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 leukemia

LSD1 inhibition by tranylcypromine derivatives interferes with GFI1-mediated repression of PU.1 target genes and induces differentiation in AML

A Correction to this article was published on 05 March 2019

This article has been updated

Abstract

LSD1 has emerged as a promising epigenetic target in the treatment of acute myeloid leukemia (AML). We used two murine AML models based on retroviral overexpression of Hoxa9/Meis1 (H9M) or MN1 to study LSD1 loss of function in AML. The conditional knockout of Lsd1 resulted in differentiation with both granulocytic and monocytic features and increased ATRA sensitivity and extended the survival of mice with H9M-driven AML. The conditional knockout led to an increased expression of multiple genes regulated by the important myeloid transcription factors GFI1 and PU.1. These include the transcription factors GFI1B and IRF8. We also compared the effect of different irreversible and reversible inhibitors of LSD1 in AML and could show that only tranylcypromine derivatives were capable of inducing a differentiation response. We employed a conditional knock-in model of inactive, mutant LSD1 to study the effect of only interfering with LSD1 enzymatic activity. While this was sufficient to initiate differentiation, it did not result in a survival benefit in mice. Hence, we believe that targeting both enzymatic and scaffolding functions of LSD1 is required to efficiently treat AML. This finding as well as the identified biomarkers may be relevant for the treatment of AML patients with LSD1 inhibitors.

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

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Change history

  • 05 March 2019

    The original version of this Article contained an error in the Acknowledgements section.

References

  1. Harris WJ, Huang X, Lynch JT, Spencer GJ, Hitchin JR, Li Y, et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell. 2012;21:473–87.

    PubMed  CAS  Google Scholar 

  2. Schenk T, Chen WC, Gollner S, Howell L, Jin L, Hebestreit K, et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med. 2012;18:605–11.

    PubMed  PubMed Central  CAS  Google Scholar 

  3. Mohammad HP, Smitheman KN, Kamat CD, Soong D, Federowicz KE, Van Aller GS, et al. A DNA hypomethylation signature predicts antitumor activity of LSD1 inhibitors in SCLC. Cancer Cell. 2015;28:57–69.

    PubMed  CAS  Google Scholar 

  4. Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004;119:941–53.

    PubMed  CAS  Google Scholar 

  5. Lee MG, Wynder C, Cooch N, Shiekhattar R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature. 2005;437:432–5.

    PubMed  CAS  Google Scholar 

  6. Shi YJ, Matson C, Lan F, Iwase S, Baba T, Shi Y. Regulation of LSD1 histone demethylase activity by its associated factors. Mol Cell. 2005;19:857–64.

    PubMed  CAS  Google Scholar 

  7. Wang Y, Zhang H, Chen Y, Sun Y, Yang F, Yu W, et al. LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell. 2009;138:660–72.

    PubMed  CAS  Google Scholar 

  8. Metzger E, Wissmann M, Yin N, Muller JM, Schneider R, Peters AH, et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature. 2005;437:436–9.

    PubMed  CAS  Google Scholar 

  9. Cai C, He HH, Gao S, Chen S, Yu Z, Gao Y, et al. Lysine-specific demethylase 1 has dual functions as a major regulator of androgen receptor transcriptional activity. Cell Rep. 2014;9:1618–27.

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Kerenyi MA, Shao Z, Hsu YJ, Guo G, Luc S, O’Brien K, et al. Histone demethylase Lsd1 represses hematopoietic stem and progenitor cell signatures during blood cell maturation. eLife. 2013;2:e00633.

    PubMed  PubMed Central  Google Scholar 

  11. Sprussel A, Schulte JH, Weber S, Necke M, Handschke K, Thor T, et al. Lysine-specific demethylase 1 restricts hematopoietic progenitor proliferation and is essential for terminal differentiation. Leukemia. 2012;26:2039–51.

    PubMed  CAS  Google Scholar 

  12. Morera L, Lubbert M, Jung M. Targeting histone methyltransferases and demethylases in clinical trials for cancer therapy. Clin Epigenetics. 2016;8:57.

    PubMed  PubMed Central  Google Scholar 

  13. Lee MG, Wynder C, Schmidt DM, McCafferty DG, Shiekhattar R. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chem Biol. 2006;13:563–7.

    PubMed  CAS  Google Scholar 

  14. Schmidt DM, McCafferty DG. trans-2-Phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1. Biochemistry. 2007;46:4408–16.

    PubMed  CAS  Google Scholar 

  15. Binda C, Valente S, Romanenghi M, Pilotto S, Cirilli R, Karytinos A, et al. Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2. J Am Chem Soc. 2010;132:6827–33.

