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:

Acute myeloid leukaemia disrupts endogenous myelo-erythropoiesis by compromising the adipocyte bone marrow niche

Abstract

Acute myeloid leukaemia (AML) is distinguished by the generation of dysfunctional leukaemic blasts, and patients characteristically suffer from fatal infections and anaemia due to insufficient normal myelo-erythropoiesis. Direct physical crowding of bone marrow (BM) by accumulating leukaemic cells does not fully account for this haematopoietic failure. Here, analyses from AML patients were applied to both in vitro co-culture platforms and in vivo xenograft modelling, revealing that human AML disease specifically disrupts the adipocytic niche in BM. Leukaemic suppression of BM adipocytes led to imbalanced regulation of endogenous haematopoietic stem and progenitor cells, resulting in impaired myelo-erythroid maturation. In vivo administration of PPARγ agonists induced BM adipogenesis, which rescued healthy haematopoietic maturation while repressing leukaemic growth. Our study identifies a previously unappreciated axis between BM adipogenesis and normal myelo-erythroid maturation that is therapeutically accessible to improve symptoms of BM failure in AML via non-cell autonomous targeting of the niche.

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: Healthy myelo-erythroid populations are suppressed in human AML patients and patient-derived xenografts.
Figure 2: AML disrupts the composition of the haematopoietic niche by reducing BM adipocytes.
Figure 3: Adipocytes localized within red BM are uniquely sensitive to the deleterious effects of AML.
Figure 4: AML patient BM-MSCs are biased against adipogenic differentiation.
Figure 5: BM adipocytes support healthy myelo-erythroid maturation but inhibit leukaemic progenitors.
Figure 6: The preservation of BM adipocytes improves human myelo-erythropoiesis in conditions of leukaemic disease.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Dohner, H. et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 115, 453–474 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Estey, E. & Dohner, H. Acute myeloid leukaemia. Lancet 368, 1894–1907 (2006).

    Article  PubMed  Google Scholar 

  3. Sasine, J. P. & Schiller, G. J. Acute myeloid leukemia: how do we measure success? Curr. Hematol. Malig. Rep. 6, 528–536 (2016).

    Article  Google Scholar 

  4. Boyd, A. L. et al. Niche displacement of human leukemic stem cells uniquely allows their competitive replacement with healthy HSPCs. J. Exp. Med. 211, 1925–1935 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Glait-Santar, C. et al. Functional niche competition between normal hematopoietic stem and progenitor cells and myeloid leukemia cells. Stem Cells 33, 3635–3642 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Boyd, A. L. & Bhatia, M. Bone marrow localization and functional properties of human hematopoietic stem cells. Curr. Opin. Hematol. 21, 249–255 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Morrison, S. J. & Scadden, D. T. The bone marrow niche for haematopoietic stem cells. Nature 505, 327–334 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. Schepers, K. et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 13, 285–299 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3, 730–737 (1997).

    Article  CAS  PubMed  Google Scholar 

  11. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994).

