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:

Genome-wide localization of small molecules

Subjects

Abstract

A vast number of small-molecule ligands, including therapeutic drugs under development and in clinical use, elicit their effects by binding specific proteins associated with the genome. An ability to map the direct interactions of a chemical entity with chromatin genome-wide could provide important insights into chemical perturbation of cellular function. Here we describe a method that couples ligand-affinity capture and massively parallel DNA sequencing (Chem-seq) to identify the sites bound by small chemical molecules throughout the human genome. We show how Chem-seq can be combined with ChIP-seq to gain unique insights into the interaction of drugs with their target proteins throughout the genome of tumor cells. These methods will be broadly useful to enhance understanding of therapeutic action and to characterize the specificity of chemical entities that interact with DNA or genome-associated proteins.

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: Chem-seq from intact cells or cellular lysates reveals genomic sites bound by the BET bromodomain-targeting drug JQ1.
Figure 2: Genome-wide drug target analysis.
Figure 3: Chem-seq reveals genomic occupancy of a protein kinase inhibitor and a DNA-intercalating drug.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Bell, O., Tiwari, V.K., Thoma, N.H. & Schubeler, D. Determinants and dynamics of genome accessibility. Nat. Rev. Genet. 12, 554–564 (2011).

    Article  CAS  Google Scholar 

  2. Gilbert, D.M. Evaluating genome-scale approaches to eukaryotic DNA replication. Nat. Rev. Genet. 11, 673–684 (2010).

    Article  CAS  Google Scholar 

  3. Hawkins, R.D., Hon, G.C. & Ren, B. Next-generation genomics: an integrative approach. Nat. Rev. Genet. 11, 476–486 (2010).

    Article  CAS  Google Scholar 

  4. MacQuarrie, K.L., Fong, A.P., Morse, R.H. & Tapscott, S.J. Genome-wide transcription factor binding: beyond direct target regulation. Trends Genet. 27, 141–148 (2011).

    Article  CAS  Google Scholar 

  5. Neph, S. et al. Circuitry and dynamics of human transcription factor regulatory networks. Cell 150, 1274–1286 (2012).

    Article  CAS  Google Scholar 

  6. Zhou, V.W., Goren, A. & Bernstein, B.E. Charting histone modifications and the functional organization of mammalian genomes. Nat. Rev. Genet. 12, 7–18 (2011).

    Article  Google Scholar 

  7. Ng, H.H. & Surani, M.A. The transcriptional and signalling networks of pluripotency. Nat. Cell Biol. 13, 490–496 (2011).

    Article  CAS  Google Scholar 

  8. Northrup, D.L. & Zhao, K. Application of ChIP-Seq and related techniques to the study of immune function. Immunity 34, 830–842 (2011).

    Article  CAS  Google Scholar 

  9. Orkin, S.H. & Hochedlinger, K. Chromatin connections to pluripotency and cellular reprogramming. Cell 145, 835–850 (2011).

    Article  CAS  Google Scholar 

  10. Young, R.A. Control of the embryonic stem cell state. Cell 144, 940–954 (2011).

    Article  CAS  Google Scholar 

  11. Arrowsmith, C.H., Bountra, C., Fish, P.V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 11, 384–400 (2012).

    Article  CAS  Google Scholar 

  12. Copeland, R.A., Solomon, M.E. & Richon, V.M. Protein methyltransferases as a target class for drug discovery. Nat. Rev. Drug Discov. 8, 724–732 (2009).

    Article  CAS  Google Scholar 

  13. Dawson, M.A. et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533 (2011).

    Article  CAS  Google Scholar 

  14. Deshpande, A.J., Bradner, J. & Armstrong, S.A. Chromatin modifications as therapeutic targets in MLL-rearranged leukemia. Trends Immunol. 33, 563–570 (2012).

    Article  CAS  Google Scholar 

  15. Sandoval, J. & Esteller, M. Cancer epigenomics: beyond genomics. Curr. Opin. Genet. Dev. 22, 50–55 (2012).

    Article  CAS  Google Scholar 

  16. You, J.S. & Jones, P.A. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell 22, 9–20 (2012).

    Article  CAS  Google Scholar 

  17. Raida, M. Drug target deconvolution by chemical proteomics. Curr. Opin. Chem. Biol. 15, 570–575 (2011).

    Article  CAS  Google Scholar 

  18. Bantscheff, M. et al. Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat. Biotechnol. 29, 255–265 (2011).

    Article  CAS  Google Scholar 

  19. Ito, T. et al. Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350 (2010).

    Article  CAS  Google Scholar 

  20. 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 

  21. Tung, S.Y., Hong, J.Y., Walz, T., Moazed, D. & Liou, G.G. Chromatin affinity-precipitation using a small metabolic molecule: its application to analysis of O-acetyl-ADP-ribose. Cell. Mol. Life Sci. 69, 641–650 (2012).

