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:

HBO1 is required for the maintenance of leukaemia stem cells

Abstract

Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by transcriptional dysregulation that results in a block in differentiation and increased malignant self-renewal. Various epigenetic therapies aimed at reversing these hallmarks of AML have progressed into clinical trials, but most show only modest efficacy owing to an inability to effectively eradicate leukaemia stem cells (LSCs)1. Here, to specifically identify novel dependencies in LSCs, we screened a bespoke library of small hairpin RNAs that target chromatin regulators in a unique ex vivo mouse model of LSCs. We identify the MYST acetyltransferase HBO1 (also known as KAT7 or MYST2) and several known members of the HBO1 protein complex as critical regulators of LSC maintenance. Using CRISPR domain screening and quantitative mass spectrometry, we identified the histone acetyltransferase domain of HBO1 as being essential in the acetylation of histone H3 at K14. H3 acetylated at K14 (H3K14ac) facilitates the processivity of RNA polymerase II to maintain the high expression of key genes (including Hoxa9 and Hoxa10) that help to sustain the functional properties of LSCs. To leverage this dependency therapeutically, we developed a highly potent small-molecule inhibitor of HBO1 and demonstrate its mode of activity as a competitive analogue of acetyl-CoA. Inhibition of HBO1 phenocopied our genetic data and showed efficacy in a broad range of human cell lines and primary AML cells from patients. These biological, structural and chemical insights into a therapeutic target in AML will enable the clinical translation of these findings.

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: HBO1 is an essential dependency in LSCs.
Fig. 2: Loss of HBO1 impairs LSC maintenance in vivo.
Fig. 3: HBO1 regulates expression of the 5′ end of the HOXA cluster in AML.
Fig. 4: Treatment with WM-3835 reduces AML growth.

Similar content being viewed by others

Data availability

The shRNA screen sequencing data have been deposited to the NCBI Sequence Archieve under the accession number GSE120813. Crystal structure data for HBO1–BPRF2 in complex with WM-3835 and acetyl-CoA have been submitted to the PDB under accession numbers 6MAJ (WM-3835) and 6MAK (acetyl-CoA). Source Data are provided for Figs. 14. Any other relevant data are available from the corresponding author upon reasonable request.

Code availability

All code used in this study is publically available.

References

  1. Dawson, M. A. The cancer epigenome: concepts, challenges, and therapeutic opportunities. Science 355, 1147–1152 (2017).

    ADS  PubMed  CAS  Google Scholar 

  2. Thomas, D. & Majeti, R. Biology and relevance of human acute myeloid leukemia stem cells. Blood 129, 1577–1585 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  3. Wang, T. et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  4. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Reports 17, 1193–1205 (2016).

    PubMed  CAS  Google Scholar 

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

    ADS  PubMed  PubMed Central  CAS  Google Scholar 

  6. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    ADS  PubMed  PubMed Central  CAS  Google Scholar 

  7. Fong, C. Y. et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 (2015).

    ADS  PubMed  PubMed Central  CAS  Google Scholar 

  8. Grimwade, D., Ivey, A. & Huntly, B. J. Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance. Blood 127, 29–41 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  9. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Kueh, A. J., Dixon, M. P., Voss, A. K. & Thomas, T. HBO1 is required for H3K14 acetylation and normal transcriptional activity during embryonic development. Mol. Cell. Biol. 31, 845–860 (2011).

    PubMed  CAS  Google Scholar 

  11. Doyon, Y. et al. ING tumor suppressor proteins are critical regulators of chromatin acetylation required for genome expression and perpetuation. Mol. Cell 21, 51–64 (2006).

    PubMed  CAS  Google Scholar 

  12. Sauer, T. et al. MYST2 acetyltransferase expression and histone H4 Lysine acetylation are suppressed in AML. Exp. Hematol. 43, 794–802 (2015).

    PubMed  CAS  Google Scholar 

  13. Weinert, B.T. et al. Time-resolved analysis reveals rapid dynamics and broad scope of the CBP/p300 acetylome. Cell 174, 231–244 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  14. Saksouk, N. et al. HBO1 HAT complexes target chromatin throughout gene coding regions via multiple PHD finger interactions with histone H3 tail. Mol. Cell 33, 257–265 (2009).