    Article  CAS  PubMed  Google Scholar 

  12. Miraki-Moud, F. et al. Acute myeloid leukemia does not deplete normal hematopoietic stem cells but induces cytopenias by impeding their differentiation. Proc. Natl Acad. Sci. USA 110, 13576–13581 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Goyama, S., Wunderlich, M. & Mulloy, J. C. Xenograft models for normal and malignant stem cells. Blood 125, 2630–2640 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Shafat, M. S. et al. Leukemic blasts program bone marrow adipocytes to generate a protumoral microenvironment. Blood 129, 1320–1332 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Ye, H. et al. Leukemic stem cells evade chemotherapy by metabolic adaptation to an adipose tissue niche. Cell Stem Cell 19, 23–37 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Scheller, E. L., Cawthorn, W. P., Burr, A. A., Horowitz, M. C. & MacDougald, O. A. Marrow adipose tissue: trimming the fat. Trends Endocrinol. Metab. 27, 392–403 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Scheller, E. L. et al. Region-specific variation in the properties of skeletal adipocytes reveals regulated and constitutive marrow adipose tissues. Nat. Commun. 6, 7808 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Spalding, K. L. et al. Dynamics of fat cell turnover in humans. Nature 453, 783–787 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. Medyouf, H. The microenvironment in human myeloid malignancies: emerging concepts and therapeutic implications. Blood 129, 1617–1626 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Guezguez, B. et al. Regional localization within the bone marrow influences the functional capacity of human HSCs. Cell Stem Cell 13, 175–189 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Xie, Y. et al. Detection of functional haematopoietic stem cell niche using real-time imaging. Nature 457, 97–101 (2009).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Frisch, B. J. et al. Functional inhibition of osteoblastic cells in an in vivo mouse model of myeloid leukemia. Blood 119, 540–550 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Krevvata, M. et al. Inhibition of leukemia cell engraftment and disease progression in mice by osteoblasts. Blood 124, 2834–2846 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Chen, Q. et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 23, 1128–1139 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kubo, H. et al. Identification of mesenchymal stem cell (MSC)-transcription factors by microarray and knockdown analyses, and signature molecule-marked MSC in bone marrow by immunohistochemistry. Genes Cells 14, 407–424 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Medyouf, H. et al. Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit. Cell Stem Cell 14, 824–837 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Pearce, D. J. et al. AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML. Blood 107, 1166–1173 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kode, A. et al. Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts. Nature 506, 240–244 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Takam Kamga, P. et al. Notch signalling drives bone marrow stromal cell-mediated chemoresistance in acute myeloid leukemia. Oncotarget 7, 21713–21727 (2016).

    PubMed  Google Scholar 

  34. Lu, H., Ward, M. G., Adeola, O. & Ajuwon, K. M. Regulation of adipocyte differentiation and gene expression-crosstalk between TGFβ and wnt signaling pathways. Mol. Biol. Rep. 40, 5237–5245 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Wagegg, M. et al. Hypoxia promotes osteogenesis but suppresses adipogenesis of human mesenchymal stromal cells in a hypoxia-inducible factor-1 dependent manner. PLoS ONE 7, e46483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Moura, I. C., Hermine, O., Lacombe, C. & Mayeux, P. Erythropoiesis and transferrin receptors. Curr. Opin. Hematol. 22, 193–198 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Ganz, T. & Nemeth, E. Iron metabolism: interactions with normal and disordered erythropoiesis. Cold Spring Harb. Perspect. Med. 2, a011668 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hardouin, P., Rharass, T. & Lucas, S. Bone marrow adipose tissue: to be or not to be a typical adipose tissue? Front. Endocrinol. 7, 85 (2016).

    Article  Google Scholar 

  39. Brown, K. K. et al. A novel N-aryl tyrosine activator of peroxisome proliferator-activated receptor-gamma reverses the diabetic phenotype of the Zucker diabetic fatty rat. Diabetes 48, 1415–1424 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Faber, K. et al. CDX2-driven leukemogenesis involves KLF4 repression and deregulated PPARγ signaling. J. Clin. Invest. 123, 299–314 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Konopleva, M. et al. Peroxisome proliferator-activated receptor gamma and retinoid X receptor ligands are potent inducers of differentiation and apoptosis in leukemias. Mol. Cancer Ther. 3, 1249–1262 (2004).

    CAS  PubMed  Google Scholar 

  42. Tsao, T. et al. Role of peroxisome proliferator-activated receptor-γ and its coactivator DRIP205 in cellular responses to CDDO (RTA-401) in acute myelogenous leukemia. Cancer Res. 70, 4949–4960 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hartwell, K. A. et al. Niche-based screening identifies small-molecule inhibitors of leukemia stem cells. Nat. Chem. Biol. 9, 840–848 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Elghetany, M. T., Ge, Y., Patel, J., Martinez, J. & Uhrova, H. Flow cytometric study of neutrophilic granulopoiesis in normal bone marrow using an expanded panel of antibodies: correlation with morphologic assessments. J. Clin. Lab. Anal. 18, 36–41 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lakschevitz, F. S. et al. Identification of neutrophil surface marker changes in health and inflammation using high-throughput screening flow cytometry. Exp. Cell Res. 342, 200–209 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Ambrosi, T. H. et al. Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell 20, 771–784 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhou, B. O. et al. Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF. Nat. Cell Biol. 19, 891–903 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

  49. 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 

  50. Yu, V. W. et al. Distinctive mesenchymal-parenchymal cell pairings govern B cell differentiation in the bone marrow. Stem Cell Rep. 7, 220–235 (2016).