    PubMed  CAS  Google Scholar 

  16. Huang Y, Stewart TM, Wu Y, Baylin SB, Marton LJ, Perkins B, et al. Novel oligoamine analogues inhibit lysine-specific demethylase 1 and induce reexpression of epigenetically silenced genes. Clin Cancer Res. 2009;15:7217–28.

    PubMed  PubMed Central  CAS  Google Scholar 

  17. Huang Y, Greene E, Murray Stewart T, Goodwin AC, Baylin SB, Woster PM, et al. Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc Natl Acad Sci USA. 2007;104:8023–8.

    PubMed  CAS  Google Scholar 

  18. Fu X, Zhang P, Yu B. Advances toward LSD1 inhibitors for cancer therapy. Future Med Chem. 2017;9:1227–42.

    PubMed  CAS  Google Scholar 

  19. Duteil D, Tosic M, Lausecker F, Nenseth HZ, Muller JM, Urban S, et al. Lsd1 ablation triggers metabolic reprogramming of brown adipose tissue. Cell Rep. 2016;17:1008–21.

    PubMed  PubMed Central  CAS  Google Scholar 

  20. Willmann D, Lim S, Wetzel S, Metzger E, Jandausch A, Wilk W, et al. Impairment of prostate cancer cell growth by a selective and reversible lysine-specific demethylase 1 inhibitor. Int J Cancer. 2012;131:2704–9.

    PubMed  CAS  Google Scholar 

  21. Hitchin JR, Blagg J, Burke R, Burns S, Cockerill MJ, Fairweather EE, et al. Development and evaluation of selective, reversible LSD1 inhibitors derived from fragments. MedChemComm . 2013;4:1513.

    CAS  Google Scholar 

  22. Fiskus W, Sharma S, Shah B, Portier BP, Devaraj SG, Liu K, et al. Highly effective combination of LSD1 (KDM1A) antagonist and pan-histone deacetylase inhibitor against human AML cells. Leukemia. 2014;28:2155–64.

    PubMed  PubMed Central  CAS  Google Scholar 

  23. Zhu D, Holz S, Metzger E, Pavlovic M, Jandausch A, Jilg C, et al. Lysine-specific demethylase 1 regulates differentiation onset and migration of trophoblast stem cells. Nat Commun. 2014;5:3174.

    PubMed  Google Scholar 

  24. Khandanpour C, Phelan JD, Vassen L, Schutte J, Chen R, Horman SR, et al. Growth factor independence 1 antagonizes a p53-induced DNA damage response pathway in lymphoblastic leukemia. Cancer Cell. 2013;23:200–14.

    PubMed  PubMed Central  CAS  Google Scholar 

  25. Kroon E, Krosl J, Thorsteinsdottir U, Baban S, Buchberg AM, Sauvageau G. Hoxa9 transforms primary bone marrow cells through specific collaboration with Meis1a but not Pbx1b. EMBO J. 1998;17:3714–25.

    PubMed  PubMed Central  CAS  Google Scholar 

  26. Pineault N, Abramovich C, Ohta H, Humphries RK. Differential and common leukemogenic potentials of multiple NUP98-Hox fusion proteins alone or with Meis1. Mol Cell Biol. 2004;24:1907–17.

    PubMed  PubMed Central  CAS  Google Scholar 

  27. Heuser M, Yun H, Berg T, Yung E, Argiropoulos B, Kuchenbauer F, et al. Cell of origin in AML: susceptibility to MN1-induced transformation is regulated by the MEIS1/AbdB-like HOX protein complex. Cancer Cell. 2011;20:39–52.

    PubMed  PubMed Central  CAS  Google Scholar 

  28. Heuser M, Argiropoulos B, Kuchenbauer F, Yung E, Piper J, Fung S, et al. MN1 overexpression induces acute myeloid leukemia in mice and predicts ATRA resistance in patients with AML. Blood. 2007;110:1639–47.

    PubMed  CAS  Google Scholar 

  29. Oellerich T, Oellerich MF, Engelke M, Munch S, Mohr S, Nimz M, et al. beta2 Integrin-derived signals induce cell survival and proliferation of AML blasts by activating a Syk/STAT signaling axis. Blood. 2013;121:3889–99. S1-66

    PubMed  CAS  Google Scholar 

  30. Wilson NK, Foster SD, Wang X, Knezevic K, Schutte J, Kaimakis P, et al. Combinatorial transcriptional control in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators. Cell Stem Cell. 2010;7:532–44.

    PubMed  CAS  Google Scholar 

  31. Hones JM, Botezatu L, Helness A, Vadnais C, Vassen L, Robert F, et al. GFI1 as a novel prognostic and therapeutic factor for AML/MDS. Leukemia. 2016;30:1237–45.