    Article  CAS  Google Scholar 

  22. Chung, C.W. et al. Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J. Med. Chem. 54, 3827–3838 (2011).

    Article  CAS  Google Scholar 

  23. Delmore, J.E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  Google Scholar 

  24. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  Google Scholar 

  25. Loven, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  CAS  Google Scholar 

  26. Nicodeme, E. et al. Suppression of inflammation by a synthetic histone mimic. Nature 468, 1119–1123 (2010).

    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. Santo, L. et al. AT7519, A novel small molecule multi-cyclin-dependent kinase inhibitor, induces apoptosis in multiple myeloma via GSK-3beta activation and RNA polymerase II inhibition. Oncogene 29, 2325–2336 (2010).

    Article  CAS  Google Scholar 

  29. Zhou, Q., Li, T. & Price, D.H. RNA polymerase II elongation control. Annu. Rev. Biochem. 81, 119–143 (2012).

    Article  CAS  Google Scholar 

  30. Squires, M.S. et al. AT7519, a cyclin-dependent kinase inhibitor, exerts its effects by transcriptional inhibition in leukemia cell lines and patient samples. Mol. Cancer Ther. 9, 920–928 (2010).

    Article  CAS  Google Scholar 

  31. Kouzine, F. et al. Transcription-dependent dynamic supercoiling is a short-range genomic force. Nat. Struct. Mol. Biol. 20, 396–403 (2013).

    Article  CAS  Google Scholar 

  32. Naughton, C. et al. Transcription forms and remodels supercoiling domains unfolding large-scale chromatin structures. Nat. Struct. Mol. Biol. 20, 387–395 (2013).

    Article  CAS  Google Scholar 

  33. Marson, A. et al. Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 134, 521–533 (2008).

    Article  CAS  Google Scholar 

  34. Lee, T.I., Johnstone, S.E. & Young, R.A. Chromatin immunoprecipitation and microarray-based analysis of protein location. Nat. Protoc. 1, 729–748 (2006).

    Article  CAS  Google Scholar 

  35. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  Google Scholar 

  36. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  Google Scholar 

  37. Guenther, M.G., Levine, S.S., Boyer, L.A., Jaenisch, R. & Young, R.A. A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130, 77–88 (2007).

    Article  CAS  Google Scholar 

  38. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  Google Scholar 

  39. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  Google Scholar 

  40. Kagey, M.H. et al. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467, 430–435 (2010).

    Article  CAS  Google Scholar 

  41. Rahl, P.B. et al. c-Myc regulates transcriptional pause release. Cell 141, 432–445 (2010).

    Article  CAS  Google Scholar 

  42. 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).

    Article  CAS  Google Scholar 

  43. Ross-Innes, C.S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank T. Volkert, J. Love, J.-A. Kwon and S. Gupta at the Whitehead Genome Technology Core for Solexa sequencing, E. Cohick for help with cell culture, and A. Federation and R. St. Pierre at the Dana-Farber Cancer Institute for biochemistry support. This work was supported by US National Institutes of Health grants HG002668 (R.A.Y.), CA109901 (R.A.Y.) and CA146445 (R.A.Y., T.I.L.), Swedish Research Council Postdoctoral Fellowship VR-B0086301 (J.L.), American Cancer Society Postdoctoral Fellowship PF-11-042- 01-DMC (P.B.R.), the Leukemia & Lymphoma Society (J.Q. and J.E.B.) and the Damon-Runyon Cancer Research Foundation (J.E.B.).

Author information

Authors and Affiliations

Authors

Contributions

J.J.M., P.B.R., J.E.B. and R.A.Y. conceived of the Chem-seq method, L.A. and M.G.G. developed the method, L.A. generated bio-JQ1 and bio-psoralen Chem-seq data, M.G.G. generated bio-AT7519 Chem-seq data, Z.P.F. developed computational methods and analyzed the data, J.Q. and J.J.M. synthesized biotinylated derivatives of chemical probes, J.Q. performed protein biochemistry, L.A. generated ChIP-seq data for BRD2, BRD3, BRD4, CDK7, CDK8, H4K20me3 and H3K27me3, P.B.R. and J.L. generated ChIP-seq data for RNA Pol II and CDK9, W.B.S. generated cellular proliferation data, A.A.S. contributed to optimize Chem-seq, T.I.L. provided advice on method development, and J.E.B. and R.A.Y. supervised the research.

Corresponding authors

Correspondence to James E Bradner or Richard A Young.

Ethics declarations

Competing interests

J.E.B. and R.A.Y. are founders of Syros Pharmaceuticals. J.J.M., P.B.R., M.G.G. and J.L. are employees of Syros Pharmaceuticals.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6, Supplementary Methods and Supplementary Table 1 (PDF 4352 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Anders, L., Guenther, M., Qi, J. et al. Genome-wide localization of small molecules. Nat Biotechnol 32, 92–96 (2014). https://doi.org/10.1038/nbt.2776

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2776

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