    Article  CAS  Google Scholar 

  51. Panaroni, C. & Wu, J. Y. Interactions between B lymphocytes and the osteoblast lineage in bone marrow. Calcif. Tissue Int. 93, 261–268 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Adler, B. J., Green, D. E., Pagnotti, G. M., Chan, M. E. & Rubin, C. T. High fat diet rapidly suppresses B lymphopoiesis by disrupting the supportive capacity of the bone marrow niche. PLoS ONE 9, e90639 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Doucette, C. R. et al. A high fat diet increases bone marrow adipose tissue (MAT) but does not alter trabecular or cortical bone mass in C57BL/6J mice. J. Cell. Physiol. 230, 2032–2037 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sahdo, B. et al. Body temperature during hibernation is highly correlated with a decrease in circulating innate immune cells in the brown bear (Ursus arctos): a common feature among hibernators? Int. J. Med. Sci. 10, 508–514 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Boyd, A. L., Salci, K. R., Shapovalova, Z., McIntyre, B. A. & Bhatia, M. Nonhematopoietic cells represent a more rational target of in vivo hedgehog signaling affecting normal or acute myeloid leukemia progenitors. Exp. Hematol. 41, 858–869 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Shultz, L. D. et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J. Immunol. 154, 180–191 (1995).

    CAS  PubMed  Google Scholar 

  57. Tersey, S. A. et al. Islet β-cell endoplasmic reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic mouse model. Diabetes 61, 818–827 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

  59. Pricola, K. L., Kuhn, N. Z., Haleem-Smith, H., Song, Y. & Tuan, R. S. Interleukin-6 maintains bone marrow-derived mesenchymal stem cell stemness by an ERK1/2-dependent mechanism. J. Cell. Biochem. 108, 577–588 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Fleury, A. et al. Hedgehog associated to microparticles inhibits adipocyte differentiation via a non-canonical pathway. Sci. Rep. 6, 23479 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Huang, Y. et al. γ-secretase inhibitor induces adipogenesis of adipose-derived stem cells by regulation of Notch and PPAR-γ. Cell Prolif. 43, 147–156 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Styner, M., Sen, B., Xie, Z., Case, N. & Rubin, J. Indomethacin promotes adipogenesis of mesenchymal stem cells through a cyclooxygenase independent mechanism. J. Cell. Biochem. 111, 1042–1050 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Yanik, S. C., Baker, A. H., Mann, K. K. & Schlezinger, J. J. Organotins are potent activators of PPARγ and adipocyte differentiation in bone marrow multipotent mesenchymal stromal cells. Toxicol. Sci. 122, 476–488 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Benoit, Y. D. et al. Cooperation between HNF-1α, Cdx2, and GATA-4 in initiating an enterocytic differentiation program in a normal human intestinal epithelial progenitor cell line. Am. J. Physiol. Gastrointest. Liver Physiol. 298, G504–G517 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by a research grant to M.B. from the Marta and Owen Boris Foundation and Canadian Cancer Society Research Institute. A.L.B. was supported by graduate research scholarships: Ontario Graduate Scholarship, National Science and Engineering Research Council (NSERC) and the Jans Graduate Scholarship in Stem Cell Research. J.C.R. was supported by a doctoral scholarship from the Canadian Institute of Health Research (CIHR) and K.R.S. was supported by the NSERC Create M3 program. Y.D.B. was funded by the Quebec Health Research Funds (FRQS) and CIHR, and holds a fellowship from the Cancer Research Society (CRS). M.B. is a Canada Research Chair in Stem Cell Biology and Regenerative Medicine. We acknowledge M. Graham for human sample processing, as well as A. Fiebig-Comyn, W. Whittaker, L. Robson and A. Scott for assistance with animal husbandry and in vivo experiments. We thank L. May, M. J. Smith and M. Bruni for assistance with histology and immunofluorescence staining. We also thank C. Magruder and C. Hopkins for illustrative images.

Author information

Authors and Affiliations

Authors

Contributions

A.L.B., J.C.R., K.R.S., L.A., Y.D.B., Z.S., M.N., D.P.P. and C.J.V.C. performed experiments. M.F.J. performed microCT imaging and analysis. Z.S. performed bioinformatic analyses. M.A. performed clinical analyses. T.J.C. wrote custom image analysis scripts. C.A.R., R.F., B.L., D.S.A., M.S. and A.X. provided critical patient samples and clinical expertise. A.L.B. and M.B. interpreted data and wrote the manuscript. M.B. directed the study.

