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:

Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes

Abstract

The development of selective histone deacetylase (HDAC) inhibitors with anti-cancer and anti-inflammatory properties remains challenging in large part owing to the difficulty of probing the interaction of small molecules with megadalton protein complexes. A combination of affinity capture and quantitative mass spectrometry revealed the selectivity with which 16 HDAC inhibitors target multiple HDAC complexes scaffolded by ELM-SANT domain subunits, including a novel mitotic deacetylase complex (MiDAC). Inhibitors clustered according to their target profiles with stronger binding of aminobenzamides to the HDAC NCoR complex than to the HDAC Sin3 complex. We identified several non-HDAC targets for hydroxamate inhibitors. HDAC inhibitors with distinct profiles have correspondingly different effects on downstream targets. We also identified the anti-inflammatory drug bufexamac as a class IIb (HDAC6, HDAC10) HDAC inhibitor. Our approach enables the discovery of novel targets and inhibitors and suggests that the selectivity of HDAC inhibitors should be evaluated in the context of HDAC complexes and not purified catalytic subunits.

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: Mapping of HDAC drug target complexes in chemical space and in proteome space.
Figure 2: HDAC inhibitor drug targets and target complexes are defined by chemoproteomics profiling of drugs and compounds used as research tools.
Figure 3: Deconvolution of protein complexes by co-IP analysis confirms the identification of novel HDAC complexes.
Figure 4: Class I HDACs and DNTTIP1 form a mitotic deacetylase complex (MiDAC).
Figure 5: Differential effects of HDAC inhibitors on histone and tubulin acetylation.
Figure 6: The nonsteroidal anti-inflammatory drug bufexamac is a novel class IIb HDAC inhibitor.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  CAS  Google Scholar 

  2. Choudhary, C. et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 325, 834–840 (2009).

    Article  CAS  Google Scholar 

  3. Zhao, S. et al. Regulation of cellular metabolism by protein lysine acetylation. Science 327, 1000–1004 (2010).

    Article  CAS  Google Scholar 

  4. Karberg, S. Switching on epigenetic therapy. Cell 139, 1029–1031 (2009).

    Article  CAS  Google Scholar 

  5. Taunton, J., Hassig, C.A. & Schreiber, S.L. A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p. Science 272, 408–411 (1996).

    Article  CAS  Google Scholar 

  6. Gregoretti, I.V., Lee, Y.M. & Goodson, H.V. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J. Mol. Biol. 338, 17–31 (2004).

    Article  CAS  Google Scholar 

  7. Yang, X.J. & Seto, E. The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat. Rev. Mol. Cell Biol. 9, 206–218 (2008).

    Article  CAS  Google Scholar 

  8. Cunliffe, V.T. Eloquent silence: developmental functions of Class I histone deacetylases. Curr. Opin. Genet. Dev. 18, 404–410 (2008).

    Article  CAS  Google Scholar 

  9. You, A., Tong, J.K., Grozinger, C.M. & Schreiber, S.L. CoREST is an integral component of the CoREST-human histone deacetylase complex. Proc. Natl. Acad. Sci. USA 98, 1454–1458 (2001).

    Article  CAS  Google Scholar 

  10. Tong, J.K., Hassig, C.A., Schnitzler, G.R., Kingston, R.E. & Schreiber, S.L. Chromatin deacetylation by an ATP-dependent nucleosome remodelling complex. Nature 395, 917–921 (1998).

    Article  CAS  Google Scholar 

  11. Zhang, Y., Iratni, R., Erdjument-Bromage, H., Tempst, P. & Reinberg, D. Histone deacetylases and SAP18, a novel polypeptide, are components of a human Sin3 complex. Cell 89, 357–364 (1997).

    Article  CAS  Google Scholar 

  12. Karagianni, P. & Wong, J. HDAC3: taking the SMRT-N-CoRrect road to repression. Oncogene 26, 5439–5449 (2007).

    Article  CAS  Google Scholar 

  13. Guenther, M.G., Barak, O. & Lazar, M.A. The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Mol. Cell. Biol. 21, 6091–6101 (2001).

    Article  CAS  Google Scholar 

  14. Bradner, J.E. et al. Chemical phylogenetics of histone deacetylases. Nat. Chem. Biol. 6, 238–243 (2010).

    Article  CAS  Google Scholar 

  15. Lahm, A. et al. Unraveling the hidden catalytic activity of vertebrate class IIa histone deacetylases. Proc. Natl. Acad. Sci. USA 104, 17335–17340 (2007).

    Article  CAS  Google Scholar 

  16. Boyault, C., Sadoul, K., Pabion, M. & Khochbin, S. HDAC6, at the crossroads between cytoskeleton and cell signaling by acetylation and ubiquitination. Oncogene 26, 5468–5476 (2007).

