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:

Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia

Abstract

Acute promyelocytic leukemia (APL), a cytogenetically distinct subtype of acute myeloid leukemia (AML), characterized by the t(15;17)-associated PML-RARA fusion, has been successfully treated with therapy utilizing all-trans-retinoic acid (ATRA) to differentiate leukemic blasts. However, among patients with non-APL AML, ATRA-based treatment has not been effective. Here we show that, through epigenetic reprogramming, inhibitors of lysine-specific demethylase 1 (LSD1, also called KDM1A), including tranylcypromine (TCP), unlocked the ATRA-driven therapeutic response in non-APL AML. LSD1 inhibition did not lead to a large-scale increase in histone 3 Lys4 dimethylation (H3K4me2) across the genome, but it did increase H3K4me2 and expression of myeloid-differentiation–associated genes. Notably, treatment with ATRA plus TCP markedly diminished the engraftment of primary human AML cells in vivo in nonobese diabetic (NOD)-severe combined immunodeficient (SCID) mice, suggesting that ATRA in combination with TCP may target leukemia-initiating cells. Furthermore, initiation of ATRA plus TCP treatment 15 d after engraftment of human AML cells in NOD-SCID γ (with interleukin-2 (IL-2) receptor γ chain deficiency) mice also revealed the ATRA plus TCP drug combination to have a potent anti-leukemic effect that was superior to treatment with either drug alone. These data identify LSD1 as a therapeutic target and strongly suggest that it may contribute to AML pathogenesis by inhibiting the normal pro-differentiative function of ATRA, paving the way for new combinatorial therapies for AML.

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: LSD1 inhibitors potentiate ATRA-induced differentiation of AML cells.
Figure 2: Treatment with ATRA and TCP together diminishes the engraftment of primary AML samples.
Figure 3: Treatment with ATRA plus TCP enhances the expression of a subset of genes associated with the myeloid differentiation pathway.
Figure 4: Gene-specific increases in H3K4me2 induced by treatment with ATRA plus TCP correlate with the upregulation of myeloid-differentiation–associated gene expression.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Pollyea, D.A., Kohrt, H.E. & Medeiros, B.C. Acute myeloid leukaemia in the elderly: a review. Br. J. Haematol. 152, 524–542 (2011).

    Article  Google Scholar 

  2. Krug, U. et al. Complete remission and early death after intensive chemotherapy in patients aged 60 years or older with acute myeloid leukaemia: a web-based application for prediction of outcomes. Lancet 376, 2000–2008 (2010).

    Article  CAS  Google Scholar 

  3. Altucci, L. & Gronemeyer, H. The promise of retinoids to fight against cancer. Nat. Rev. Cancer 1, 181–193 (2001).

    Article  CAS  Google Scholar 

  4. Petrie, K., Zelent, A. & Waxman, S. Differentiation therapy of acute myeloid leukemia: past, present and future. Curr. Opin. Hematol. 16, 84–91 (2009).

    Article  CAS  Google Scholar 

  5. Altucci, L. et al. Retinoic acid–induced apoptosis in leukemia cells is mediated by paracrine action of tumor-selective death ligand TRAIL. Nat. Med. 7, 680–686 (2001).

    Article  CAS  Google Scholar 

  6. Glasow, A., Prodromou, N., Xu, K., von Lindern, M. & Zelent, A. Retinoids and myelomonocytic growth factors cooperatively activate RARA and induce human myeloid leukemia cell differentiation via MAP kinase pathways. Blood 105, 341–349 (2005).

    Article  CAS  Google Scholar 

  7. Glasow, A. et al. DNA methylation-independent loss of RARA gene expression in acute myeloid leukemia. Blood 111, 2374–2377 (2008).

    Article  CAS  Google Scholar 

  8. Bernstein, B.E. et al. Genomic maps and comparative analysis of histone modifications in human and mouse. Cell 120, 169–181 (2005).

    Article  CAS  Google Scholar 

  9. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).

    Article  CAS  Google Scholar 

  10. Berglund, L. et al. A genecentric Human Protein Atlas for expression profiles based on antibodies. Mol. Cell. Proteomics 7, 2019–2027 (2008).