Corresponding author

Correspondence to Mickie Bhatia.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Healthy human myelo-erythroid cells are reproducibly suppressed by AML co-transplantation.

(ag) Experimental design (a), representative FACS plots (b,d,f) and quantification (c,e,g) of human populations in xenograft BM following healthy human HSPC transplantation alone or together with human AML. Data points represent group averages from n = 5 (e) or n = 6 (c,g) independent experiments. (h) Absolute BM cell number recovered by mechanical dissociation. Data points represent independent xenografted mice from 2 experiments (precise n values indicated in the figure). Findings are representative of 6 experiments. (il) Experimental design (i), representative FACS plots (j) and quantification (k,l) of healthy human CD45+CD33+ populations in xenograft BM following healthy human HSPC transplantation alone or together with human AML. (k) n = 37 (alone) and n = 35 (with AML) independent xenografted mice pooled from 6 experiments. (l) Data points represent group averages from n = 6 independent experiments. (mo) Experimental design (m) to compare competitive BM repopulation between HSPCs from two healthy human donors (analyzed in n,o). Constant cell doses from human Donor A were competitively transplanted against escalating doses of competitor cells from human Donor B. (n,o) FACS quantification of overall chimerism (n) and myeloid/lymphoid cell frequencies (o) specific to healthy human Donor A. Data represent n = 5 (low dose) and n = 4 (high dose) independent xenografted mice from 1 experiment, and reproduced in 3 additional experiments. (ps) Experimental design (p), representative FACS plots (q) and quantification (r,s) of healthy human CD45+CD34+ populations in xenograft BM following healthy human HSPC transplantation alone or together with human AML. (r) n = 34 (alone) and n = 32 (with AML) independent xenografted mice pooled from 6 experiments. (s) Data points represent group averages from n = 6 independent experiments. All FACS plots are representative of at least 5 independent experiments; blue coloring indicates healthy human cells, red indicates human AML cells, and grey indicates residual mouse BM cells. Quantification was based on patient and donor-specific HLA-A2 gating. Numbers within data points indicate AML Patient IDs. All data are means ± s.e.m. Statistical significance was assessed by Fisher’s Exact test (c), paired t-tests (e,g,l,s), unpaired t-tests (h,n,o), or Mann–Whitney U tests (k,r). Source data can be found in Supplementary Table 8.

Supplementary Figure 2 AML reduces adipocyte numbers more profoundly in BM versus non-BM sites.

(a) Absolute PLN+ adipocyte counts and DAPI+ nucleated cell counts per xenograft femur section. n = 9 (healthy) and n = 8 (AML) pooled from 5 experiments (values correspond to frequencies reported in Fig. 2h). (b) Representative images of xenograft femur sections showing that AML engraftment reduces the number of adipocytes per unit area, whereas nucleated cell counts remain stable. Scale bar, 70 μm. (c) Body weights as a function of human leukemic burden in BM. Values reflect n = 7, 2, 7, 7 independent xenografted mice (healthy human chimerism, 20% AML, 20–80% AML, and >80% AML) pooled from 3 experiments (1 healthy sample, AML Patients #2 and #6). (d,e) Nonfasted serum glucose levels in mice reconstituted with healthy human hematopoieisis or human AML (Patient #1). n = 5 biologically independent xenografted mice/group from 1 experiment (1 mouse with <20% AML, 3 mice with 20–80% AML, and 1 mouse with >80% AML). (f) Representative immunofluorescence images of inguinal fat pad tissue collected from mice engrafted with human AML (Patient #2) or matched non-transplanted controls. PLN, perilipin. Low magnification images (left) represent full tissue montage images of up to 347 fields (scale bar, 1.3 mm), while high magnification images (right), represent individual fields (scale bar, 70 μm). BM leukemic chimerism was >90%. (g,h) Adipocyte number (g) and cross-sectional area measurements (h) from fat pad sections shown in f. Area measurements reflect n = 73422 (control) and n = 46242 (AML) independent adipocytes from individual tissue sections. Analysis is representative of 3 independent mice/group from separate experiments. All data are means ± s.e.m. Statistical significance was assessed by unpaired t-tests (a,d), Kruskal–Wallis test (c), or Mann–Whitney U test (h). No statistical analyses were performed in e. Source data can be found in Supplementary Table 8.

