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

Drug targeting of NR4A nuclear receptors for treatment of acute myeloid leukemia

Abstract

NR4As are AML tumor suppressors that are frequently silenced in human acute myeloid leukemia (AML). Despite their potential as novel targets for therapeutic intervention, mechanisms of NR4A silencing and strategies for their reactivation remain poorly defined. Here we show that NR4A silencing in AML occurs through blockade of transcriptional elongation rather than epigenetic promoter silencing. By intersection of NR4A-regulated gene signatures captured upon acute, exogenous expression of NR4As in human AML cells with in silico chemical genomics screening, we identify several FDA-approved drugs including dihydroergotamine (DHE) that reactivate NR4A expression and regulate NR4A-dependent gene signatures. We show that DHE induces NR4A expression via recruitment of the super elongation complex to enable elongation of NR4A promoter paused RNA polymerase II. Finally, DHE exhibits AML selective NR4A-dependent anti-leukemic activity in cytogenetically distinct human AML cells in vitro and delays AML progression in mice revealing its potential as a novel therapeutic agent in AML.

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

Similar content being viewed by others

References

  1. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645–8.

    Article  CAS  Google Scholar 

  2. Jordan CT. The leukemic stem cell. Best Pract Res Clin Haematol. 2007;20:13–8.

    Article  CAS  Google Scholar 

  3. Kadia TM, Ravandi F, Cortes J, Kantarjian H. Toward individualized therapy in acute myeloid leukemia: a contemporary review. JAMA Oncol. 2015;1:820–8.

    Article  Google Scholar 

  4. Cancer Genome Atlas Research Network. et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368:2059–74.

    Article  Google Scholar 

  5. Goardon N, Marchi E, Atzberger A, Quek L, Schuh A, Soneji S, et al. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell. 2011;19:138–52.

    Article  CAS  Google Scholar 

  6. Wiseman DH, Greystoke BF, Somervaille TC. The variety of leukemic stem cells in myeloid malignancy. Oncogene. 2014;33:3091–8.

    Article  CAS  Google Scholar 

  7. Klco JM, Spencer DH, Miller CA, Griffith M, Lamprecht TL, O’Laughlin M, et al. Functional heterogeneity of genetically defined subclones in acute myeloid leukemia. Cancer Cell. 2014;25:379–92.

    Article  CAS  Google Scholar 

  8. Maxwell MA, Muscat GE. The NR4A subgroup: immediate early response genes with pleiotropic physiological roles. Nucl Recept Signal. 2006;4:e002.

    Article  Google Scholar 

  9. Safe S, Jin UH, Morpurgo B, Abudayyeh A, Singh M, Tjalkens RB. Nuclear receptor 4A (NR4A) family - orphans no more. J Steroid Biochem Mol Biol. 2016;157:48–60.

    Article  CAS  Google Scholar 

  10. Hamers AA, Hanna RN, Nowyhed H, Hedrick CC, de Vries CJ. NR4A nuclear receptors in immunity and atherosclerosis. Curr Opin Lipidol. 2013;24:381–5.

    Article  CAS  Google Scholar 

  11. Mullican SE, Zhang S, Konopleva M, Ruvolo V, Andreeff M, Milbrandt J, et al. Abrogation of nuclear receptors Nr4a3 and Nr4a1 leads to development of acute myeloid leukemia. Nat Med. 2007;13:730–5.

    Article  CAS  Google Scholar 

  12. Sirin O, Lukov GL, Mao R, Conneely OM, Goodell MA. The orphan nuclear receptor Nurr1 restricts the proliferation of haematopoietic stem cells. Nat Cell Biol. 2010;12:1213–9.

    Article  CAS  Google Scholar 

  13. Cheng LE, Chan FK, Cado D, Winoto A. Functional redundancy of the Nur77 and Nor-1 orphan steroid receptors in T-cell apoptosis. EMBO J. 1997;16:1865–75.