    PubMed  PubMed Central  CAS  Google Scholar 

  15. Shi, J. et al. Discovery of cancer drug targets by CRISPR–Cas9 screening of protein domains. Nat. Biotechnol. 33, 661–667 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  16. Krivtsov, A. V. et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006).

    ADS  PubMed  CAS  Google Scholar 

  17. Mupo, A. et al. A powerful molecular synergy between mutant nucleophosmin and Flt3-ITD drives acute myeloid leukemia in mice. Leukemia 27, 1917–1920 (2013).

    PubMed  PubMed Central  CAS  Google Scholar 

  18. Alharbi, R. A., Pettengell, R., Pandha, H. S. & Morgan, R. The role of HOX genes in normal hematopoiesis and acute leukemia. Leukemia 27, 1000–1008 (2013).

    PubMed  CAS  Google Scholar 

  19. Ballabio, E. & Milne, T. A. Epigenetic control of gene expression in leukemogenesis: cooperation between wild type MLL and MLL fusion proteins. Mol. Cell. Oncol. 1, e955330 (2014).

    PubMed  PubMed Central  Google Scholar 

  20. Skucha, A. et al. MLL-fusion-driven leukemia requires SETD2 to safeguard genomic integrity. Nat. Commun. 9, 1983 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  21. Church, M., Smith, K. C., Alhussain, M. M., Pennings, S. & Fleming, A. B. Sas3 and Ada2(Gcn5)-dependent histone H3 acetylation is required for transcription elongation at the de-repressed FLO1 gene. Nucleic Acids Res. 45, 4413–4430 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  22. Shen, W. et al. Solution structure of human Brg1 bromodomain and its specific binding to acetylated histone tails. Biochemistry 46, 2100–2110 (2007).

    PubMed  CAS  Google Scholar 

  23. Huber, F. M. et al. Histone-binding of DPF2 mediates its repressive role in myeloid differentiation. Proc. Natl Acad. Sci. USA 114, 6016–6021 (2017).

    PubMed  CAS  PubMed Central  Google Scholar 

  24. Dann, G. P. et al. ISWI chromatin remodellers sense nucleosome modifications to determine substrate preference. Nature 548, 607–611 (2017).

    ADS  PubMed  PubMed Central  CAS  Google Scholar 

  25. Baell, J. B. et al. Inhibitors of histone acetyltransferases KAT6A/B induce senescence and arrest tumour growth. Nature 560, 253–257 (2018).

    ADS  PubMed  CAS  Google Scholar 

  26. Majewski, I. J. et al. Polycomb repressive complex 2 (PRC2) restricts hematopoietic stem cell activity. PLoS Biol. 6, e93 (2008).

    PubMed  PubMed Central  Google Scholar 

  27. Voss, A. K., Collin, C., Dixon, M. P. & Thomas, T. Moz and retinoic acid coordinately regulate H3K9 acetylation, Hox gene expression, and segment identity. Dev. Cell 17, 674–686 (2009).

    PubMed  CAS  Google Scholar 

  28. Kinkel, S. A. et al. Jarid2 regulates hematopoietic stem cell function by acting with polycomb repressive complex 2. Blood 125, 1890–1900 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  29. Keniry, A. et al. Setdb1-mediated H3K9 methylation is enriched on the inactive X and plays a role in its epigenetic silencing. Epigenetics Chromatin 9, 16 (2016).

    PubMed  PubMed Central  Google Scholar 

  30. Dai, Z. et al. edgeR: a versatile tool for the analysis of shRNA-seq and CRISPR-Cas9 genetic screens. F1000Res. 3, 95 (2014).

    PubMed  PubMed Central  Google Scholar 

  31. Shechter, D., Dormann, H. L., Allis, C. D. & Hake, S. B. Extraction, purification and analysis of histones. Nat. Protocols 2, 1445–1457 (2007).

    PubMed  CAS  Google Scholar 

  32. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    PubMed  CAS  Google Scholar 

  33. Brumatti, G. et al. HoxA9 regulated Bcl-2 expression mediates survival of myeloid progenitors and the severity of HoxA9-dependent leukemia. Oncotarget 4, 1933–1947 (2013).

    PubMed  PubMed Central  Google Scholar 

  34. Tchasovnikarova, I. A. et al. Epigenetic silencing by the HUSH complex mediates position-effect variegation in human cells. Science 348, 1481–1485 (2015).