    PubMed  CAS  Google Scholar 

  32. Fraszczak J, Vadnais C, Rashkovan M, Ross J, Beauchemin H, Chen R. et al. Reduced expression but not deficiency of GFI1 causes a fatal myeloproliferative disease in mice. Leukemia. 2019;33:110–21.

    PubMed  CAS  Google Scholar 

  33. Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347:70–8.

    PubMed  CAS  Google Scholar 

  34. Saleque S, Kim J, Rooke HM, Orkin SH. Epigenetic regulation of hematopoietic differentiation by Gfi-1 and Gfi-1b is mediated by the cofactors CoREST and LSD1. Mol Cell. 2007;27:562–72.

    PubMed  CAS  Google Scholar 

  35. Roe JS, Mercan F, Rivera K, Pappin DJ, Vakoc CR. BET bromodomain inhibition suppresses the function of hematopoietic transcription factors in acute myeloid leukemia. Mol Cell. 2015;58:1028–39.

    PubMed  PubMed Central  CAS  Google Scholar 

  36. Khandanpour C, Krongold J, Schutte J, Bouwman F, Vassen L, Gaudreau MC, et al. The human GFI136N variant induces epigenetic changes at the Hoxa9 locus and accelerates K-RAS driven myeloproliferative disorder in mice. Blood. 2012;120:4006–17.

    PubMed  CAS  Google Scholar 

  37. Feng Z, Yao Y, Zhou C, Chen F, Wu F, Wei L, et al. Pharmacological inhibition of LSD1 for the treatment of MLL-rearranged leukemia. J Hematol Oncol. 2016;9:24.

    PubMed  PubMed Central  Google Scholar 

  38. Verhaak RG, Goudswaard CS, van Putten W, Bijl MA, Sanders MA, Hugens W, et al. Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance. Blood. 2005;106:3747–54.

    PubMed  CAS  Google Scholar 

  39. Heuser M, Beutel G, Krauter J, Dohner K, von Neuhoff N, Schlegelberger B, et al. High meningioma 1 (MN1) expression as a predictor for poor outcome in acute myeloid leukemia with normal cytogenetics. Blood. 2006;108:3898–905.

    PubMed  CAS  Google Scholar 

  40. Sugino N, Kawahara M, Tatsumi G, Kanai A, Matsui H, Yamamoto R, et al. A novel LSD1 inhibitor NCD38 ameliorates MDS-related leukemia with complex karyotype by attenuating leukemia programs via activating super-enhancers. Leukemia. 2017;31:2303–14.

    PubMed  CAS  Google Scholar 

  41. Dahl R, Iyer SR, Owens KS, Cuylear DD, Simon MC. The transcriptional repressor GFI-1 antagonizes PU.1 activity through protein-protein interaction. J Biol Chem. 2007;282:6473–83.

    PubMed  CAS  Google Scholar 

  42. Spooner CJ, Cheng JX, Pujadas E, Laslo P, Singh H. A recurrent network involving the transcription factors PU.1 and Gfi1 orchestrates innate and adaptive immune cell fates. Immunity. 2009;31:576–86.

    PubMed  PubMed Central  CAS  Google Scholar 

  43. Maes T, Mascaro C, Tirapu I, Estiarte A, Ciceri F, Lunardi S, et al. ORY-1001, a potent and selective covalent KDM1A Inhibitor, for the treatment of acute leukemia. Cancer Cell. 2018;33:495–511. e12

    PubMed  CAS  Google Scholar 

  44. Maiques-Diaz A, Spencer GJ, Lynch JT, Ciceri F, Williams EL, Amaral FMR, et al. Enhancer activation by pharmacologic displacement of LSD1 from GFI1 induces differentiation in acute myeloid leukemia. Cell Rep. 2018;22:3641–59.

    PubMed  PubMed Central  CAS  Google Scholar 

  45. Cusan M, Cai SF, Mohammad HP, Krivtsov A, Chramiec A, Loizou E, et al. LSD1 inhibition exerts its antileukemic effect by recommissioning PU.1- and C/EBPalpha-dependent enhancers in AML. Blood. 2018;131:1730–42.

    PubMed  PubMed Central  CAS  Google Scholar 

  46. Zhou J, Wu J, Li B, Liu D, Yu J, Yan X, et al. PU.1 is essential for MLL leukemia partially via crosstalk with the MEIS/HOX pathway. Leukemia. 2014;28:1436–48.

    PubMed  CAS  Google Scholar 

  47. Rosenbauer F, Wagner K, Kutok JL, Iwasaki H, Le Beau MM, Okuno Y, et al. Acute myeloid leukemia induced by graded reduction of a lineage-specific transcription factor, PU.1. Nat Genet. 2004;36:624–30.