    Article  CAS  Google Scholar 

  14. Woronicz JD, Calnan B, Ngo V, Winoto A. Requirement for the orphan steroid receptor Nur77 in apoptosis of T-cell hybridomas. Nature. 1994;367:277–81.

    Article  CAS  Google Scholar 

  15. Lee SL, Wesselschmidt RL, Linette GP, Kanagawa O, Russell JH, Milbrandt J. Unimpaired thymic and peripheral T cell death in mice lacking the nuclear receptor NGFI-B (Nur77). Science. 1995;269:532–5.

    Article  CAS  Google Scholar 

  16. Sekiya T, Kashiwagi I, Yoshida R, Fukaya T, Morita R, Kimura A, et al. Nr4a receptors are essential for thymic regulatory T cell development and immune homeostasis. Nat Immunol. 2013;14:230–7.

    Article  CAS  Google Scholar 

  17. Sekiya T, Kondo T, Shichita T, Morita R, Ichinose H, Yoshimura A. Suppression of Th2 and Tfh immune reactions by Nr4a receptors in mature T reg cells. J Exp Med. 2015;212:1623–40.

    Article  CAS  Google Scholar 

  18. Nowyhed HN, Huynh TR, Blatchley A, Wu R, Thomas GD, Hedrick CC. The nuclear receptor nr4a1 controls CD8 T cell development through transcriptional suppression of runx3. Sci Rep. 2015;5:9059.

    Article  CAS  Google Scholar 

  19. Hanna RN, Carlin LM, Hubbeling HG, Nackiewicz D, Green AM, Punt JA, et al. The transcription factor NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C- monocytes. Nat Immunol. 2011;12:778–85.

    Article  CAS  Google Scholar 

  20. Tacke R, Hilgendorf I, Garner H, Waterborg C, Park K, Nowyhed H, et al. The transcription factor NR4A1 is essential for the development of a novel macrophage subset in the thymus. Sci Rep. 2015;5:10055.

    Article  Google Scholar 

  21. Hanna RN, Shaked I, Hubbeling HG, Punt JA, Wu R, Herrley E, et al. NR4A1 (Nur77) deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis. Circ Res. 2012;110:416–27.

    Article  CAS  Google Scholar 

  22. Ramirez-Herrick AM, Mullican SE, Sheehan AM, Conneely OM. Reduced NR4A gene dosage leads to mixed myelodysplastic/myeloproliferative neoplasms in mice. Blood. 2011;117:2681–90.

    Article  CAS  Google Scholar 

  23. Deutsch AJ, Rinner B, Wenzl K, Pichler M, Troppan K, Steinbauer E, et al. NR4A1-mediated apoptosis suppresses lymphomagenesis and is associated with a favorable cancer-specific survival in patients with aggressive B-cell lymphomas. Blood. 2014;123:2367–77.

    Article  CAS  Google Scholar 

  24. Wenzl K, Troppan K, Neumeister P, Deutsch AJ. The nuclear orphan receptor NR4A1 and NR4A3 as tumor suppressors in hematologic neoplasms. Curr Drug Targets. 2015;16:38–46.

    Article  CAS  Google Scholar 

  25. Boudreaux SP, Ramirez-Herrick AM, Duren RP, Conneely OM. Genome-wide profiling reveals transcriptional repression of MYC as a core component of NR4A tumor suppression in acute myeloid leukemia. Oncogenesis. 2012;1:e19.

    Article  CAS  Google Scholar 

  26. Pellagatti A, Cazzola M, Giagounidis AA, Malcovati L, Porta MG, Killick S, et al. Gene expression profiles of CD34+cells in myelodysplastic syndromes: involvement of interferon-stimulated genes and correlation to FAB subtype and karyotype. Blood. 2006;108:337–45.