    ADS  PubMed  PubMed Central  CAS  Google Scholar 

  35. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 3 (2011).

    Google Scholar 

  36. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  37. Lavallée, V. P. et al. The transcriptomic landscape and directed chemical interrogation of MLL-rearranged acute myeloid leukemias. Nat. Genet. 47, 1030–1037 (2015).

    PubMed  Google Scholar 

  38. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  39. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

    MathSciNet  MATH  Google Scholar 

  40. Wu, D. et al. ROAST: rotation gene set tests for complex microarray experiments. Bioinformatics 26, 2176–2182 (2010).

    PubMed  PubMed Central  CAS  Google Scholar 

  41. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    PubMed  CAS  Google Scholar 

  42. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  43. Orlando, D. A. et al. Quantitative ChIP-seq normalization reveals global modulation of the epigenome. Cell Reports 9, 1163–1170 (2014).

    PubMed  CAS  Google Scholar 

  44. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    PubMed  PubMed Central  Google Scholar 

  45. Robinson, J. T. et al. Integrative Genomics Viewer. Nat. Biotechnol. 29, 24–26 (2011).

    PubMed  PubMed Central  CAS  Google Scholar 

  46. Tao, Y., Zhong, C., Zhu, J., Xu, S. & Ding, J. Structural and mechanistic insights into regulation of HBO1 histone acetyltransferase activity by BRPF2. Nucleic Acids Res. 45, 5707–5719 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  47. Waterman, D. G. et al. Diffraction-geometry refinement in the DIALS framework. Acta Crystallogr. D 72, 558–575 (2016).

    CAS  Google Scholar 

  48. Kabsch, W. Xds. Acta Crystallogr. D 66, 125–132 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Evans, P. R. & Murshudov, G. N. How good are my data and what is the resolution? Acta Crystallogr. D 69, 1204–1214 (2013).

    PubMed  CAS  PubMed Central  Google Scholar 

  50. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    PubMed  PubMed Central  CAS  Google Scholar 

  51. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    PubMed  CAS  PubMed Central  Google Scholar 

  52. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D 68, 352–367 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Murshudov, G. N., Vagin, A. A. & Dodson, E. J. Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr. D 53, 240–255 (1997).

    PubMed  CAS  Google Scholar 

  54. Gilan, O. et al. Functional interdependence of BRD4 and DOT1L in MLL leukemia. Nat. Struct. Mol. Biol. 23, 673–681 (2016).

    PubMed  CAS  Google Scholar 

  55. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

    PubMed Central  Google Scholar 

  56. Bagger, F. O. et al. BloodSpot: a database of gene expression profiles and transcriptional programs for healthy and malignant haematopoiesis. Nucleic Acids Res. 44, D917–D924 (2016).

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. Lovitt, J. Wingerd, S. Jackson and E. Allan for their technical contributions to this project. The work in the Dawson, Blewitt and Burns laboratories was supported by the Cancer Council Victoria Venture Grant Scheme, and Dawson laboratory work was supported by project grant funding from the National Health and Medical Research Council of Australia (1085015). We thank the following funders for fellowship and grant support: Leukaemia Foundation Australia senior fellowship, Cancer Council Victoria Dunlop Fellopship and Howard Hughes Medical Institute international research scholarship (M.A.D.); Victoria Cancer Agency early-career (L.M.) and mid-career (E.Y.N.L.) fellowships; CSL Centenary fellowship (S.-J.D.), Snowdome Foundation (P.Y.), Maddie Riewoldt’s Vision Foundation (Y.-C.C.), Bellberry-Viertel Senior Medical Research Fellowship (M.E.B), Novo Nordisk Foundation Hallas Møller Fellowship NNF14OC0008541 (C.C.), National Health and Medical Research Council of Australia through project grants 1081421 (J.B.B. and T.T.), 575558, 1084248 (A.K.V. and T.T.), research fellowship 1081421 (A.K.V.) and postgraduate scholarship (K.-L.C.). Salary support for M.Z., M.d.S., H.F., C.C., P.S.K., P.A.S., I.P.S. and B.J.M. was provided by the Cancer Therapeutics CRC, funded through the Australian Government’s Cooperative Research Centre programme. The Novo Nordisk Foundation Center for Protein Research is supported financially by the Novo Nordisk Foundation (grant agreement NNF14CC0001). This work was made possible through the Victorian State Government Operation Infrastructure Support and Australian National Health and the Medical Research Council Research Institute Infrastructure Support Scheme. We thank the Australian Synchrotron and beamline scientists for help with data collection; this research was undertaken in part using the MX2 beamline at the Australian Synchrotron and made use of the ACRF detector.

