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:

Foxc1 is a critical regulator of haematopoietic stem/progenitor cell niche formation

Abstract

Haematopoietic stem and progenitor cells are maintained by special microenvironments known as niches in bone marrow1,2,3,4,5,6. Many studies have identified diverse candidate cells that constitute niches for haematopoietic stem cells in the marrow, including osteoblasts7,8,9,10, endothelial cells11,12,13,14, Schwann cells15, α-smooth muscle actin-expressing macrophages16 and mesenchymal progenitors such as CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells17,18, stem cell factor-expressing cells13, nestin-expressing cells19 and platelet-derived growth factor receptor-α (PDGFR-α)+Sca-1+CD45Ter119 (PαS) cells20. However, the molecular basis of the formation of the niches remains unclear. Here we find that the transcription factor Foxc1 is preferentially expressed in the adipo-osteogenic progenitor CAR cells essential for haematopoietic stem and progenitor cell maintenance in vivo5,13,18 in the developing and adult bone marrow. When Foxc1 was deleted in all marrow mesenchymal cells or CAR cells, from embryogenesis onwards, osteoblasts appeared normal, but haematopoietic stem and progenitor cells were markedly reduced and marrow cavities were occupied by adipocytes (yellow adipose marrow) with reduced CAR cells. Inducible deletion of Foxc1 in adult mice depleted haematopoietic stem and progenitor cells and reduced CXCL12 and stem cell factor expression in CAR cells but did not induce a change to yellow marrow. These data suggest a role for Foxc1 in inhibiting adipogenic processes in CAR progenitors. Foxc1 might also promote CAR cell development, upregulating CXCL12 and stem cell factor expression. This study identifies Foxc1 as a specific transcriptional regulator essential for development and maintenance of the mesenchymal niches for haematopoietic stem and progenitor cells.

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: Foxc1 is preferentially expressed in CAR cells.
Figure 2: Foxc1 deletion results in yellow marrow with reduced HSPCs and CAR cells.
Figure 3: Essential roles of Foxc1 in CAR cells.
Figure 4: Foxc1 is essential for maintenance of HSPC niche functions in adult bone marrow.

Similar content being viewed by others

References

  1. Morrison, S. J. & Spradling, A. C. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 132, 598–611 (2008)

    Article  CAS  Google Scholar 

  2. Li, L. & Clevers, H. Coexistence of quiescent and active adult stem cells in mammals. Science 327, 542–545 (2010)

    Article  ADS  CAS  Google Scholar 

  3. Ehninger, A. & Trumpp, A. The bone marrow stem cell niche grows up: mesenchymal stem cells and macrophages move in. J. Exp. Med. 208, 421–428 (2011)

    Article  CAS  Google Scholar 

  4. Bianco, P. Bone and the hematopoietic niche: a tale of two stem cells. Blood 117, 5281–5288 (2011)

    Article  CAS  Google Scholar 

  5. Nagasawa, T., Omatsu, Y. & Sugiyama, T. Control of hematopoietic stem cells by the bone marrow stromal niche: the role of reticular cells. Trends Immunol. 32, 315–320 (2011)

    Article  CAS  Google Scholar 

  6. Mercier, F. E., Ragu, C. & Scadden, D. T. The bone marrow at the crossroads of blood and immunity. Nature Rev. Immunol. 12, 49–60 (2011)

    Article  Google Scholar 

  7. Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836–841 (2003)

    Article  ADS  CAS  Google Scholar 

  8. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841–846 (2003)

    Article  ADS  CAS  Google Scholar 

  9. Arai, F. et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 118, 149–161 (2004)

    Article  CAS  Google Scholar 

  10. Sugimura, R. et al. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell 150, 351–365 (2012)

    Article  CAS  Google Scholar 

  11. Kiel, M. J., Yilmaz, O. H., Iwashita, T., Terhorst, C. & Morrison, S. J. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121, 1109–1121 (2005)

    Article  CAS  Google Scholar 

  12. 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  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  14. Ding, L. & Morrison, S. J. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 495, 231–235 (2013)