Supplementary Figure 3 Osteoblasts and yellow marrow adipocytes persist in BM despite substantial levels of leukemic disease.

(a) Representative PLN immunofluorescence images of femur sections from AML-engrafted mice or non-transplanted controls, corresponding to high magnification images shown in Fig. 3e. Scale bar, 250 μm. (b,c) PLN quantification of absolute adipocyte numbers in tail vertebrae sections. N numbers (and data points) represent independent xenografted mice from 2 experiments (precise n values indicated in the figure). Data values for Patient #2 correspond to frequencies shown in Fig. 3g. (d) High magnification images of H&E-stained mouse femurs engrafted with healthy human hematopoietic cells or human AML (Patient #2). Chimerism levels are similar between healthy and AML grafts (>90% human chimerism). Yellow arrowheads indicate osteoblasts. Scale bar, 20 μm. (e) Immunofluorescence quantification of Osx + osteoblasts in femur sections from AML-engrafted mice (Patient #6) or healthy human HSPC-engrafted mice. n = 2 mice (healthy) and n = 4 mice (AML) from 1 experiment. This was reproduced in an additional experiment using an independent AML patient sample (Fig. 3k). (fh) MicroCT quantification of mineralized bone; additional parameters corresponding to data shown in Fig. 3l–n (AML-xenografts from Patient #2). n = 3 biologically independent mice/group from 1 experiment. All data are means ± s.e.m. Statistical significance was assessed by Mann–Whitney U test (b) or unpaired t-tests (c,fh). No statistical analyses were performed in e. Source data can be found in Supplementary Table 8.

Supplementary Figure 4 Characterization of MSCs and hematopoietic repopulating cells isolated from BM aspirates of healthy human donors or AML patients.

(a, b) Phase contrast images and FACS plots of MSCs isolated from the BM of healthy adult donors (a) and AML patients (b). Scale bars, 100 μm. (c) FACS analysis of human hematopoietic repopulation in the BM of mice transplanted with 10 × 106 BM mononuclear cells from AML Patients #12 or #13. Plots are representative of n = 4 (Patient#12), and n = 2 (Patient#13) independent xenografted mice, with similar results. (d,e) Heatmap (d) and bar graphs (e) showing the frequency of mature BODIPY+ adipocytes differentiated from healthy human BM-MSCs that were cultured in different cocktails of adipogenesis-inducing agents (variable concentrations of dexamethasone, IBMX, and insulin, together with a constant concentration of 2% FBS across all conditions). Each cocktail was tested in the presence or absence of 10 ng ml−1 IL-6 as a known inhibitor of adipogenesis59. Dashed lines indicate the final cocktail selected, based on the greatest sensitivity to detect inhibitory effects of IL-6. (d) Each heatmap value represents the average of two replicate wells. (e) Only values from 500 μM IBMX conditions are plotted. Source data can be found in Supplementary Table 8.

Supplementary Figure 5 In vivo GW1929 treatment expands adipose tissue and promotes healthy myelo-erythropoiesis.

