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:

Precise small-molecule recognition of a toxic CUG RNA repeat expansion

Abstract

Excluding the ribosome and riboswitches, developing small molecules that selectively target RNA is a longstanding problem in chemical biology. A typical cellular RNA is difficult to target because it has little tertiary, but abundant secondary structure. We designed allele-selective compounds that target such an RNA, the toxic noncoding repeat expansion (r(CUG)exp) that causes myotonic dystrophy type 1 (DM1). We developed several strategies to generate allele-selective small molecules, including non-covalent binding, covalent binding, cleavage and on-site probe synthesis. Covalent binding and cleavage enabled target profiling in cells derived from individuals with DM1, showing precise recognition of r(CUG)exp. In the on-site probe synthesis approach, small molecules bound adjacent sites in r(CUG)exp and reacted to afford picomolar inhibitors via a proximity-based click reaction only in DM1-affected cells. We expanded this approach to image r(CUG)exp in its natural context.

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: A rationally designed small molecule improves DM1-associated defects in patient-derived cells.
Figure 2: Covalent (Chem-CLIP) approach to inhibit r(CUG)exp dysfunction and assess cellular selectivity.
Figure 3: Cleavage-based (Ribo-SNAP) approach to inhibit r(CUG)exp dysfunction and assess cellular selectivity.
Figure 4: r(CUG)exp-dependent synthesis of multivalent RNA-binding compounds in vitro and in cells.
Figure 5: FRET-based approach to image RNA targets in cells.

Similar content being viewed by others

References

  1. Schlünzen, F. et al. Structural basis for the interaction of antibiotics with the peptidyl transferase center in eubacteria. Nature 413, 814–821 (2001).

    Article  Google Scholar 

  2. Carter, A.P. et al. Functional insights from the structure of the 30S ribosomal subunit and its interactions with antibiotics. Nature 407, 340–348 (2000).

    Article  CAS  Google Scholar 

  3. Blount, K.F. et al. Novel riboswitch-binding flavin analog that protects mice against Clostridium difficile infection without inhibiting cecal flora. Antimicrob. Agents Chemother. 59, 5736–5746 (2015).

    Article  CAS  Google Scholar 

  4. Howe, J.A. et al. Selective small-molecule inhibition of an RNA structural element. Nature 526, 672–677 (2015).

    Article  CAS  Google Scholar 

  5. Brook, J.D. et al. Molecular basis of myotonic dystrophy: expansion of a trinucleotide (CTG) repeat at the 3′ end of a transcript encoding a protein kinase family member. Cell 68, 799–808 (1992).

    Article  CAS  Google Scholar 

  6. Jiang, H., Mankodi, A., Swanson, M.S., Moxley, R.T. & Thornton, C.A. Myotonic dystrophy type 1 is associated with nuclear foci of mutant RNA, sequestration of muscleblind proteins and deregulated alternative splicing in neurons. Hum. Mol. Genet. 13, 3079–3088 (2004).

    Article  CAS  Google Scholar 

  7. Taneja, K.L., McCurrach, M., Schalling, M., Housman, D. & Singer, R.H. Foci of trinucleotide repeat transcripts in nuclei of myotonic dystrophy cells and tissues. J. Cell Biol. 128, 995–1002 (1995).

    Article  CAS  Google Scholar 

  8. Furling, D., Lemieux, D., Taneja, K. & Puymirat, J. Decreased levels of myotonic dystrophy protein kinase (DMPK) and delayed differentiation in human myotonic dystrophy myoblasts. Neuromuscul. Disord. 11, 728–735 (2001).

    Article  CAS  Google Scholar 

  9. Hamshere, M.G., Newman, E.E., Alwazzan, M., Athwal, B.S. & Brook, J.D. Transcriptional abnormality in myotonic dystrophy affects DMPK but not neighboring genes. Proc. Natl. Acad. Sci. USA 94, 7394–7399 (1997).

    Article  CAS  Google Scholar 

  10. Johnson, L.F., Abelson, H.T., Penman, S. & Green, H. The relative amounts of the cytoplasmic RNA species in normal, transformed and senescent cultured cell lines. J. Cell. Physiol. 90, 465–470 (1977).

