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:

Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF

Abstract

Endothelial cells and leptin receptor+ (LepR+) stromal cells are critical sources of haematopoietic stem cell (HSC) niche factors, including stem cell factor (SCF), in bone marrow. After irradiation or chemotherapy, these cells are depleted while adipocytes become abundant. We discovered that bone marrow adipocytes synthesize SCF. They arise from Adipoq-Cre/ER+ progenitors, which represent 5% of LepR+ cells, and proliferate after irradiation. Scf deletion using Adipoq-Cre/ER inhibited haematopoietic regeneration after irradiation or 5-fluorouracil treatment, depleting HSCs and reducing mouse survival. Scf from LepR+ cells, but not endothelial, haematopoietic or osteoblastic cells, also promoted regeneration. In non-irradiated mice, Scf deletion using Adipoq-Cre/ER did not affect HSC frequency in long bones, which have few adipocytes, but depleted HSCs in tail vertebrae, which have abundant adipocytes. A-ZIP/F1 ‘fatless’ mice exhibited delayed haematopoietic regeneration in long bones but not in tail vertebrae, where adipocytes inhibited vascularization. Adipocytes are a niche component that promotes haematopoietic regeneration.

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: Irradiation disrupted sinusoids and depleted HSCs, endothelial cells, and LepR+ stromal cells while dramatically increasing adipocytes in the bone marrow.
Figure 2: Scf was highly expressed by LepR+ stromal cells and adipocytes in the bone marrow before and after irradiation.
Figure 3: Scf from LepR+ stromal cells, but not endothelial cells, is necessary for haematopoietic regeneration and mouse survival after irradiation.
Figure 4: Adipoq-Cre/ER recombines in most adipocytes and in a subset of LepR+ stromal cells in the bone marrow.
Figure 5: Adipoq-Cre/ER+ bone marrow stromal cells form adipocytes, but rarely osteoblasts, in vivo.
Figure 6: Scf from adipocytes is required for the regeneration of HSCs and haematopoiesis and mouse survival after irradiation.
Figure 7: Scf from adipocytes is required for normal HSC frequency and haematopoiesis in the caudal vertebrae of normal young adult mice.
Figure 8: A-ZIP/F1 ‘fatless’ mice exhibit delayed haematopoietic regeneration in the femur but accelerated regeneration of haematopoiesis and vasculature in caudal vertebrae.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

References

  1. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Acar, M. et al. Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal. Nature 526, 126–130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell 15, 154–168 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Oguro, H., Ding, L. & Morrison, S. J. SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell 13, 102–116 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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  PubMed  Google Scholar 

  8. Omatsu, Y., Seike, M., Sugiyama, T., Kume, T. & Nagasawa, T. Foxc1 is a critical regulator of haematopoietic stem/progenitor cell niche formation. Nature 508, 536–540 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. 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  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kunisaki, Y. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. Kfoury, Y. & Scadden, D. T. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell 16, 239–253 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Mizoguchi, T. et al. Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development. Dev. Cell 29, 340–349 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rodeheffer, M. S., Birsoy, K. & Friedman, J. M. Identification of white adipocyte progenitor cells in vivo. Cell 135, 240–249 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Festa, E. et al. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell 146, 761–771 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lee, P. et al. Irisin and FGF21 are cold-induced endocrine activators of brown fat function in humans. Cell Metab. 19, 302–309 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Park, D. et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell 10, 259–272 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hanoun, M. et al. Acute myelogenous leukemia-induced sympathetic neuropathy promotes malignancy in an altered hematopoietic stem cell niche. Cell Stem Cell 15, 365–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Worthley, D. L. et al. Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential. Cell 160, 269–284 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  23. Mendez-Ferrer, S., Lucas, D., Battista, M. & Frenette, P. S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature 452, 442–447 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Lucas, D. et al. Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration. Nat. Med. 19, 695–703 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chow, A. et al. Bone marrow CD169 + macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J. Exp. Med. 208, 261–271 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Christopher, M. J., Rao, M., Liu, F., Woloszynek, J. R. & Link, D. C. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J. Exp. Med. 208, 251–260 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bruns, I. et al. Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion. Nat. Med. 20, 1315–1320 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhao, M. et al. Megakaryocytes maintain homeostatic quiescence and promote post-injury regeneration of hematopoietic stem cells. Nat. Med. 20, 1321–1326 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Calvo, W., Fliedner, T. M., Herbst, E., Hugl, E. & Bruch, C. Regeneration of blood-forming organs after autologous leukocyte transfusion in lethally irradiated dogs. II. Distribution and cellularity of the marrow in irradiated and transfused animals. Blood 47, 593–601 (1976).