(a) Schematic showing 4-day Transwell co-culture of healthy human Lin cells with healthy human BM-MSCs or differentiated human adipocytes (analysis in b,c). (b) FACS quantification of CD15+ granulocyte-lineage cells. Data represent group averages from n = 3 biologically independent experiments. (c) Quantification of human erythroid progenitors using methylcellulose CFU assays. n = 7 (MSCs) and n = 14 (adipocytes) independently assayed wells, pooled from 3 experiments. (d) Heatmap of probe sets most preferentially expressed in mature human osteoblasts versus adipocytes (GSE945129). (e) Normalized PPARγ transcript expression levels in mature human osteoblasts versus adipocytes (n = 3 biologically independent samples/group, GSE945129). (f) Experimental design to validate in vivo GW1929 dosing in non-transplanted mice (analysis in gk; all n = 5 (vehicle) and n = 4 (GW1929) independent mice from 1 experiment). (g,h) Effects of GW1929 treatment on previously reported39 PPARγ-sensitive parameters of fat mass (g) and non-fasted serum glucose (h). (ik) Whole femur dot plots (i) and image-based quantification of BM adipocyte frequency (j) and size (k). PLN, perilipin. (l) Experimental design to evaluate effects of in vivo GW1929 treatment on BM adipocytes and healthy human myelo-erythroid cells (analysis in m,n). (m,n) Correlation of BM adipocyte frequencies versus human erythroid colony number (m) or FACS-quantified healthy human CD45+CD15+ granulocytes (n). Data points represent n = 9 independently assayed wells (m) or n = 7 independent xenografted mice (n) from 1 experiment. Findings were reproduced in 3 additional experiments with independent healthy human cells (Fig. 6e, g). (o) Schematic showing 4-day in vitro culture of healthy human Lin cells in 0.1% DMSO (vehicle) or 20 μM GW1929 (analysis in p,q). (p) FACS quantification of CD15+ granulocyte-lineage cells. Data represent group averages from n = 6 biologically independent experiments. (q) Quantification of human erythroid progenitors using methylcellulose CFU assays. Data points represent n = 6 independently assayed wells/group, pooled from 2 experiments. All data are means ± s.e.m. Statistical significance was assessed by paired t-tests (b,p), Mann–Whitney U tests (c,q), two-tailed unpaired t-test (e), one-tailed unpaired t-tests (g,h,j,k), or Pearson’s correlation (m,n). Source data can be found in Supplementary Table 8.

Supplementary Figure 6 Adipogenesis-promoting therapy limits leukemic progression in a non-cell autonomous manner.

(a,b) Schematic (a) and FACS/Trypan blue quantification (b) of leukemic blasts disseminated to the spleen from BM progenitors. Human AML-xenografts (Patient #2) were treated with corn oil (vehicle) or 10 mg kg−1 GW1929, as outlined in Fig. 5m. n = 3 (vehicle) and n = 4 (GW1929) independent xenografted mice from 1 experiment. (c) FACS quantification of human AML dissemination in the BM and spleens of xenografted mice (Patient #2). Data points represent individual mice analyzed at various time points post-engraftment, from 1 experiment that was performed independently from drug treatment experiments in panel b. Findings have been replicated in 2 additional experiments. (d) Western blots showing PPARγ protein levels in human AML cells versus healthy human BM-MSCs and in vitro-differentiated human adipocytes. Unprocessed blots can be found in Supplementary Fig. 7. Images are representative of 3 independent Western blots, each with similar results. (e,f) Viability (left) and leukemic progenitor frequency (right) following overnight culture of human AML cells with 0.1% DMSO (0 μM GW1929) or 5–20 μM GW1929. N numbers (and data points) represent independently assayed wells shown separately for 2 experiments (precise n values are indicated in the figure). All data are means ± s.e.m. Statistical significance was assessed by unpaired t-test (b) or one-way ANOVA with Newman–Keuls Multiple Comparison Test (e,f). Source data can be found in Supplementary Table 8.

Supplementary Figure 7 Unprocessed Western blots.

In blots corresponding to Supplementary Fig. 6, tracks for samples from AML Patients #2 and #6 have been reflected along the vertical axis so that the sequence of the samples matches the order of their appearance in the text.

Supplementary information

Supplementary Information

Supplementary Information (PDF 4664 kb)

Life Sciences Reporting Summary (PDF 86 kb)

Supplementary Table 1

Supplementary Information (XLSX 12 kb)

Supplementary Table 2

Supplementary Information (XLSX 19 kb)

Supplementary Table 3

Supplementary Information (XLSX 48 kb)

Supplementary Table 4

Supplementary Information (XLSX 35 kb)

Supplementary Table 5

Supplementary Information (XLSX 43 kb)

Supplementary Table 6

Supplementary Information (XLSX 53 kb)

Supplementary Table 7

Supplementary Information (XLSX 49 kb)

Supplementary Table 8

Supplementary Information (XLSX 107 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boyd, A., Reid, J., Salci, K. et al. Acute myeloid leukaemia disrupts endogenous myelo-erythropoiesis by compromising the adipocyte bone marrow niche. Nat Cell Biol 19, 1336–1347 (2017). https://doi.org/10.1038/ncb3625

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3625

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer