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:

A PML–PPAR-δ pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance

Abstract

Stem-cell function is an exquisitely regulated process. Thus far, the contribution of metabolic cues to stem-cell function has not been well understood. Here we identify a previously unknown promyelocytic leukemia (PML)–peroxisome proliferator-activated receptor δ (PPAR-δ)–fatty-acid oxidation (FAO) pathway for the maintenance of hematopoietic stem cells (HSCs). We have found that loss of PPAR-δ or inhibition of mitochondrial FAO induces loss of HSC maintenance, whereas treatment with PPAR-δ agonists improved HSC maintenance. PML exerts its essential role in HSC maintenance through regulation of PPAR signaling and FAO. Mechanistically, the PML–PPAR-δ–FAO pathway controls the asymmetric division of HSCs. Deletion of Ppard or Pml as well as inhibition of FAO results in the symmetric commitment of HSC daughter cells, whereas PPAR-δ activation increased asymmetric cell division. Thus, our findings identify a metabolic switch for the control of HSC cell fate with potential therapeutic implications.

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: PPAR-δ is essential for HSC maintenance.
Figure 2: Pharmacological activation of PPAR-δ enhances HSC maintenance.
Figure 3: Pharmacological inhibition of mitochondrial FAO with etomoxir induces HSC exhaustion.
Figure 4: Pharmacological activation of PPAR-δ rescues the maintenance defect of Pml-deleted HSCs.
Figure 5: An immunophenotyping assay to characterize asymmetric division in HSCs.
Figure 6: PPAR-δ and FAO regulate asymmetric division in the HSC compartment.

Similar content being viewed by others

References

  1. Ergen, A.V. & Goodell, M.A. Mechanisms of hematopoietic stem cell aging. Exp. Gerontol. 45, 286–290 (2010).

    Article  CAS  Google Scholar 

  2. 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 

  3. Fuchs, E., Tumbar, T. & Guasch, G. Socializing with the neighbors: stem cells and their niche. Cell 116, 769–778 (2004).

    Article  CAS  Google Scholar 

  4. Lemischka, I.R. & Moore, K.A. Stem cells: interactive niches. Nature 425, 778–779 (2003).

    Article  CAS  Google Scholar 

  5. 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 

  6. Wilson, A. & Trumpp, A. Bone-marrow haematopoietic-stem-cell niches. Nat. Rev. Immunol. 6, 93–106 (2006).

    Article  CAS  Google Scholar 

  7. Morrison, S.J. & Kimble, J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 441, 1068–1074 (2006).

    Article  CAS  Google Scholar 

  8. Lansdorp, P.M. Self-renewal of stem cells. Biol. Blood Marrow Transplant. 3, 171–178 (1997).

    CAS  Google Scholar 

  9. Metcalf, D. Cell-cell signalling in the regulation of blood cell formation and function. Immunol. Cell Biol. 76, 441–447 (1998).

    Article  CAS  Google Scholar 

  10. Ogawa, M. Stochastic model revisited. Int. J. Hematol. 69, 2–5 (1999).

    CAS  Google Scholar 

  11. Michalik, L. et al. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol. Rev. 58, 726–741 (2006).

    Article  CAS  Google Scholar 

  12. Braissant, O., Foufelle, F., Scotto, C., Dauca, M. & Wahli, W. Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-α, -β and -γ in the adult rat. Endocrinology 137, 354–366 (1996).

    Article  CAS  Google Scholar 

  13. Takahashi, S., Tanaka, T. & Sakai, J. New therapeutic target for metabolic syndrome: PPARδ. Endocr. J. 54, 347–357 (2007).

    Article  CAS  Google Scholar 

  14. Clapham, J.C. & Arch, J.R. Thermogenic and metabolic antiobesity drugs: rationale and opportunities. Diabetes Obes. Metab. 9, 259–275 (2007).

    Article  CAS  Google Scholar 

  15. Narkar, V.A. et al. AMPK and PPARδ agonists are exercise mimetics. Cell 134, 405–415 (2008).

    Article  CAS  Google Scholar 

  16. Ito, K. et al. PML targeting eradicates quiescent leukaemia-initiating cells. Nature 453, 1072–1078 (2008).

    Article  CAS  Google Scholar 

  17. Bernardi, R. & Pandolfi, P.P. Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nat. Rev. Mol. Cell Biol. 8, 1006–1016 (2007).

    Article  CAS  Google Scholar 

  18. Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).

    Article  CAS  Google Scholar 

  19. Takubo, K. et al. Regulation of the HIF-1α level is essential for hematopoietic stem cells. Cell Stem Cell 7, 391–402 (2010).

    Article  CAS  Google Scholar 

  20. Gan, B. et al. Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature 468, 701–704 (2010).

    Article  CAS  Google Scholar 

  21. Gurumurthy, S. et al. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature 468, 659–663 (2010).

    Article  CAS  Google Scholar 

  22. Nakada, D., Saunders, T.L. & Morrison, S.J. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 468, 653–658 (2010).