    Article  CAS  Google Scholar 

  11. Lim, S. et al. Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology. Carcinogenesis 31, 512–520 (2010).

    Article  CAS  Google Scholar 

  12. Hayami, S. et al. Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers. Int. J. Cancer 128, 574–586 (2011).

    Article  CAS  Google Scholar 

  13. Lee, M.G., Wynder, C., Schmidt, D.M., McCafferty, D.G. & Shiekhattar, R. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chem. Biol. 13, 563–567 (2006).

    Article  CAS  Google Scholar 

  14. Huang, Y. et al. Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc. Natl. Acad. Sci. USA 104, 8023–8028 (2007).

    Article  CAS  Google Scholar 

  15. Dalton, W.T. Jr. et al. HL-60 cell line was derived from a patient with FAB-M2 and not FAB-M3. Blood 71, 242–247 (1988).

    PubMed  Google Scholar 

  16. Warner, J.K. et al. Direct evidence for cooperating genetic events in the leukemic transformation of normal human hematopoietic cells. Leukemia 19, 1794–1805 (2005).

    Article  CAS  Google Scholar 

  17. Wang, H., Zheng, X., Behm, F.G. & Ratnam, M. Differentiation-independent retinoid induction of folate receptor type β, a potential tumor target in myeloid leukemia. Blood 96, 3529–3536 (2000).

    CAS  PubMed  Google Scholar 

  18. Wolf, D. & Rotter, V. Major deletions in the gene encoding the p53 tumor antigen cause lack of p53 expression in HL-60 cells. Proc. Natl. Acad. Sci. USA 82, 790–794 (1985).

    Article  CAS  Google Scholar 

  19. McDermott, S.P. et al. A small molecule screening strategy with validation on human leukemia stem cells uncovers the therapeutic efficacy of kinetin riboside. Blood 119, 1200–1207 (2012).

    Article  CAS  Google Scholar 

  20. Howell, A.L., Stukel, T.A., Bloomfield, C.D., Davey, F.R. & Ball, E.D. Induction of differentiation in blast cells and leukemia colony-forming cells from patients with acute myeloid leukemia. Blood 75, 721–729 (1990).

    CAS  PubMed  Google Scholar 

  21. 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  Google Scholar 

  22. Lapidot, T. et al. Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice. Science 255, 1137–1141 (1992).

    Article  CAS  Google Scholar 

  23. Doulatov, S. et al. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nat. Immunol. 11, 585–593 (2010).

    Article  CAS  Google Scholar 

  24. McKenzie, J.L., Gan, O.I., Doedens, M., Wang, J.C. & Dick, J.E. Individual stem cells with highly variable proliferation and self-renewal properties comprise the human hematopoietic stem cell compartment. Nat. Immunol. 7, 1225–1233 (2006).

    Article  CAS  Google Scholar 

  25. Yin, B. et al. A retroviral mutagenesis screen reveals strong cooperation between Bcl11a overexpression and loss of the Nf1 tumor suppressor gene. Blood 113, 1075–1085 (2009).

    Article  CAS  Google Scholar 

  26. Milojkovic, D. et al. Antiapoptotic microenvironment of acute myeloid leukemia. J. Immunol. 173, 6745–6752 (2004).

    Article  CAS  Google Scholar 

  27. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011).

    Article  CAS  Google Scholar 

  28. Wang, J. et al. The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat. Genet. 41, 125–129 (2009).

    Article  CAS  Google Scholar 

  29. Florean, C., Schnekenburger, M., Grandjenette, C., Dicato, M. & Diederich, M. Epigenomics of leukemia: from mechanisms to therapeutic applications. Epigenomics 3, 581–609 (2011).

    Article  CAS  Google Scholar 

  30. Binda, C. et al. Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2. J. Am. Chem. Soc. 132, 6827–6833 (2010).

    Article  CAS  Google Scholar 

  31. Ueda, R. et al. Identification of cell-active lysine specific demethylase 1-selective inhibitors. J. Am. Chem. Soc. 131, 17536–17537 (2009).