    CAS  PubMed  Google Scholar 

  31. Meunier, P., Aaron, J., Edouard, C. & Vignon, G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin. Orthop. Relat. Res. 80, 147–154 (1971).

    Article  CAS  PubMed  Google Scholar 

  32. Abella, E. et al. Bone marrow changes in anorexia nervosa are correlated with the amount of weight loss and not with other clinical findings. Am. J. Clin. Pathol. 118, 582–588 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Gimble, J. M., Robinson, C. E., Wu, X. & Kelly, K. A. The function of adipocytes in the bone marrow stroma: an update. Bone 19, 421–428 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Moitra, J. et al. Life without white fat: a transgenic mouse. Genes Dev. 12, 3168–3181 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nunez, N. P. et al. Accelerated tumor formation in a fatless mouse with type 2 diabetes and inflammation. Cancer Res. 66, 5469–5476 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Ablamunits, V. et al. Susceptibility to induced and spontaneous carcinogenesis is increased in fatless A-ZIP/F-1 but not in obese ob/ob mice. Cancer Res. 66, 8897–8902 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Hooper, A. T. et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 4, 263–274 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhou, B. O., Ding, L. & Morrison, S. J. Hematopoietic stem and progenitor cells regulate the regeneration of their niche by secreting Angiopoietin-1. eLife 4, e05521 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kopp, H. G. et al. Tie2 activation contributes to hemangiogenic regeneration after myelosuppression. Blood 106, 505–513 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Li, X. M., Hu, Z., Jorgenson, M. L., Wingard, J. R. & Slayton, W. B. Bone marrow sinusoidal endothelial cells undergo nonapoptotic cell death and are replaced by proliferating sinusoidal cells in situ to maintain the vascular niche following lethal irradiation. Exp. Hematol. 36, 1143–1156 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Knospe, W. H., Blom, J. & Crosby, W. H. Regeneration of locally irradiated bone marrow. I. Dose dependent, long-term changes in the rat, with particular emphasis upon vascular and stromal reaction. Blood 28, 398–415 (1966).

    CAS  PubMed  Google Scholar 

  42. Greenberg, A. S. et al. Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. J. Biol. Chem. 266, 11341–11346 (1991).

    CAS  PubMed  Google Scholar 

  43. Liu, F. et al. Expression and activity of osteoblast-targeted Cre recombinase transgenes in murine skeletal tissues. Int. J. Dev. Biol. 48, 645–653 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. 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 

  45. Sassmann, A., Offermanns, S. & Wettschureck, N. Tamoxifen-inducible Cre-mediated recombination in adipocytes. Genesis 48, 618–625 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Jeffery, E. et al. Characterization of Cre recombinase models for the study of adipose tissue. Adipocyte 3, 206–211 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang, C. C. & Lodish, H. F. Cytokines regulating hematopoietic stem cell function. Curr. Opin. Hematol. 15, 307–311 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Broudy, V. C. Stem cell factor and hematopoiesis. Blood 90, 1345–1364 (1997).

    CAS  PubMed  Google Scholar 

  49. Randall, T. D. & Weissman, I. L. Phenotypic and functional changes induced at the clonal level in hematopoietic stem cells after 5-fluorouracil treatment. Blood 89, 3596–3606 (1997).

    CAS  PubMed  Google Scholar 

  50. Spindler, T. J., Tseng, A. W., Zhou, X. & Adams, G. B. Adipocytic cells augment the support of primitive hematopoietic cells in vitro but have no effect in the bone marrow niche under homeostatic conditions. Stem Cells Dev. 23, 434–441 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Corre, J. et al. Human bone marrow adipocytes support complete myeloid and lymphoid differentiation from human CD34 cells. Br. J. Haematol. 127, 344–347 (2004).