Author information

Authors and Affiliations

Authors

Contributions

M.A.D. developed the overall concept behind the study and supervised the project with important advice from L.M., M.E.B., C.J.B., A.K.V., T.T., C.C., F.G. and B.J.M. The manuscript was written by L.M., B.J.M. and M.A.D., with input from all the authors. L.M. performed the majority of the experiments with help from J.A., E.Y.N.L., Y.-C.C., C.-F.W., M.M.Y., P.Y., K.-L.C., K.K., M.S.B., M.L.B., J.L., Y.Y., R.A.B., T.T., H.F., M.d.S., N.N., P.S.K., M.Z., V.M.A. and C.C.-P. Critical reagents and interpretation of the research findings were provided by A.K.V., F.G., S.-J.D., P.A.S., C.C.-P., T.S.P., J.B.B., T.T., O. Dovey, G.S.V., C.C. and I.P.S. The shRNA library was designed and built by L.J.G., T.W., J.L., J.C. and M.E.B. A.H. and C.C. performed mass spectrometry analysis of histone acetylation sites. Protein production, SPR and structural studies were done by S.N., R.S., M.L.D., O. Dolezal, J.N., B.R. and T.S.P. WM-3835 was developed by J.B.B., D.J.L. and N.N. with analytical support by Y.S.

Corresponding author

Correspondence to Mark A. Dawson.

Ethics declarations

Competing interests

M.A.D. has been a member of advisory boards for CTX CRC, Storm Therapeutics, Celgene and Cambridge Epigenetix. The Dawson, Thomas and Voss laboratories receive research funding from CTx CRC. The remaining authors declare no competing financial interests.

Additional information

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

Peer review information Nature thanks Stefan Knapp and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Fig. 1 HBO1 is an AML-specific dependency.

a, RNA interference screen multidimensional scaling plot of shRNA sequences from bulk leukaemic blasts (bulk) and LSCs over 14 days of screening. Screens were performed in duplicate (R1 and R2). b, Expression of Ezh2, Lsd1 or Prmt5 mRNA, relative to non-targeting shRNA in LSCs that express shRNAs. n = 3.Mean + s.e.m. c, Negative-selection competition assays in bulk leukaemic blasts and LSCs that express shRNAs against Ezh2, Lsd1, Prmt5 or Dot1l. n = 3. Mean + s.e.m. Validation of the shRNAs against Dot1l have previously been published54. d. Expression of Hbo1 mRNA, relative to non-targeting shRNA, in LSCs that express shRNAs. n = 3. Mean + s.e.m.

Extended Data Fig. 2 Expression levels of HBO1.

a. Box plot of HBO1 expression levels in cancer types from The Cancer Genome Atlas (TCGA)55. The upper limit, centre and lower limit of each box denotes the upper quartile, median and lower quartile of the data, respectively. Cohort abbreviations refer to TCGA study abbreviations (https://gdc.cancer.gov/resources-tcga-users/tcga-code-tables/tcga-study-abbreviations). b, Dot plot of levels of HBO1 expression in normal haematopoietic and AML cells from BloodSpot56; horizontal line indicates mean expression.

Extended Data Fig. 3 HBO1 depletion increases apoptosis, cell-cycle arrest and myeloid differentiation in mouse LSCs.

a, Negative-selection competition assays in LSCs that express shRNAs against Moz, Qkf or Hbo1. n = 3. Mean + s.e.m. b, Negative-selection competition assays in LSCs that express Cas9, transduced with sgRNAs targeting Moz or Qkf. n = 3. Mean + s.e.m. c, Expression of members of the HBO1 complex, relative to non-targeting shRNA in LSCs that express shRNAs targeting individual members of the complex. n = 3. Mean + s.e.m. d, Apoptosis of LSCs that express shRNAs targeting Hbo1. n = 3. Mean + s.e.m. e, Cell-cycle profile of LSCs that express shRNAs targeting Hbo1. n = 3. Mean + s.e.m. f, Surface expression of GR1 and CD11b in LSCs that express shRNAs against Hbo1. n = 3. g. Apoptosis of LSCs that express Cas9 and sgRNAs targeting Hbo1. n = 3. Mean + s.e.m. h, Surface expression of KIT (also known as CD117) in LSCs that express sgRNAs targeting Hbo1. Representative of n = 2 biological replicates.