    Article  CAS  Google Scholar 

  32. Culhane, J.C., Wang, D., Yen, P.M. & Cole, P.A. Comparative analysis of small molecules and histone substrate analogues as LSD1 lysine demethylase inhibitors. J. Am. Chem. Soc. 132, 3164–3176 (2010).

    Article  CAS  Google Scholar 

  33. Huang, Y. et al. Novel oligoamine analogues inhibit lysine-specific demethylase 1 and induce reexpression of epigenetically silenced genes. Clin. Cancer Res. 15, 7217–7228 (2009).

    Article  CAS  Google Scholar 

  34. Sharma, S.K. et al. (Bis)urea and (bis)thiourea inhibitors of lysine-specific demethylase 1 as epigenetic modulators. J. Med. Chem. 53, 5197–5212 (2010).

    Article  CAS  Google Scholar 

  35. Whyte, I.M. Monoamineoxidase inhibitors. in Medical Toxicology (ed. Dart, R.C.) 823–833 (Lippincott Williams and Wilkins, Philadelphia, Pennsylvania, USA, 2003).

  36. McKenzie, J.L., Gan, O.I., Doedens, M. & Dick, J.E. Reversible cell surface expression of CD38 on CD34-positive human hematopoietic repopulating cells. Exp. Hematol. 35, 1429–1436 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

T.S., L.H., K.P. and A.Z. were supported by a Specialist Programme Grant from Leukaemia and Lymphoma Research. T.S., L.H., K.P. and A.Z. were also supported in part by the Samuel Waxman Cancer Research Foundation. S.G. and C.M.-T. were supported by the Interdisziplinäres Zentrum für Klinische Forschung (IZKF) and the Deutsche Forschungsgemeinschaft (1328/6-1, 8-1 and 9-1). W.C.C., L.J., J.C.Y.W. and J.E.D. were supported by the Cancer Stem Cell Consortium with funding from the Government of Canada through Genome Canada and the Ontario Genomics Institute (OGI-047) and through the Canadian Institute of Health Research (CSC-105367), as well as with funding from Canadian Institutes for Health Research (CIHR), the Canadian Cancer Society Research Institute, the Terry Fox Foundation; Genome Canada through the Ontario Genomics Institute; Ontario Institute for Cancer Research with funds from the province of Ontario; and a Canada Research Chair. W.C.C., L.J., J.C.Y.W. and J.E.D. were also funded in part by the Ontario Ministry of Health and Long Term Care (OMOHLTC). The views expressed here do not necessarily reflect those of the OMOHLTC. R.A.C. Jr. was supported by a National Cancer Institute Grant (CA51085). The authors would like to thank D. Leongamornlert, L. Jasnos and S. Bashir for valuable discussions on the interpretation of the ChIP-Seq data and the statistical analyses. The authors would also like to thank M. Greaves for support and critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

T.S. performed cell drug treatments, ChIP-Seq, quantitative PCR, RNAi and western blotting; L.H. performed cell drug treatments, FACS, the superoxide assay, immunohistochemical staining and light microscopy; S.G. and C.M.-T. performed ChIP-Seq and primary AML sample drug treatments; W.C.C., L.J., A.C.P., J.C.Y.W. and J.E.D. performed in vivo treatments of AML in NOD-SCID and NSG mice; K.H., H.-U.K. and M.D. performed bioinformatic analyses of expression microarray and ChIP-Seq data; A.B., K.M. and M.D.M. isolated primary AML samples; P.W., L.M. and R.A.C. Jr. collaborated on the 2d LSD1 inhibitor; T.S., K.P. and A.Z. designed the study and analyzed the data; K.P. and A.Z. wrote the paper. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Arthur Zelent.

Ethics declarations

Competing interests

L.M. was employed by Progen Pharmaceuticals. R.A.C. and P.W. have received funding from Progen Pharmaceuticals.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11, Supplementary Tables 1–3 and Supplementary Methods (PDF 4269 kb)

Supplementary Data 1 (XLS 45 kb)

Supplementary Data 2 (XLS 483 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schenk, T., Chen, W., Göllner, S. et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med 18, 605–611 (2012). https://doi.org/10.1038/nm.2661

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2661

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