    Article  PubMed  Google Scholar 

  52. Glettig, D. L. & Kaplan, D. L. Extending human hematopoietic stem cell survival in vitro with adipocytes. Biores Open Access 2, 179–185 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Belaid-Choucair, Z. et al. Human bone marrow adipocytes block granulopoiesis through neuropilin-1-induced granulocyte colony-stimulating factor inhibition. Stem Cells 26, 1556–1564 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. DiMascio, L. et al. Identification of adiponectin as a novel hemopoietic stem cell growth factor. J. Immunol. 178, 3511–3520 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Poloni, A. et al. Molecular and functional characterization of human bone marrow adipocytes. Exp. Hematol. 41, 558–566 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Yue, R., Zhou, B. O., Shimada, I. S., Zhao, Z. & Morrison, S. J. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell 18, 782–796 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  58. 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  PubMed  PubMed Central  Google Scholar 

  59. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  61. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat. Methods 2, 419–426 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. He, W. et al. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc. Natl Acad. Sci. USA 100, 15712–15717 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kalajzic, Z. et al. Directing the expression of a green fluorescent protein transgene in differentiated osteoblasts: comparison between rat type I collagen and rat osteocalcin promoters. Bone 31, 654–660 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Morrison, S. J. et al. Transient Notch activation initiates an irreversible switch from neurogenesis to gliogenesis by neural crest stem cells. Cell 101, 499–510 (2000).

    Article  CAS  PubMed  Google Scholar 

  65. Bianco, P., Kuznetsov, S. A., Riminucci, M. & Gehron Robey, P. Postnatal skeletal stem cells. Methods Enzymol. 419, 117–148 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

S.J.M. is a Howard Hughes Medical Institute Investigator, the Mary McDermott Cook Chair in Pediatric Genetics, the Kathryn and Gene Bishop Distinguished Chair in Pediatric Research, the director of the Hamon Laboratory for Stem Cells and Cancer, and a Cancer Prevention and Research Institute of Texas Scholar. B.O.Z. was supported by a fellowship from the Leukemia and Lymphoma Society and the Thousand Talents Plan-Youth in China. This work was funded by the National Institute on Aging (R37 AG024945) and the Cancer Prevention and Research Institute of Texas. We thank E. Jeffery for advice on the manuscript, N. Loof and the Moody Foundation Flow Cytometry Facility, and K. Correll for mouse colony management.

Author information

Authors and Affiliations

Authors

Contributions

B.O.Z. performed most of the experiments. H.Y. and R.Y. helped in some imaging and transplantation experiments, respectively. Z.Z. performed statistical analyses. J.J.R. provided human bone marrow specimens. O.N. participated in the interpretation of results from A-ZIP/F1 mice. B.O.Z. and S.J.M. designed the experiments, interpreted the results, and wrote the manuscript.

Corresponding authors

Correspondence to Bo O. Zhou or Sean J. Morrison.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Irradiation depletes bone marrow hematopoiesis.

One million mechanically dissociated bone marrow cells from wild-type mice were transplanted into irradiated wild-type mice. The statistical significance of differences among treatments was assessed using repeated measures one-way ANOVAs with the Geisser-Greenhouse method for sphericity correction and Tukey’s multiple comparisons tests (ae). indicates statistical significance relative to non-irradiated controls (con) while #indicates statistical significance between 2 and 4 weeks after irradiation (P < 0.05, or ## P < 0.01, P < 0.001). All data represent mean ± s.d. from two femurs and two tibias in n = 5 mice/genotype/condition from 5 independent experiments. (ae) Numbers of B (a) and T (b) cells in the bone marrow as well as WBC (c), RBC (d), and platelet (e) counts in non-irradiated control (Con) mice as well as mice at 2 and 4 weeks after irradiation and bone marrow transplantation. (f) Confocal imaging showed that perilipin+ adipocytes were uniformly Tomato+ in Leprcre; R26tdTomao mice 2 weeks after irradiation and bone marrow transplantation (representative image from 3 independent experiments).

Supplementary Figure 2 Scf was expressed by adipocytes but not by hematopoietic cells or osteoblasts in the bone marrow of non-irradiated or irradiated mice.