    Article  CAS  Google Scholar 

  11. Velagapudi, S.P., Gallo, S.M. & Disney, M.D. Sequence-based design of bioactive small molecules that target precursor microRNAs. Nat. Chem. Biol. 10, 291–297 (2014).

    Article  CAS  Google Scholar 

  12. Disney, M.D. et al. Inforna 2.0: a platform for the sequence-based design of small molecules targeting structured RNAs. ACS Chem. Biol. 11, 1720–1728 (2016).

    Article  CAS  Google Scholar 

  13. Pushechnikov, A. et al. Rational design of ligands targeting triplet repeating transcripts that cause RNA dominant disease: application to myotonic muscular dystrophy type 1 and spinocerebellar ataxia type 3. J. Am. Chem. Soc. 131, 9767–9779 (2009).

    Article  CAS  Google Scholar 

  14. Lee, M.M., Pushechnikov, A. & Disney, M.D. Rational and modular design of potent ligands targeting the RNA that causes myotonic dystrophy 2. ACS Chem. Biol. 4, 345–355 (2009).

    Article  CAS  Google Scholar 

  15. Childs-Disney, J.L., Hoskins, J., Rzuczek, S.G., Thornton, C.A. & Disney, M.D. Rationally designed small molecules targeting the RNA that causes myotonic dystrophy type 1 are potently bioactive. ACS Chem. Biol. 7, 856–862 (2012).

    Article  CAS  Google Scholar 

  16. Velagapudi, S.P. et al. Design of a small molecule against an oncogenic noncoding RNA. Proc. Natl. Acad. Sci. USA 113, 5898–5903 (2016).

    Article  CAS  Google Scholar 

  17. Rzuczek, S.G. et al. Features of modularly assembled compounds that impart bioactivity against an RNA target. ACS Chem. Biol. 8, 2312–2321 (2013).

    Article  CAS  Google Scholar 

  18. Childs-Disney, J.L. et al. Induction and reversal of myotonic dystrophy type 1 pre-mRNA splicing defects by small molecules. Nat. Commun. 4, 2044 (2013).

    Article  Google Scholar 

  19. Wojtkowiak-Szlachcic, A. et al. Short antisense-locked nucleic acids (all-LNAs) correct alternative splicing abnormalities in myotonic dystrophy. Nucleic Acids Res. 43, 3318–3331 (2015).

    Article  CAS  Google Scholar 

  20. Mastroyiannopoulos, N.P., Feldman, M.L., Uney, J.B., Mahadevan, M.S. & Phylactou, L.A. Woodchuck post-transcriptional element induces nuclear export of myotonic dystrophy 3′ untranslated region transcripts. EMBO Rep. 6, 458–463 (2005).

    Article  CAS  Google Scholar 

  21. Guan, L. & Disney, M.D. Covalent small-molecule-RNA complex formation enables cellular profiling of small-molecule-RNA interactions. Angew. Chem. Int. Edn Engl. 52, 10010–10013 (2013).

    Article  CAS  Google Scholar 

  22. Yang, W.Y., Wilson, H.D., Velagapudi, S.P. & Disney, M.D. Inhibition of non-ATG translational events in cells via covalent small molecules targeting RNA. J. Am. Chem. Soc. 137, 5336–5345 (2015).

    Article  CAS  Google Scholar 

  23. Carter, B.J. et al. Site-specific cleavage of RNA by Fe(II)-bleomycin. Proc. Natl. Acad. Sci. USA 87, 9373–9377 (1990).

    Article  CAS  Google Scholar 

  24. Lewis, W.G. et al. Click chemistry in situ: acetylcholinesterase as a reaction vessel for the selective assembly of a femtomolar inhibitor from an array of building blocks. Angew. Chem. Int. Edn Engl. 41, 1053–1057 (2002).

    Article  CAS  Google Scholar 

  25. Rzuczek, S.G., Park, H. & Disney, M.D. A toxic RNA catalyzes the in cellulo synthesis of its own inhibitor. Angew. Chem. Int. Edn Engl. 53, 10956–10959 (2014).

    Article  CAS  Google Scholar 

  26. Wang, E.T. et al. Transcriptome-wide regulation of pre-mRNA splicing and mRNA localization by muscleblind proteins. Cell 150, 710–724 (2012).