    Article  CAS  Google Scholar 

  17. Marks, P.A. & Breslow, R. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat. Biotechnol. 25, 84–90 (2007).

    Article  CAS  Google Scholar 

  18. Bolden, J.E., Peart, M.J. & Johnstone, R.W. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 5, 769–784 (2006).

    Article  CAS  Google Scholar 

  19. Zhang, Y. et al. Analysis of the NuRD subunits reveals a histone deacetylase core complex and a connection with DNA methylation. Genes Dev. 13, 1924–1935 (1999).

    Article  CAS  Google Scholar 

  20. Salisbury, C.M. & Cravatt, B.F. Activity-based probes for proteomic profiling of histone deacetylase complexes. Proc. Natl. Acad. Sci. USA 104, 1171–1176 (2007).

    Article  CAS  Google Scholar 

  21. Bantscheff, M. et al. Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 25, 1035–1044 (2007).

    Article  CAS  Google Scholar 

  22. Ong, S.E. et al. Identifying the proteins to which small-molecule probes and drugs bind in cells. Proc. Natl. Acad. Sci. USA 106, 4617–4622 (2009).

    Article  CAS  Google Scholar 

  23. Sharma, K. et al. Proteomics strategy for quantitative protein interaction profiling in cell extracts. Nat. Methods 6, 741–744 (2009).

    Article  CAS  Google Scholar 

  24. Bantscheff, M., Scholten, A. & Heck, A.J. Revealing promiscuous drug-target interactions by chemical proteomics. Drug Discov. Today 14, 1021–1029 (2009).

    Article  CAS  Google Scholar 

  25. Khan, N. et al. Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. Biochem. J. 409, 581–589 (2008).

    Article  CAS  Google Scholar 

  26. Bantscheff, M. et al. Robust and sensitive iTRAQ quantification on an LTQ Orbitrap mass spectrometer. Mol. Cell. Proteomics 7, 1702–1713 (2008).

    Article  CAS  Google Scholar 

  27. Jones, P. et al. Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases. Bioorg. Med. Chem. Lett. 18, 1814–1819 (2008).

    Article  CAS  Google Scholar 

  28. Kruhlak, M.J. et al. Regulation of global acetylation in mitosis through loss of histone acetyltransferases and deacetylases from chromatin. J. Biol. Chem. 276, 38307–38319 (2001).

    CAS  PubMed  Google Scholar 

  29. Savitski, M.M. et al. Targeted data acquisition for improved reproducibility and robustness of proteomic mass spectrometry assays. J. Am. Soc. Mass Spectrom. 21, 1668–1679 (2010).

    Article  CAS  Google Scholar 

  30. Chou, C.J., Herman, D. & Gottesfeld, J.M. Pimelic diphenylamide 106 is a slow, tight-binding inhibitor of class I histone deacetylases. J. Biol. Chem. 283, 35402–35409 (2008).

    Article  CAS  Google Scholar 

  31. Wang, L., Charroux, B., Kerridge, S. & Tsai, C.C. Atrophin recruits HDAC1/2 and G9a to modify histone H3K9 and to determine cell fates. EMBO Rep. 9, 555–562 (2008).

    Article  CAS  Google Scholar 

  32. Mulligan, P. et al. CDYL bridges REST and histone methyltransferases for gene repression and suppression of cellular transformation. Mol. Cell 32, 718–726 (2008).

    Article  CAS  Google Scholar 

  33. Savitski, M.M., Mathieson, T., Becher, I. & Bantscheff, M. H-score, a mass accuracy driven rescoring approach for improved Peptide identification in modification rich samples. J. Proteome Res. 9, 5511–5516 (2010).

    Article  CAS  Google Scholar 

  34. Trommer, H. et al. Examinations of the antioxidative properties of the topically administered drug bufexamac reveal new insights into its mechanism of action. J. Pharm. Pharmacol. 55, 1379–1388 (2003).

    Article  CAS  Google Scholar 

  35. Alberts, B. The cell as a collection of protein machines: preparing the next generation of molecular biologists. Cell 92, 291–294 (1998).

    Article  CAS  Google Scholar 

  36. Gavin, A.C. et al. Proteome survey reveals modularity of the yeast cell machinery. Nature 440, 631–636 (2006).

    Article  CAS  Google Scholar 

  37. Malovannaya, A. et al. Streamlined analysis schema for high-throughput identification of endogenous protein complexes. Proc. Natl. Acad. Sci. USA 107, 2431–2436 (2010).

    Article  CAS  Google Scholar 

  38. Beckers, T. et al. Distinct pharmacological properties of second generation HDAC inhibitors with the benzamide or hydroxamate head group. Int. J. Cancer 121, 1138–1148 (2007).