(a) Virtually all Scf-GFPhigh cells were Tomato+ in the bone marrow of irradiated and non-irradiated Leprcre; R26tdTomao; ScfGFP mice (representative plots from 3 independent experiments). (b) Scf-GFP was not expressed by CD45+ or Ter119+ hematopoietic cells in the bone marrow of non-irradiated or irradiated ScfGFP mice (representative plots from 3 independent experiments). (c) Tomato+ osteoblasts did not express Scf-GFP in non-irradiated or irradiated Col1a12.3-cre; R26tdTomato; ScfGFP mice (representative plots from 3 independent experiments). (d,e) Scf-GFP was not detectably expressed by Tomato+ osteoblasts or osteocytes in femur sections from non-irradiated or irradiated Col1a12.3-cre; R26tdTomato; ScfGFP mice (representative images from 3 independent experiments). (f,g) Scf-GFP and Tomato were expressed by perilipin+ adipocytes in the bone marrow of normal Leprcre; R26tdTomao; ScfGFP mice (f) as well as 2 weeks after irradiation and wild-type bone marrow transplantation (g; representative images from 3 independent experiments).

Supplementary Figure 3 Scf is expressed by bone marrow adipocytes.

(a) Confocal imaging showed five consecutive optical images (each 1.5 μm) demonstrating the expression of Scf-GFP in perilipin+ adipocytes from non-irradiated mice (representative images from 3 independent experiments). (bf) Quantitative RT-PCR analysis of leptin (b), Ob-Rb (c), Adipoq (d), perilipin (e) and FABP4 (f) transcript levels (normalized to β-Actin) in LepR+ stromal cells (LepR+), bone marrow adipocytes (Adip-BM) and intraperitoneal adipocytes (Adip-IP) relative to unfractionated whole bone marrow cells (WBM). The transcript levels in WBM were normalized to 1. All data represent mean ± s.d. from n = 3 mice in 3 independent experiments. One-way ANOVAs with Tukey’s multiple comparisons tests were used to assess statistical significance (, P < 0.05; , P < 0.01; , P < 0.001). (g) Abundant adipocytes in human tibia bone marrow sections from an 8 year-old donor (representative image from 3 independent experiments).

Supplementary Figure 4 Scf from osteoblasts and hematopoietic cells is dispensable for the regeneration of HSCs and hematopoiesis after irradiation.

(af) White blood cell (a) red blood cell (b), and platelet counts (c), as well as bone marrow cellularity (d) and numbers of LSK cells (e) and HSCs (f) from paired Col1a12.3-cre; ScfGFP/fl mice and ScfGFP/fl controls that were non-irradiated (Con) or analyzed at 2 or 4 weeks after irradiation and bone marrow transplantation (n = 5 mice/genotype/condition from 3 independent experiments). HSCs could not be detected at 2 weeks after irradiation. Two-way ANOVAs with Sidak’s multiple comparisons tests (ae) or two-tailed Student’s t-tests with Holm-Sidak’s multiple comparisons test (f) were used to assess the significance of differences between Col1a12.3-cre; ScfGFP/fl mice and ScfGFP/fl controls. (g) Competitive reconstitution assay in which 106 donor bone marrow cells from the indicated primary recipient mice were transplanted 4 weeks after irradiation along with recipient-type competitor cells into irradiated secondary recipient mice (n = 12 recipient mice/genotype from 3 independent experiments). The statistical significance of differences was assessed using two-way repeated measures ANOVAs with Sidak’s multiple comparisons tests. (hm) White blood cell (h) red blood cell (i), and platelet counts (j), as well as bone marrow cellularity (k) and numbers of LSK cells (l) and HSCs (m) from paired Vav1-cre; ScfGFP/fl mice and ScfGFP/fl controls that were non-irradiated (Con) or analyzed at 2 or 4 weeks after irradiation and bone marrow transplantation (n = 5 mice/genotype/condition from 3 independent experiments). Two-way ANOVAs with Sidak’s multiple comparisons tests (hl) or two-tailed Student’s t-tests with Holm-Sidak’s multiple comparisons test (m) were used to assess the significance of differences between Vav1-cre; ScfGFP/fland ScfGFP/fl mice. (n) Competitive reconstitution assay in which 106 donor bone marrow cells from the indicated primary recipient mice were transplanted 4 weeks after irradiation along with recipient-type competitor cells into irradiated secondary recipient mice (n = 12 recipient mice/genotype from 3 independent experiments). The statistical significance of differences was assessed using two-way repeated measures ANOVAs with Sidak’s multiple comparisons tests. All data in this figure represent mean ± s.d.