    Article  CAS  Google Scholar 

  23. Breems, D.A., Blokland, E.A., Neben, S. & Ploemacher, R.E. Frequency analysis of human primitive haematopoietic stem cell subsets using a cobblestone area forming cell assay. Leukemia 8, 1095–1104 (1994).

    CAS  Google Scholar 

  24. Peters, J.M. & Gonzalez, F.J. Sorting out the functional role(s) of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) in cell proliferation and cancer. Biochim. Biophys. Acta 1796, 230–241 (2009).

    CAS  Google Scholar 

  25. Tanaka, T. et al. Activation of peroxisome proliferator-activated receptor δ induces fatty acid β-oxidation in skeletal muscle and attenuates metabolic syndrome. Proc. Natl. Acad. Sci. USA 100, 15924–15929 (2003).

    Article  CAS  Google Scholar 

  26. Wurch, T., Junquero, D., Delhon, A. & Pauwels, J. Pharmacological analysis of wild-type α, γ and δ subtypes of the human peroxisome proliferator-activated receptor. Naunyn Schmiedebergs Arch. Pharmacol. 365, 133–140 (2002).

    Article  CAS  Google Scholar 

  27. Adams, G.B. et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature 439, 599–603 (2006).

    Article  CAS  Google Scholar 

  28. Lapidot, T., Dar, A. & Kollet, O. How do stem cells find their way home? Blood 106, 1901–1910 (2005).

    Article  CAS  Google Scholar 

  29. Wolf, N.S. Dissecting the hematopoietic microenvironment. I. Stem cell lodgment and commitment, and the proliferation and differentiation of erythropoietic descendants in the S1–S1d mouse. Cell Tissue Kinet. 7, 89–98 (1974).

    CAS  Google Scholar 

  30. Wang, Y.X. PPARs: diverse regulators in energy metabolism and metabolic diseases. Cell Res. 20, 124–137 (2010).

    Article  CAS  Google Scholar 

  31. Tutwiler, G.F., Brentzel, H.J. & Kiorpes, T.C. Inhibition of mitochondrial carnitine palmitoyl transferase A in vivo with methyl 2-tetradecylglycidate (methyl palmoxirate) and its relationship to ketonemia and glycemia. Proc. Soc. Exp. Biol. Med. 178, 288–296 (1985).

    Article  CAS  Google Scholar 

  32. Wolf, H.P. Possible new therapeutic approach in diabetes mellitus by inhibition of carnitine palmitoyltransferase 1 (CPT1). Horm. Metab. Res. Suppl. 26, 62–67 (1992).

    CAS  Google Scholar 

  33. Samudio, I. et al. Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J. Clin. Invest. 120, 142–156 (2010).

    Article  CAS  Google Scholar 

  34. Kitamura, Y.I. et al. FoxO1 protects against pancreatic beta cell failure through NeuroD and MafA induction. Cell Metab. 2, 153–163 (2005).

    Article  CAS  Google Scholar 

  35. Carracedo, A. et al. A metabolic prosurvival role for PML in breast cancer. J. Clin. Invest. 122 (2012).

  36. Carlesso, N., Aster, J.C., Sklar, J. & Scadden, D.T. Notch1-induced delay of human hematopoietic progenitor cell differentiation is associated with altered cell cycle kinetics. Blood 93, 838–848 (1999).

    CAS  Google Scholar 

  37. 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 

  38. Berger, J.P., Akiyama, T.E. & Meinke, P.T. PPARs: therapeutic targets for metabolic disease. Trends Pharmacol. Sci. 26, 244–251 (2005).

    Article  CAS  Google Scholar 

  39. Wagner, K.D. & Wagner, N. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) acts as regulator of metabolism linked to multiple cellular functions. Pharmacol. Ther. 125, 423–435 (2010).

    Article  CAS  Google Scholar 

  40. Risérus, U. et al. Activation of peroxisome proliferator-activated receptor (PPAR)δ promotes reversal of multiple metabolic abnormalities, reduces oxidative stress, and increases fatty acid oxidation in moderately obese men. Diabetes 57, 332–339 (2008).

    Article  Google Scholar 

  41. Samudio, I., Fiegl, M. & Andreeff, M. Mitochondrial uncoupling and the Warburg effect: molecular basis for the reprogramming of cancer cell metabolism. Cancer Res. 69, 2163–2166 (2009).

    Article  CAS  Google Scholar 

  42. Brown, N.F., Stefanovic-Racic, M., Sipula, I.J. & Perdomo, G. The mammalian target of rapamycin regulates lipid metabolism in primary cultures of rat hepatocytes. Metabolism 56, 1500–1507 (2007).