    Article  ADS  CAS  Google Scholar 

  15. Yamazaki, S. et al. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 147, 1146–1158 (2011)

    Article  CAS  Google Scholar 

  16. Ludin, A. et al. Monocytes-macrophages that express α-smooth muscle actin preserve primitive hematopoietic cells in the bone marrow. Nature Immunol. 13, 1072–1082 (2012)

    Article  CAS  Google Scholar 

  17. Sugiyama, T., Kohara, H., Noda, M. & Nagasawa, T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 25, 977–988 (2006)

    Article  CAS  Google Scholar 

  18. Omatsu, Y. et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity 33, 387–399 (2010)

    Article  CAS  Google Scholar 

  19. Mendez-Ferrer, S. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466, 829–834 (2010)

    Article  ADS  CAS  Google Scholar 

  20. Greenbaum, A. et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature 495, 227–230 (2013)

    Article  ADS  CAS  Google Scholar 

  21. Nagasawa, T. et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 382, 635–638 (1996)

    Article  ADS  CAS  Google Scholar 

  22. Sacchetti, B. et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 131, 324–336 (2007)

    Article  CAS  Google Scholar 

  23. Chan, C. K. et al. Endochondral ossification is required for haematopoietic stem-cell niche formation. Nature 457, 490–494 (2009)

    Article  ADS  CAS  Google Scholar 

  24. Morikawa, S. et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J. Exp. Med. 206, 2483–2496 (2009)

    Article  CAS  Google Scholar 

  25. Kume, T. et al. The forkhead/winged helix gene Mf1 is disrupted in the pleiotropic mouse mutation congenital hydrocephalus. Cell 93, 985–996 (1998)

    Article  CAS  Google Scholar 

  26. Sasman, A. et al. Generation of conditional alleles for Foxc1 and Foxc2 in mice. Genesis 50, 766–774 (2012)

    Article  CAS  Google Scholar 

  27. Logan, M. et al. Expression of Cre Recombinase in the developing mouse limb bud driven by a Prxl enhancer. Genesis 33, 77–80 (2002)

    Article  CAS  Google Scholar 

  28. Naveiras, O. et al. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature 460, 259–263 (2009)

    Article  ADS  CAS  Google Scholar 

  29. Kodama, H., Nose, M., Niida, S. & Nishikawa, S. Involvement of the c-kit receptor in the adhesion of hematopoietic stem cells to stromal cells. Exp. Hematol. 22, 979–984 (1994)

    CAS  PubMed  Google Scholar 

  30. Nehls, M. et al. Two genetically separable steps in the differentiation of thymic epithelium. Science 272, 886–889 (1996)

    Article  ADS  CAS  Google Scholar 

  31. Ara, T. et al. Long-term hematopoietic stem cells require stromal cell-derived factor-1 for colonizing bone marrow during ontogeny. Immunity 19, 257–267 (2003)

    Article  CAS  Google Scholar 

  32. Rodda, S. J. & McMahon, A. P. Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development 133, 3231–3244 (2006)

    Article  CAS  Google Scholar 

  33. DeFalco, J. et al. Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus. Science 291, 2608–2613 (2001)

    Article  ADS  CAS  Google Scholar 

  34. Ruzankina, Y. et al. Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1, 113–126 (2007)

    Article  CAS  Google Scholar 

  35. Koni, P. A. et al. Conditional vascular cell adhesion molecule 1 deletion in mice: impaired lymphocyte migration to bone marrow. J. Exp. Med. 193, 741–754 (2001)

    Article  CAS  Google Scholar 

  36. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nature Neurosci. 13, 133–140 (2010)

    Article  CAS  Google Scholar 

  37. Joe, A. W. et al. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nature Cell Biol. 12, 153–163 (2010)

    Article  CAS  Google Scholar 

  38. Uezumi, A., Fukada, S., Yamamoto, N., Takeda, S. & Tsuchida, K. Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nature Cell Biol. 12, 143–152 (2010)

    Article  CAS  Google Scholar 

  39. Harrison, D. E., Jordan, C. T., Zhong, R. K. & Astle, C. M. Primitive hemopoietic stem cells: direct assay of most productive populations by competitive repopulation with simple binomial, correlation and covariance calculations. Exp. Hematol. 21, 206–219 (1993)

    CAS  PubMed  Google Scholar 

  40. Morita, S., Kojima, T. & Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther. 7, 1063–1066 (2000)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We appreciate the technical assistance provided by K. Kawaguchi and G. Kondoh, and thank I. Sasagawa for secretarial assistance. This research was supported by JST, CREST and the Ministry of Education, Culture, Sports, Science and Technology (MEXT)/Japan Society for the Promotion of Science (JSPS) KAKENHI.