Supplementary Figure 5 Percentage of LepR+ cells in Tomato+ bone marrow cells from Leprcre; R26tdTomato mice dropped after irradiation.

(ac) Confocal imaging of thin femur sections from Leprcre; R26tdTomato mice co-stained with anti-LepR and anti-perilipin antibodies. (Representative results from 3 independent experiments). (d) Quantification of the percentage of LepR+ cells in Tomato+ bone marrow cells from Leprcre; R26tdTomato mice dropped after irradiation. Data represent mean ± s.d. from n = 3 mice in 3 independent experiments. A one-way ANOVA with Tukey’s multiple comparisons test was used to assess statistical significance (P < 0.05,  or ###P < 0.001). (e) Western blot of SCF protein levels in the bone marrow of Adipoq-cre/ER; ScfGFP/fl mice, Leprcre; ScfGFP/fl mice, and ScfGFP/fl controls, either non-irradiated (Con) or 2 weeks after irradiation and transplantation of wild-type bone marrow cells (n = 3 mice/genotype from 3 independent experiments).

Supplementary Figure 6 Adipoq-Cre/ER+ CFU-F had increased adipogenic activity as compared to Adipoq-Cre/ER CFU-F in culture.

(a,b) Representative images of CFU-F colonies derived from Tomato (a) or Tomato+ bone marrow stromal cells (b) from Adipoq-cre/ER; R26tdTomato mice. The colonies were stained with anti-perilipin antibody after culturing in DMEM plus 20% fetal bovine serum for one week. The average number of perilipin+ adipocytes that spontaneously differentiated per CFU-F colony was quantified in Fig. 4j. (c,d) Most bone marrow adipocytes that formed after irradiation were Tomato+ in Adipoq-cre/ER; R26tdTomato mice that had been administered tamoxifen 2 weeks before irradiation (n = 4 mice/time point from 3 independent experiments). (e) Whole-mount imaging of a thick femur section 12 days after 5-FU treatment (representative image from n = 3 mice from 3 independent experiments).

Supplementary Figure 7 We did not detect any effect of Scf deletion on the morphology or frequency of megakaryocytes, endothelial cells, or blood vessels in the bone marrow.

(a,b) Few CD41+ megakaryocyte lineage cells expressed c-kit in mechanically dissociated bone marrow cells (a) or in femur sections (b; results are representative of n = 3 mice analyzed in 3 independent experiments). (c,d) Few endothelial cells expressed c-kit in enzymatically dissociated bone marrow cells (c) or in blood vessels identified in bone marrow sections by laminin staining (d; n = 3 mice from 3 independent experiments). (eh) The morphologies and frequencies of megakaryocytes, endothelial cells, and blood vessels in the bone marrow did not significantly differ between Adipoq-cre/ER; ScfGFP/fl mice (mut) and ScfGFP/fl controls (con) at 4 weeks after irradiation and bone marrow transplantation. Two-tailed Student’s t-tests were used to assess the significance of differences between genotypes but the differences were not statistically significant (n = 5 mice per genotype analyzed in 3 independent experiments). (ik) Diphtheria toxin ablated Tomato+ cells in the bone marrow of Adipoq-cre/ER; R26iDTR; R26tdTomato mice at 3 days after treatment but adipocytes regenerated from unrecombined Tomato negative progenitors within 14 days after treatment (n = 5 mice per genotype analyzed in 3 independent experiments). All data in this figure represent mean ± s.d.

Supplementary Figure 8

Unprocessed scans of western-blots from Supplementary Fig. 5e.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1400 kb)

Supplementary Information

Supplementary Information (PDF 78 kb)

Supplementary Table 1

Supplementary Information (XLSX 11 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, B., Yu, H., Yue, R. et al. Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF. Nat Cell Biol 19, 891–903 (2017). https://doi.org/10.1038/ncb3570

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3570

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