Extended Data Fig. 4 In vivo depletion of HBO1 increases disease latency.

a, Kaplan–Meier curves of NSG mice transplanted with quinary MLL–AF9 leukaemic cells that express shRNAs targeting Hbo1 or Moz. n = 6 mice per group. b, Schematic of wild-type and Hbo1-mutant alleles with numbered black boxes representing exons. Genotyping primers are indicated (adapted from ref. 10). c, Kaplan–Meier curves of C57BL/6 mice injected with 106 Hbo1flox/+ Mx1-cre primary MLL–AF9 leukaemic cells. Mice were treated with saline or pIpC. n = 12 mice per group. Right, representative genotyping of recipient bone marrow at end point. d, Kaplan–Meier curves of C57BL/6 mice injected with Moz+/+, Moz+/ or Moz−/− MLL–AF9 leukaemic cells. n = 5 mice per group. e, Kaplan–Meier curves of BALB/c mice injected with Moz+/+ or Moz−/− HOXA9 and MEIS1 leukaemic cells. n = 5 mice per group.

Extended Data Fig. 5 HBO1 is a dependency in various subtypes of AML.

a, Negative-selection competition assays in human AML cell lines that express Cas9 and sgRNAs targeting HBO1. n = 3. Mean + s.e.m. Driver mutations are in parentheses. b, Immunoblot of HBO1 and H3K14ac in OCI-AML3 cells that express Cas9 and sgRNAs targeting HBO1. n = 3. Mean + s.e.m. c, Cell-cycle profile of OCI-AML3 cells that express Cas9 and sgRNAs targeting HBO1. n = 3. Mean + s.e.m. d, Apoptosis of OCI-AML3 cells that express Cas9 and sgRNAs targeting HBO1. n = 3. Mean + s.e.m. e, Surface expression of CD11b in OCI-AML3 cells that express Cas9 and sgRNAs targeting HBO1. n = 3.

Extended Data Fig. 6 HBO1 dependency in other cancers.

Negative-selection competition assays in human cancer cell lines that express Cas9 and sgRNAs targeting HBO1. n = 3. Mean + s.e.m. Cancer type is in parentheses.

Extended Data Fig. 7 HBO1 depletion increases myeloid signature and decreases global H3K14ac.

a, Barcode plot evaluating changes in myeloid development signature after HBO1 depletion with Hbo1 e11.1 and Hbo1 e12.2 sgRNAs in LSCs. n = 3. b, Bar plot of changes in gene expression after Hbo1 deletion in LSCs, ranked by expression levels. Green bars show the top 25 most-downregulated genes after Hbo1 deletion. c, HOXA9 and HOXA10 mRNA expression in Molm13 and OCI-AML3 cells that express Cas9 and sgRNA targeting HBO1. n = 3. Mean ± s.e.m. d, Surface expression of CD11b in LSCs that overexpress Hoxa9 or Hoxa10, and sgRNAs targeting Hbo1. e, Immunoblot of overexpressed HOXA9 or HOXA10 in LSCs that express Cas9. Representative of n = 3 biological replicates. f, Hoxa9- and Hoxa10-rescue negative-selection competition assays. Representative of n = 3 biological replicates. g, ChIP–seq profiles of H3K14ac and RNA Pol-II at the Pbx3 locus in LSCs that express Cas9 and an sgRNA targeting Hbo1. Representative of n = 3 biological replicates. h, RNA Pol-II travelling ratio distribution for highly expressed genes, divided according to H3K14ac levels from ChIP–seq. i, Surface expression of GR1 in LSCs that overexpress sgRNAs targeting Smarca5. Representative of n = 3 biological replicates.