Author information

Authors and Affiliations

Authors

Contributions

Y.O. and T.N. designed and performed the experiments, analysed the data and prepared the paper. T.N. supervised the study. M.S. and T.S. performed the experiments. T.K. contributed materials and tools. All authors discussed results and edited the manuscript.

Corresponding author

Correspondence to Takashi Nagasawa.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Progenitors of CAR cells and their development in fetal and postnatal bone marrow.

a, b, Immunohistochemical analysis of E14.5 (a) and E16.5 (b) femurs of Osx–GFP mice with antibodies against PDGFR-β (red) and CD31 (blue). Osx–GFP+ cells (green) expressing PDGFR-β (arrowheads) were observed inside the marrow cavity at E16.5 (b with magnified views of the boxed area on the right) but not at E14.5 (a). B, cortical bone (b, right). c, Immunohistochemical analysis of the newborn femur of CXCL12–GFP mice with antibodies against PDGFR-β (red). CXCL12–GFP+ cells (green) express PDGFR-β. d, Immunofluorescent profiles of Osx–GFP+PDGFR-βhi cells (boxed area) in the limbs of E14.5 and E16.5 Osx–GFP mice. eg Relative mRNA expression levels of PDGFR-β, Lepr, CXCL12, SCF, PPARγ, C/EBPα and Osx in Osx–GFP+PDGFR-βhi, Osx–GFP+PDGFR-βlo, CXCL12–GFP+ cells, osteoblasts (Ob), PαS, Osx–GFPPDGFR-β+Sca-1CD31CD45Ter119 (Osx+) cells and endothelial cells (ECs) from femurs of E16.5 Osx–GFP mice and newborn CXCL12–GFP mice (e) as well as 14- to 20-week-old CXCL12–GFP mice (g), or in CXCL12–GFP+ cells from newborn, 1-week-old, 3-week-old and 14- to 20-week-old CXCL12–GFP mice (f) (n = 3). *P < 0.05.

Extended Data Figure 2 Expression of PDGFR-α in Sca-1+CD31 cells.

Immunohistochemical analysis of the bone marrow cavity of wild-type mice with antibodies against Sca-1 (red), CD31 (blue) and PDGFR-α (green). All Sca-1+CD31 mesenchymal cells surrounding arteries expressed PDGFR-α.

Extended Data Figure 3 Expression levels of Foxc2 were similar in CAR cells and osteoblasts.

ac, Relative mRNA expression levels of Foxc2 in CAR cells, osteoblasts (Ob), PαS cells, bone marrow endothelial cells (ECs), c-kit+Sca-1+Lin (KSL) cells, macrophages and muscle PαS cells from adult mice (a), in Osx–GFP+PDGFR-βhi, Osx–GFP+PDGFR-βlo, CXCL12–GFP+ CAR cells, osteoblasts, PαS, Osx–GFPPDGFR-β+Sca-1CD31CD45Ter119 (Osx+) cells and endothelial cells from femurs of E16.5 Osx–GFP mice (b) and newborn mice (b, c), and in CAR cells and osteoblasts from newborn, 1-week-old, 3-week-old and 14- to 20-week-old CXCL12–GFP mice (c) (n = 3). Error bars, s.d. *P < 0.05.