    Article  CAS  Google Scholar 

  27. Wheeler, T.M. et al. Reversal of RNA dominance by displacement of protein sequestered on triplet repeat RNA. Science 325, 336–339 (2009).

    Article  CAS  Google Scholar 

  28. Lima, W.F., Monia, B.P., Ecker, D.J. & Freier, S.M. Implication of RNA structure on antisense oligonucleotide hybridization kinetics. Biochemistry 31, 12055–12061 (1992).

    Article  CAS  Google Scholar 

  29. Lakowicz, J.R. Principles of Fluorescence Spectroscopy (Springer, 2006).

  30. Paige, J.S., Wu, K.Y. & Jaffrey, S.R. RNA mimics of green fluorescent protein. Science 333, 642–646 (2011).

    Article  CAS  Google Scholar 

  31. Strack, R.L., Disney, M.D. & Jaffrey, S.R. A superfolding Spinach2 reveals the dynamic nature of trinucleotide repeat-containing RNA. Nat. Methods 10, 1219–1224 (2013).

    Article  CAS  Google Scholar 

  32. Bertrand, E. et al. Localization of ASH1 mRNA particles in living yeast. Mol. Cell 2, 437–445 (1998).

    Article  CAS  Google Scholar 

  33. Baker, M. RNA imaging in situ. Nat. Methods 9, 787–790 (2012).

    Article  CAS  Google Scholar 

  34. Chen, C.Z. et al. Two high-throughput screening assays for aberrant RNA-protein interactions in myotonic dystrophy type 1. Anal. Bioanal. Chem. 402, 1889–1898 (2012).

    Article  CAS  Google Scholar 

  35. François, V. et al. Selective silencing of mutated mRNAs in DM1 by using modified hU7-snRNAs. Nat. Struct. Mol. Biol. 18, 85–87 (2011).

    Article  Google Scholar 

  36. Shapiro, I.M. et al. An EMT-driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype. PLoS Genet. 7, e1002218 (2011).

    Article  CAS  Google Scholar 

  37. Warf, M.B., Nakamori, M., Matthys, C.M., Thornton, C.A. & Berglund, J.A. Pentamidine reverses the splicing defects associated with myotonic dystrophy. Proc. Natl. Acad. Sci. USA 106, 18551–18556 (2009).

    Article  CAS  Google Scholar 

  38. Holt, I. et al. Defective mRNA in myotonic dystrophy accumulates at the periphery of nuclear splicing speckles. Genes Cells 12, 1035–1048 (2007).

    Article  CAS  Google Scholar 

  39. Hampf, M. & Gossen, M. A protocol for combined Photinus and Renilla luciferase quantification compatible with protein assays. Anal. Biochem. 356, 94–99 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank T. Kodadek, G. Joyce, W. Ja, J. Childs-Disney, K. Sobczak and J. Cleveland for advice and critical review of the manuscript, and M.D.D. acknowledges J. and H. (nee McDougall) Disney. We also thank the platform for immortalization of human cells from the Institut de Myologie. This work was funded by the US National Institutes of Health (grants DP1NS096898 to M.D.D. and DP1NS096787 to R.Y.) and the Muscular Dystrophy Association (grant 380467 to M.D.D.). S.G.R. was partially supported by a postdoctoral fellowship from the Myotonic Dystrophy Foundation.

Author information

Authors and Affiliations

Authors

Contributions

M.D.D. directed the study, conceived of the ideas and designed experiments. S.G.R. designed experiments, synthesized all of the compounds and conducted all of the biochemical and cellular studies. Y.N. contributed to the synthesis of compounds. L.A.C. performed the FRET imaging. R.Y. contributed to the FRET studies. M.D.C. contributed to compound stability studies. D.F. provided critical reagents.

Corresponding author

Correspondence to Matthew D Disney.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Figures 1–14 and Supplementary Tables 1 and 2. (PDF 3349 kb)

Supplementary Note

Synthetic Procedures. (PDF 3737 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rzuczek, S., Colgan, L., Nakai, Y. et al. Precise small-molecule recognition of a toxic CUG RNA repeat expansion. Nat Chem Biol 13, 188–193 (2017). https://doi.org/10.1038/nchembio.2251

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2251

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