    Article  CAS  Google Scholar 

  27. Majeti R, Becker MW, Tian Q, Lee TL, Yan X, Liu R, et al. Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc Natl Acad Sci USA. 2009;106:3396–401.

    Article  CAS  Google Scholar 

  28. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, et al. The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006;313:1929–35.

    Article  CAS  Google Scholar 

  29. Duren RP, Boudreaux SP, Conneely OM. Genome wide mapping of NR4A binding reveals cooperativity with ETS factors to promote epigenetic activation of distal enhancers in acute myeloid leukemia cells. PLoS One. 2016;11:e0150450.

    Article  Google Scholar 

  30. Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60.

    Article  CAS  Google Scholar 

  31. Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA. A chromatin landmark and transcription initiation at most promoters in human cells. Cell. 2007;130:77–88.

    Article  CAS  Google Scholar 

  32. Jonkers I, Lis JT. Getting up to speed with transcription elongation by RNA polymerase II. Nat Rev Mol Cell Biol. 2015;16:167–77.

    Article  CAS  Google Scholar 

  33. Rickert P, Corden JL, Lees E. Cyclin C/CDK8 and cyclin H/CDK7/p36 are biochemically distinct CTD kinases. Oncogene. 1999;18:1093–102.

    Article  CAS  Google Scholar 

  34. Akhtar MS, Heidemann M, Tietjen JR, Zhang DW, Chapman RD, Eick D, et al. TFIIH kinase places bivalent marks on the carboxy-terminal domain of RNA polymerase II. Mol Cell. 2009;34:387–93.

    Article  CAS  Google Scholar 

  35. Donner AJ, Ebmeier CC, Taatjes DJ, Espinosa JM. CDK8 is a positive regulator of transcriptional elongation within the serum response network. Nat Struct Mol Biol. 2010;17:194–201.

    Article  CAS  Google Scholar 

  36. Galbraith MD, Allen MA, Bensard CL, Wang X, Schwinn MK, Qin B, et al. HIF1A employs CDK8-mediator to stimulate RNAPII elongation in response to hypoxia. Cell. 2013;153:1327–39.

    Article  CAS  Google Scholar 

  37. Baylin SB, Ohm JE. Epigenetic gene silencing in cancer—a mechanism for early oncogenic pathway addiction? Nat Rev Cancer. 2006;6:107–16.

    Article  CAS  Google Scholar 

  38. Schlesinger Y, Straussman R, Keshet I, Farkash S, Hecht M, Zimmerman J, et al. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat Genet. 2007;39:232–6.

    Article  CAS  Google Scholar 

  39. Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C, et al. Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet. 2008;40:741–50.

    Article  CAS  Google Scholar 

  40. Issa JP. DNA methylation as a therapeutic target in cancer. Clin Cancer Res. 2007;13:1634–7.

    Article  CAS  Google Scholar 

  41. Wagner JM, Hackanson B, Lubbert M, Jung M. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenetics. 2010;1:117–36.

    Article  CAS  Google Scholar 

  42. Brien GL, Valerio DG, Armstrong SA. Exploiting the epigenome to control cancer-promoting gene-expression programs. Cancer Cell. 2016;29:464–76.

    Article  CAS  Google Scholar 

  43. Gardini A, Baillat D, Cesaroni M, Hu D, Marinis JM, Wagner EJ, et al. Integrator regulates transcriptional initiation and pause release following activation. Mol Cell. 2014;56:128–39.

    Article  CAS  Google Scholar 

  44. Chen K, Chen Z, Wu D, Zhang L, Lin X, Su J, et al. Broad H3K4me3 is associated with increased transcription elongation and enhancer activity at tumor-suppressor genes. Nat Genet. 2015;47:1149–57.

    Article  CAS  Google Scholar 

  45. Hargreaves DC, Horng T, Medzhitov R. Control of inducible gene expression by signal-dependent transcriptional elongation. Cell. 2009;138:129–45.