Extended Data Fig. 8 WM-3835 inhibits cell growth and HOXA expression in AML.

a, KAT biochemical and SPR values for WM-3835 compared to WM-1119. Biochemical assay was done at 1 μM acetyl-CoA, the Km of HBO1. b, Proliferation assays of human AML cells treated with 1 μM WM-2474. n = 3. Mean + s.e.m. c, Cellular H3K14ac biomarker assay dose–response curves for WM-3835 (blue) and WM-1119 (red). n = 6. Mean ± s.e.m. d, Growth inhibition assays of the MLL–AF9 AML cell line Molm13 treated with WM-3835 at the doses indicated. Boxes represent minimum and maximum values. n = 11. Mean ± s.e.m. e, Cell-cycle profile of OCI-AML3 cells treated with WM-3835 or vehicle. n = 3. Mean + s.e.m. f, Apoptosis of OCI-AML3 cells treated with WM-3835 or vehicle. n = 3. Mean + s.e.m. g, Surface expression of CD11b in OCI-AML3 cells treated with WM-3835 or vehicle. n = 3. h, HOXA10 mRNA expression in LSCs and Molm13 cells treated with WM-3835 or vehicle. n = 3. Mean + s.e.m. i, HOXA9 and HOXA10 mRNA expression in OCI-AML3 cells treated with WM-3835. n = 3. Mean + s.e.m. j, Hoxa9 and Hoxa10 rescue proliferation assays with 1 μM WM-3835 in LSCs. n = 3. Mean ± s.e.m.

Extended Data Fig. 9 HBO1 crystal structure.

a, Data collection and refinement statistics of WM-3835 HBO1–BRPF2 co-crystal structure. b, WM-3835 binding site in HBO1–BRPF2. WM-3835 is shown in silver with element colouring and the omit electron density map, contoured to 3σ, is shown in green. c, Overlay of WM-3835 and acetyl-CoA (purple with element colouring), showing that WM-3835 binds in the acetyl-CoA binding site of HBO1. d, Ribbon diagram of HBO1–BRPF2 showing WM-3835 bound to the acetyl-CoA binding site. e, Space-filling model showing WM-3835 (yellow with element colouring) in the acetyl-CoA binding pocket of HBO1–BRPF2.

Extended Data Fig. 10 High in vitro metabolism and poor in vivo oral exposure of WM-3835.

a, Proliferation assays of human AML cell lines treated with 1 μM WM-3835. b, WM-3835 demonstrates high clearance in both human and mouse liver microsome assays. The use of dual cofactors (UDPGA and NADPH) results in an increased rate of clearance in human liver microsomes, which is consistent with glucuronidation having a role in the clearance of this compound. c, BALB/c female mice were dosed with WM-3835 at 100 mg kg−1 taken by mouth twice a day, formulated in 20% PEG400/10% solutol or vehicle. Four hours after the third dose, blood samples were collected. An average total drug concentration of 1,860 nM was observed. The free drug level was determined to be 2.6 nM after accounting for mouse plasma protein binding (fraction unbound 0.0014). This free drug level was considered too low to affect the acetylation of H3 at K14, on the basis of the in vitro H3K14ac cellular biomarker data (Fig. 4). An additional chromatographic peak eluting earlier than WM-3835 was detected in the plasma samples from the treatment group. Subsequent analyses using predicted multiple-reaction monitoring and accurate mass measurement indicated that this peak is likely to be a glucuronide conjugate of WM-3835, consistent with the in vitro metabolism data.

Supplementary information

Supplementary Information

This file contains supplementary methods detailing the chemistry for WM-3835.

Reporting Summary

Supplementary Figure

Contains scans of uncropped images of western blot gels.

Supplementary Table 1

Number of shRNAs depleted by more than 5-fold in leukaemic stem cell and blast cell populations from screen.

Supplementary Table 2

A list of acetylation sites quantified in core histones. Site-specific acetylation of core histones was quantified in HBO1 knockout and control cells using SILAC-based mass spectrometry. The data shows median SILAC ratio (HBO1 knockout/Control) from three independent biological replicates.

Supplementary Table 3

shRNA, sgRNA and mRNA primer sequences.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

MacPherson, L., Anokye, J., Yeung, M.M. et al. HBO1 is required for the maintenance of leukaemia stem cells. Nature 577, 266–270 (2020). https://doi.org/10.1038/s41586-019-1835-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1835-6

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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