    PubMed  CAS  Google Scholar 

  48. Mohr S, Doebele C, Comoglio F, Berg T, Beck J, Bohnenberger H, et al. Hoxa9 and Meis1 cooperatively induce addiction to Syk signaling by suppressing miR-146a in acute myeloid leukemia. Cancer Cell. 2017;31:549–62. e11

    PubMed  PubMed Central  CAS  Google Scholar 

  49. Olsson A, Venkatasubramanian M, Chaudhri VK, Aronow BJ, Salomonis N, Singh H, et al. Single-cell analysis of mixed-lineage states leading to a binary cell fate choice. Nature. 2016;537:698–702.

    PubMed  PubMed Central  CAS  Google Scholar 

  50. Sharma A, Yun H, Jyotsana N, Chaturvedi A, Schwarzer A, Yung E, et al. Constitutive IRF8 expression inhibits AML by activation of repressed immune response signaling. Leukemia. 2015;29:157–68.

    PubMed  CAS  Google Scholar 

  51. Thivakaran A, Botezatu L, Hoenes JM, Al-Matary YS, Olberding N, Schütte J, et al. GFI1b as a novel oncosuppressor in AML. Blood. 2016;128:2717-.

    Google Scholar 

  52. Ishikawa Y, Gamo K, Yabuki M, Takagi S, Toyoshima K, Nakayama K, et al. A novel LSD1 inhibitor T-3775440 disrupts GFI1B-containing complex leading to transdifferentiation and impaired growth of AML cells. Mol Cancer Ther. 2017;16:273–84.

    PubMed  CAS  Google Scholar 

  53. Sanchez-Castillo M, Ruau D, Wilkinson AC, Ng FS, Hannah R, Diamanti E, et al. CODEX: a next-generation sequencing experiment database for the haematopoietic and embryonic stem cell communities. Nucleic Acids Res. 2015;43(Database issue):D1117–23.

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank the High Throughput Sequencing unit of the DKFZ Genomics and Proteomics Core Facility and the DKTK Cancer Genomics Core Unit Frankfurt for providing excellent library preparation and RNA sequencing services. We thank Alexandra Walter from the Institute of Pharmaceutical Sciences in Freiburg for technical assistance with the synthesis of used compounds. We also thank the team from the Cytology Lab of the Department of Medicine II, Hematology/Oncology for excellent staining of cytospins and blood smears. This work was performed as part of the LSD1 as Anticancer target in the Clinic and In drug Discovery (LACID) Joint Initiative of the German Cancer Consortium (DKTK). TB was supported by the Deutsche Forschungsgemeinschaft (DFG) (Grants BE 4198/1-1 and BE 4198/2-1) and by the LOEWE programs “Onkogene Signaltransduktion Frankfurt” (OSF) and “Zentrum für Zell- und Gentherapie Frankfurt (CGT)”. CK, JS and LV were supported by a Max-Eder-Fellowship of the Deutsche Krebshilfe and the Fritz-Thyssen-Foundation. ML, RS and MJ were supported by the DFG CRC 992 (MEDEP).

Author contributions

TB conceived the study together with RS, HS, ML, MJ and CK. JB designed and performed the majority of the experiments and subsequent data analysis. NK designed and performed the immunoprecipitation experiment. A-MM assisted in cell culture and mouse transplantation experiments. SM contributed to the mouse transplantation experiments. JS-F and MS synthesized and provided LSD1 inhibitors and advised on their use. DD, KA-E-A and SAW helped with the generation and analyzed the RNA sequencing data. GG performed combination treatment experiments with HL60. MT, EM and RS generated and provided the Lsd1cKO and Lsd1cKI mouse models. GB established and provided primary human AML cultures. JS, LV and CK contributed the Gfi1 KD data and the GficKO mouse model.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tobias Berg.

Ethics declarations

Conflict of interest

TB served as a consultant to Riemser Pharma GmbH and received travel funding from Incyte, Abbvie, Astellas, Alexion and Celgene. GB received research funding and honoraria from Novartis, honoraria from Amgen and Celgene and travel funding from Astellas, Janssen, Jazz Pharmaceuticals and Celgene. ML received research funding from Janssen Cilag, study drug support from Ratiopharm and travel funding from Janssen Cilag and Celgene.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Barth, J., Abou-El-Ardat, K., Dalic, D. et al. LSD1 inhibition by tranylcypromine derivatives interferes with GFI1-mediated repression of PU.1 target genes and induces differentiation in AML. Leukemia 33, 1411–1426 (2019). https://doi.org/10.1038/s41375-018-0375-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-018-0375-7

This article is cited by

Search

Quick links