    Article  CAS  Google Scholar 

  46. Adelman K, Kennedy MA, Nechaev S, Gilchrist DA, Muse GW, Chinenov Y, et al. Immediate mediators of the inflammatory response are poised for gene activation through RNA polymerase II stalling. Proc Natl Acad Sci USA. 2009;106:18207–12.

    Article  CAS  Google Scholar 

  47. Brondfield S, Umesh S, Corella A, Zuber J, Rappaport AR, Gaillard C, et al. Direct and indirect targeting of MYC to treat acute myeloid leukemia. Cancer Chemother Pharmacol. 2015;76:35–46.

    Article  CAS  Google Scholar 

  48. Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 2011;478:529–33.

    Article  CAS  Google Scholar 

  49. Zuber J, Radtke I, Pardee TS, Zhao Z, Rappaport AR, Luo W, et al. Mouse models of human AML accurately predict chemotherapy response. Genes Dev. 2009;23:877–89.

    Article  CAS  Google Scholar 

  50. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008;22:659–61.

    Article  CAS  Google Scholar 

  51. Silberstein SD, McCrory DC. Ergotamine and dihydroergotamine: history, pharmacology, and efficacy. Headache. 2003;43:144–66.

    Article  Google Scholar 

  52. Mendez-Ferrer S, Battista M, Frenette PS. Cooperation of beta(2)- and beta(3)-adrenergic receptors in hematopoietic progenitor cell mobilization. Ann N Y Acad Sci. 2010;1192:139–44.

    Article  CAS  Google Scholar 

  53. Lucas D, Bruns I, Battista M, Mendez-Ferrer S, Magnon C, Kunisaki Y, et al. Norepinephrine reuptake inhibition promotes mobilization in mice: potential impact to rescue low stem cell yields. Blood. 2012;119:3962–5.

    Article  CAS  Google Scholar 

  54. Spiegel A, Shivtiel S, Kalinkovich A, Ludin A, Netzer N, Goichberg P, et al. Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+cells through Wnt signaling. Nat Immunol. 2007;8:1123–31.

    Article  CAS  Google Scholar 

  55. Yang M, Li K, Ng PC, Chuen CK, Lau TK, Cheng YS, et al. Promoting effects of serotonin on hematopoiesis: ex vivo expansion of cord blood CD34+stem/progenitor cells, proliferation of bone marrow stromal cells, and antiapoptosis. Stem Cells. 2007;25:1800–6.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by RO1 CA160747 from National Institutes of Health to OMC and by the Cell Sorting and Flow Cytometry shared resource of the Dan L. Duncan Comprehensive Cancer Center with funding from National Cancer Institute grant (P30CA125123). The authors acknowledge the joint participation by Adrienne Helis Melvin Medical Research Foundation through its direct engagement in the continuous active conduct of medical research in conjunction with Baylor College of Medicine. The authors also thank Dr. Terzah Horton and the Leukemia Research Interest Group at Texas Children’s Hospital for providing primary patient AML samples.

Statement of significance

A chemical genomics strategy identifies DHE as a novel activator of silenced NR4A tumor suppressors with repositioning potential for treatment of AML.

Author contributions

S.P.B., R.P.D., and S.G.C. carried out experiments including CMap chemical genomics integration, ChIP-experiments, bisulfite sequencing, and cell growth assays. P.R.F. did flow cytometry analysis. L.N. did transplantation assays. P.N., M.S.R., and P.R.F. were responsible for colony forming assays on primary AML samples. S.P.B. and O.M.C. conceived the strategy, designed experiments and wrote the paper.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Orla M. Conneely.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boudreaux, S.P., Duren, R.P., Call, S.G. et al. Drug targeting of NR4A nuclear receptors for treatment of acute myeloid leukemia. Leukemia 33, 52–63 (2019). https://doi.org/10.1038/s41375-018-0174-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-018-0174-1

This article is cited by

Search

Quick links