    Article  CAS  Google Scholar 

  39. Blackwell, L., Norris, J., Suto, C.M. & Janzen, W.P. The use of diversity profiling to characterize chemical modulators of the histone deacetylases. Life Sci. 82, 1050–1058 (2008).

    Article  CAS  Google Scholar 

  40. Farias, E.F. et al. Interference with Sin3 function induces epigenetic reprogramming and differentiation in breast cancer cells. Proc. Natl. Acad. Sci. USA 107, 11811–11816 (2010).

    Article  CAS  Google Scholar 

  41. Smith, K.T., Martin-Brown, S.A., Florens, L., Washburn, M.P. & Workman, J.L. Deacetylase inhibitors dissociate the histone-targeting ING2 subunit from the Sin3 complex. Chem. Biol. 17, 65–74 (2010).

    Article  CAS  Google Scholar 

  42. Kubota, T., Maezawa, S., Koiwai, K., Hayano, T. & Koiwai, O. Identification of functional domains in TdIF1 and its inhibitory mechanism for TdT activity. Genes Cells 12, 941–959 (2007).

    Article  CAS  Google Scholar 

  43. Balasubramanian, S. et al. A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI-34051 induces apoptosis in T-cell lymphomas. Leukemia 22, 1026–1034 (2008).

    Article  CAS  Google Scholar 

  44. Wang, Z. et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138, 1019–1031 (2009).

    Article  CAS  Google Scholar 

  45. Thingholm, T.E., Palmisano, G., Kjeldsen, F. & Larsen, M.R. Undesirable charge-enhancement of isobaric tagged phosphopeptides leads to reduced identification efficiency. J. Proteome Res. 9, 4045–4052 (2010).

    Article  CAS  Google Scholar 

  46. Olsen, J.V. et al. Parts per million mass accuracy on an Orbitrap mass spectrometer via lock mass injection into a C-trap. Mol. Cell. Proteomics 4, 2010–2021 (2005).

    Article  CAS  Google Scholar 

  47. Kocher, T. et al. High precision quantitative proteomics using iTRAQ on an LTQ Orbitrap: a new mass spectrometric method combining the benefits of all. J. Proteome Res. 8, 4743–4752 (2009).

    Article  CAS  Google Scholar 

  48. Sanders, S.L., Jennings, J., Canutescu, A., Link, A.J. & Weil, P.A. Proteomics of the eukaryotic transcription machinery: identification of proteins associated with components of yeast TFIID by multidimensional mass spectrometry. Mol. Cell. Biol. 22, 4723–4738 (2002).

    Article  CAS  Google Scholar 

  49. Suzuki, R. & Shimodaira, H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics 22, 1540–1542 (2006).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from the German Bundesministerium für Bildung und Forschung (Spitzencluster BioRN, Verbundprojekt Inkubator/Teilprojekt INE-TP01) to Cellzome AG. We are grateful to N. Garcia-Altrieth, M. Jundt, M. Löttgers, J.-I. Huber, M. Klös-Hudak, J. Krause, B. Kröh, A. Podszuweit, T. Rudi and K. Weis for expert technical assistance, to C. Gemünd and V. Wolowski for the development of software and database tools, and to F. Weisbrodt for help with the figures. We would like to thank T. Edwards, O. Rausch and D. Simmons for suggestions and support.

Author information

Authors and Affiliations

Authors

Contributions

A.D., D.E., A.-M.M., and K.S. performed biochemical and cell biological experiments; V.R. synthesized and sourced compounds; D.P. performed the interferon assay; I.B. analyzed histone modifications; B.D., M.D. and M. Boesche prepared peptide samples and operated mass spectrometers; M. Bantscheff, M.M.S., T.M. and G.S. established and conducted mass spectrometry data handling processes; M.M.S., Y.A., C. Huthmacher and J.S. contributed data analysis and visualization; M. Bantscheff, C. Hopf, P.G. and G.D. analyzed data, planned and supervised experiments, and conceptualized the project; G.B., U.K., G.N. and N.G.R. contributed ideas and supported the work; and M. Bantscheff and G.D. wrote the paper.

Corresponding authors

Correspondence to Marcus Bantscheff or Gerard Drewes.

Ethics declarations

Competing interests

The authors are employees of Cellzome AG or Cellzome UK Ltd. These companies funded the work.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–5, Supplementary Synthetic Procedures and Supplementary Figs. 1–10 (PDF 4173 kb)

Supplementary Data

Supplementary Data Sets 1–6 (ZIP 16869 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bantscheff, M., Hopf, C., Savitski, M. et al. Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat Biotechnol 29, 255–265 (2011). https://doi.org/10.1038/nbt.1759

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.1759

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research