Extended Data Figure 4 The numbers of functional multilineage reconstituting HSCs were markedly reduced in the bone marrow of Prx1-Cre;Foxc1f/f mice.

a, C-kit+Sca-1+Lin (KSL) cells sorted from 3-week-old control or Prx1-Cre;Foxc1f/f mice as tester progenitors and those sorted from wild-type mice as competitor progenitors were mixed at a ratio of 1:1 and injected intravenously into recipient mice. The percentages of donor-derived Gr-1+ myeloid, B220+ B and CD3+ T cells in peripheral blood were analysed for 14 weeks after transplantation (n = 3). b, The numbers of KSL cells in the bone marrow of 3-week-old control or Prx1-Cre;Foxc1f/f mice (n = 3). Because KSL cell numbers were reduced, a marked decrease in the numbers of functional multilineage reconstituting HSCs in the mutants was observed. c, The numbers of LTC-ICs in the bone marrow of 3-week-old control and Prx1-Cre;Foxc1f/f mice (n = 3). LTC-IC numbers per femurs and tibiae were assayed by limiting dilution analysis. Error bars, s.d. *P < 0.05.

Extended Data Figure 5 The numbers of HSPCs were reduced in newborn Prx1-Cre;Foxc1f/f mice although to a lesser extent than juvenile mutants.

Total haematopoietic cell counts and the numbers of cells in the CD150+CD48 KSL population (HSCs), pro-B cells, pre-B cells and proerythroblasts (pro-E) in the bone marrow of newborn control and Prx1-Cre;Foxc1f/f mice (n = 5). *P < 0.05.

Extended Data Figure 6 CAR cells can be identified as S100+ cells in the marrow cavity.

Immunohistochemical analysis of bone marrow from CXCL12–GFP mice with antibodies against S100 (red). CAR cells (green) were identified as S100+ cells in the marrow cavity.

Extended Data Figure 7 Expression of PDGFR-α in Sca-1+CD31 PαS cells in the bone marrow from Prx1-Cre;Foxc1f/f or Lepr-Cre;Foxc1f/f mice.

a, b, Immunohistochemical analysis of the bone marrow cavity of control, Prx1-Cre;Foxc1f/f (a) and Lepr-Cre;Foxc1f/f mice (b) with antibodies against Sca-1 (red), CD31 (blue) and PDGFR-α (green). Sca-1+CD31 mesenchymal cells surrounding arteries expressed PDGFR-α (arrowheads).

Extended Data Figure 8 The numbers of functional multilineage reconstituting HSCs were markedly reduced in the bone marrow of Lepr-Cre;Foxc1f/f mice.

a, KSL cells sorted from 14- to 18-week-old control or Lepr-Cre;Foxc1f/f mice as tester progenitors and those sorted from wild-type mice as competitor progenitors were mixed at a ratio of 1:1 and injected intravenously into recipient mice. The percentages of donor-derived Gr-1+ myeloid, B220+ B and CD3+ T cells in peripheral blood were analysed for 14 weeks after transplantation (n = 3). b, The numbers of KSL cells in the bone marrow of 14- to 18-week-old control or Lepr-Cre;Foxc1f/f mice (n = 3). Because KSL cell numbers were reduced, a marked decrease in the numbers of functional multilineage reconstituting HSCs in the mutants was observed. c, The numbers of LTC-ICs in the bone marrow of 14- to 18-week-old control or Lepr-Cre;Foxc1f/f mice (n = 3). LTC-IC numbers per femur and tibia were assayed by limiting dilution analysis. Error bars, s.d. *P < 0.05.

Extended Data Figure 9 HSPC maintenance and haematopoiesis were not affected when Foxc1 was deleted from endothelial cells and haematopoietic cells.

Total haematopoietic cell counts and the numbers of LT-HSCs, ST-HSCs, MPPs, CLPs, pro-B cells, pre-B cells, proerythroblasts (pro-E) and granulocyte/macrophage progenitors (GMPs) in the bone marrow of 14- to 18-week-old control and Tie2-Cre;Foxc1f/f mice, in which the Foxc1 gene was deleted in endothelial cells and haematopoietic cells (n = 3).

Extended Data Table 1 Primers for RT–PCR

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Omatsu, Y., Seike, M., Sugiyama, T. et al. Foxc1 is a critical regulator of haematopoietic stem/progenitor cell niche formation. Nature 508, 536–540 (2014). https://doi.org/10.1038/nature13071

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13071

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