    Article  CAS  Google Scholar 

  43. Sipula, I.J., Brown, N.F. & Perdomo, G. Rapamycin-mediated inhibition of mammalian target of rapamycin in skeletal muscle cells reduces glucose utilization and increases fatty acid oxidation. Metabolism 55, 1637–1644 (2006).

    Article  CAS  Google Scholar 

  44. Sengupta, S., Peterson, T.R., Laplante, M., Oh, S. & Sabatini, D.M. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature 468, 1100–1104 (2010).

    Article  CAS  Google Scholar 

  45. Wu, M. et al. Imaging hematopoietic precursor division in real time. Cell Stem Cell 1, 541–554 (2007).

    Article  CAS  Google Scholar 

  46. Sahin, E. et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 470, 359–365 (2011).

    Article  CAS  Google Scholar 

  47. Schafer, Z.T. et al. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature 461, 109–113 (2009).

    Article  CAS  Google Scholar 

  48. Wang, Z.G. et al. Role of PML in cell growth and the retinoic acid pathway. Science 279, 1547–1551 (1998).

    Article  CAS  Google Scholar 

  49. Barak, Y. et al. Effects of peroxisome proliferator-activated receptor δ on placentation, adiposity, and colorectal cancer. Proc. Natl. Acad. Sci. USA 99, 303–308 (2002).

    Article  CAS  Google Scholar 

  50. Chambers, S.M. et al. Hematopoietic fingerprints: an expression database of stem cells and their progeny. Cell Stem Cell 1, 578–591 (2007).

    Article  CAS  Google Scholar 

  51. Alimonti, A. et al. Subtle variations in Pten dose determine cancer susceptibility. Nat. Genet. 42, 454–458 (2010).

    Article  CAS  Google Scholar 

  52. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005).

    Article  CAS  Google Scholar 

  53. Gerhart-Hines, Z. et al. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1α. EMBO J. 26, 1913–1923 (2007).

    Article  CAS  Google Scholar 

  54. Deberardinis, R.J., Lum, J.J. & Thompson, C.B. Phosphatidylinositol 3-kinase–dependent modulation of carnitine palmitoyltransferase 1A expression regulates lipid metabolism during hematopoietic cell growth. J. Biol. Chem. 281, 37372–37380 (2006).

    Article  CAS  Google Scholar 

  55. Pike, L.S., Smift, A.L., Croteau, N.J., Ferrick, D.A. & Wu, M. Inhibition of fatty acid oxidation by etomoxir impairs NADPH production and increases reactive oxygen species resulting in ATP depletion and cell death in human glioblastoma cells. Biochim. Biophys. Acta 1807, 726–734 (2011).

    Article  CAS  Google Scholar 

  56. Ferrick, D.A., Neilson, A. & Beeson, C. Advances in measuring cellular bioenergetics using extracellular flux. Drug Discov. Today 13, 268–274 (2008).

    Article  CAS  Google Scholar 

  57. Iwama, A. et al. Enhanced self-renewal of hematopoietic stem cells mediated by the polycomb gene product Bmi-1. Immunity 21, 843–851 (2004).

    Article  CAS  Google Scholar 

  58. Matsuzaki, Y., Kinjo, K., Mulligan, R.C. & Okano, H. Unexpectedly efficient homing capacity of purified murine hematopoietic stem cells. Immunity 20, 87–93 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all the members of the Pandolfi laboratory for comments and discussion. We especially thank K. Hosokawa for technical advice on asymmetric division, M. Alberich-Jorda and A.M. Zubiaga for technical help and critical discussions, and P. Puigserver and M.C. Haigis for technical support with the metabolic analysis. K.I. was supported by a K99 US National Institutes of Health (NIH) grant. The work of A.C. was supported by EMBO, the Ramón y Cajal award (Spanish Ministry of Education), Instituto de Salud Carlos III (PI10/01484), the Marie Curie Reintegration grant (277043) and the Basque Government (PI2012-03). This work was supported by NIH grants to P.P.P.

Author information

Authors and Affiliations

Authors

Contributions

K.I., A.C. and P.P.P. conceived of and designed the experiments. K.I., A.C. and D.W. performed the experiments. F.A. and T.S. contributed to the setup of the asymmetric division experiments. U.A. performed the statistical analyses. Z.T.S. helped with the approach of ATP measurement. R.M.E. and C.-H.L. provided Ppard conditional knockout mice. D.E.A. provided expertise in hematology and helped with hematological analysis. K.I., A.C. and P.P.P. analyzed data. K.I., A.C. and P.P.P. wrote the paper.

Corresponding author

Correspondence to Pier Paolo Pandolfi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Tables 1 and 2 (PDF 1506 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ito, K., Carracedo, A., Weiss, D. et al. A PML–PPAR-δ pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance. Nat Med 18, 1350–1358 (2012). https://doi.org/10.1038/nm.2882

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2882

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