates hematopoietic stem cell functions under stress conditions. Leukemia 3, 615–624 (2014).

    Google Scholar 

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

    PubMed  Google Scholar 

  50. Zhang, C.C., Kaba, M., Iizuka, S., Huynh, H. & Lodish, H.F. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood 111, 3415–3423 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M.A. Goodell (Baylor College of Medicine) for the plasmid MSCV-FlagNurr1. Supported by the US National Institutes of Health (RO1 CA140979 to Z.Q.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Ho., W.L., Y.S., L.L., Y.Hu. and Z.Z. performed research; T.Z., D.P., J.G.Q., W.W. and Y.Z. provided advice, reagents and/or analytic tools; Y.Ho. and Z.Q. designed the research and performed data analysis; and Y.Ho., D.P., J.G.Q. and Z.Q. wrote the paper.

Corresponding author

Correspondence to Zhijian Qian.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Foxm1 loss leads to bone marrow hypocellularity.

(a-b) Analysis of ablation of Foxm1 as determined by semiquantitative PCR analysis of genomic DNA (a) as well as qRT-PCR analysis of mRNAs (b) from BM cells from both Foxm1fl/fl Tie2-Cre and Foxm1fl/fl mice. (c) Histological analysis of hematoxylin and eosin-stained sternum from the representative Foxm1fl/fl Tie2-Cre and Foxm1fl/fl mice.

Supplementary Figure 2 Foxm1 loss leads to a decreased number of mature blood cells but does not interfere with lineage differentiation.

(a) Histograms represent the total number of myeloid cells (Mac-1+Gr-1+), B cells (B220+ IgM+), Megakaryocyte (CD41+) and Macrophage (F4/80+) in BM from Foxm1fl/fl Tie2-Cre and Foxm1fl/fl mice. Mean ± standard deviation (SD); n = 5. *, P<0.05. (b-e) Representative histograms show the frequency of myeloid cells (Mac-1+Gr+) (b), B cells (B220+IgM+) (c), Megakaryocyte (CD41+) (d) and Macrophage (F4/80+) (e) in BM from Foxm1fl/fl Tie2-Cre and Foxm1fl/fl mice. (f-i) The frequency and representative flow cytometric histograms of four differentiation stages of erythroblasts including R1 (CD71+Ter119), R2 (CD71+Ter119+), R3 (CD71lowTer119+) and R4 (CD71Ter119+) in BM cells (f,h) and spleen cells (g,i) from Foxm1fl/fl Tie2-Cre and Foxm1fl/fl mice at age of 6-8 weeks.

Supplementary Figure 3 Foxm1 is efficiently deleted in LSK cells from Foxm1fl/flMx1-Cre mice after injection of poly(I:C).

(a) Analysis of Foxm1 deletion as determined by semiquantitative PCR analysis of genomic DNA from LSK cells from Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice 3 weeks after 3 doses of pI-pC injection. (b) qRT-PCR analysis of mRNAs from LSK cells from both Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice 3 weeks after 3 doses of pI-pC injection. (c) The total number of BM cells were reduced in Foxm1fl/fl Mx1-Cre mice as compared to Foxm1fl/fl mice (n=5, P<0.05).

Supplementary Figure 4 Induction of Foxm1 deletion reduces HSC and HPC pools in Foxm1fl/flMx1-Cre chimeric mice.

(a) Comparison of engraftment frequency of Foxm1fl/fl Mx1-Cre (CD45.2+) or control Foxm1fl/fl BM cells (CD45.2+) in WT (CD45.1+) recipient mice by flow cytometric analysis of peripheral blood at 6 weeks after transplantation. (b) Total number of LSK, LSKCD34 and LT-HSC in BM from chimeric Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice at 6 weeks after pI-pC injection. (mean±SD, n=5). *, P<0.05. (c) Total number of HPC, CMP, GMP and MEP in BM from Foxm1fl/fl Mx1-Cre and Foxm1fl/fl chimeric mice at 6 weeks after pI-pC injection (mean±SD, n=5)*. P<0.05.

Supplementary Figure 5 Foxm1 deletion results in a decreased repopulation capacity but does not affect the lineage differentiation of BM cells.

(a-c) The ratio of donor-derived CD45.2+CD45.1 from Foxm1fl/flTie2-Cre or Foxm1fl/fl vs competitor-cell-derived CD45.1+CD45.2+ in myeloid cells, B cells, and T cells in PB from first and secondary recipient mice were analyzed (mean ± SD, n = 4-5). (d) Flow cytometric analysis of the frequency of LSK, LSK CD34 in the competitive repopulated recipient mice 4 months after secondary transplantation. (e) Flow cytometric analysis of CD45.2+CD45.1 and CD45.1+CD45.2+ cells in LSK and LSK CD34 stem cell–enriched population in the competitive repopulated recipient mice 4 months after secondary transplantation. Donor cells were from Foxm1fl/flTie2-Cre or Foxm1fl/fl. (f) Lineage differentiation in the recipient mice transplanted with BM cells from Foxm1fl/flTie2-Cre and Foxm1fl/fl chimeric mice. Histogram shows percentage of donor-derived (CD45.2+CD45.1) myeloid cells (Gr-1+Mac+), B cells (B220+), and T cells (CD3+) in PB analyzed 4 months after the first and secondary transplantation (mean ± SD, n = 4-5).

Supplementary Figure 6 Induction of Foxm1 deletion alters cell-cycle progression in HSCs and HPCs from Foxm1fl/flMx1-Cre mice and chimeric Foxm1fl/flMx1-Cre mice.

(a-c) The histograms depicting the cell cycle profile of HPCs (a), LSK cells (b) and LT-HSCs (c) in Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice (mean±SD, n=3). *, P<0.05, **, P<0.005. (d) The histogram depicting cell cycle status of LSK cells in Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice (mean±SD, n= 5). *, P<0.05, **, P<0.005. (e-g) The histograms depicting the cell cycle profile of HPCs (e), LSK cells (f) and LT-HSCs (g) in chimeric Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice (mean±SD, n=5). *, P<0.05; **, P<0.005; ***, P<0.0005. (h) The histogram depicting cell cycle status of LSK cells in chimeric Foxm1fl/fl Mx1-Cre and Foxm1fl/fl mice (mean±SD, n= 5). *, P<0.05; **, P<0.005.

Supplementary Figure 7 Loss of Foxm1 increases the apoptosis of HPCs and LSK cells under stress.

(a) Histogram shows the mean frequency of apoptosis of HPCs and LSK cells from Foxm1fl/fl Tie2-Cre and Foxm1fl/fl mice (mean ± SD; n = 11). (b) Histogram shows the mean frequency of apoptosis of HPCs and LSK cells from Foxm1fl/fl Tie2-Cre and Foxm1fl/fl chimeric mice (mean ± SD; n = 5). *, P<0.05. (c) Histogram shows the mean frequency of apoptosis of HPCs and LSK cells from Foxm1fl/fl Mx1-Cre and Foxm1fl/fl chimeric mice. (mean ± SD; n = 5). *, P<0.05.

Supplementary Figure 8 Expression of the genes encoding p21 and p27 is upregulated by Nurr1 overexpression.

(a) Doxycycline-induced expression of Flag-Nurr1 in BM cells from the chimeric mice. The Flag-Nurr1 expression in BM cells was determined by Western Blot. The Foxm1/ BM cells expressing pLVX-Tet-On and pLVX-tight-puro-Nurr1 or pLVX-tight-puro or Foxm1fl/fl BM cells expressing pLVX-Tet-On and pLVX-tight-puro from chimeric mice received two-week-induction of Doxycyline. (b) qRT-PCR analysis of p21, p27 and p16 expression in LSK cells. The LSK cells, isolated from Foxm1fl/fl Tie2-Cre or control Foxm1fl/fl mice, were infected with MSCV-puro-Nurr1 or control vector and cultured with Stemspan medium with cytokines. The cells were treated with Puromycin for 2 days before analysis. Gene expression was initially normalized to Actb expression. Values represent the fold changes in gene expression relative to that in Foxm1fl/fl LSK cells expressing control vector. *, P<0.05.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1–4 (PDF 2324 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hou, Y., Li, W., Sheng, Y. et al. The transcription factor Foxm1 is essential for the quiescence and maintenance of hematopoietic stem cells. Nat Immunol 16, 810–818 (2015). https://doi.org/10.1038/ni.3204

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3204

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