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:

A small molecule mitigates hearing loss in a mouse model of Usher syndrome III

Abstract

Usher syndrome type III (USH3), characterized by progressive deafness, variable balance disorder and blindness, is caused by destabilizing mutations in the gene encoding the clarin-1 (CLRN1) protein. Here we report a new strategy to mitigate hearing loss associated with a common USH3 mutation CLRN1N48K that involves cell-based high-throughput screening of small molecules capable of stabilizing CLRN1N48K, followed by a secondary screening to eliminate general proteasome inhibitors, and finally an iterative process to optimize structure–activity relationships. This resulted in the identification of BioFocus 844 (BF844). To test the efficacy of BF844, we developed a mouse model that mimicked the progressive hearing loss associated with USH3. BF844 effectively attenuated progressive hearing loss and prevented deafness in this model. Because the CLRN1N48K mutation causes both hearing and vision loss, BF844 could in principle prevent both sensory deficiencies in patients with USH3. Moreover, the strategy described here could help identify drugs for other protein-destabilizing monogenic disorders.

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

Figure 1: HTS identifies compounds that stabilize human CLRN1N48K.
Figure 2: A dual-reporter assay eliminates pan-proteasome inhibitors.
Figure 3: The lead compound O03 stabilizes CLRN1N48K by a post-translational mechanism.
Figure 4: Design of BF844; identification of proteins bound to its pharmacophore.
Figure 5: BF844 preserves hearing in Tg;KI/KI mice.

Similar content being viewed by others

References

  1. Pakarinen, L., Karjalainen, S., Simola, K.O., Laippala, P. & Kaitalo, H. Usher's syndrome type 3 in Finland. Laryngoscope 105, 613–617 (1995).

    Article  CAS  Google Scholar 

  2. Ness, S.L. et al. Genetic homogeneity and phenotypic variability among Ashkenazi Jews with Usher syndrome type III. J. Med. Genet. 40, 767–772 (2003).

    Article  CAS  Google Scholar 

  3. Plantinga, R.F. et al. Serial audiometry and speech recognition findings in Finnish Usher syndrome type III patients. Audiol. Neurootol. 10, 79–89 (2005).

    Article  CAS  Google Scholar 

  4. Joensuu, T. et al. Mutations in a novel gene with transmembrane domains underlie Usher syndrome type 3. Am. J. Hum. Genet. 69, 673–684 (2001).

    Article  CAS  Google Scholar 

  5. Adato, A. et al. USH3A transcripts encode clarin-1, a four-transmembrane-domain protein with a possible role in sensory synapses. Eur. J. Hum. Genet. 10, 339–350 (2002).

    Article  CAS  Google Scholar 

  6. Tian, G. et al. Clarin-1, encoded by the Usher Syndrome III causative gene, forms a membranous microdomain: possible role of clarin-1 in organizing the actin cytoskeleton. J. Biol. Chem. 284, 18980–18993 (2009).

    Article  CAS  Google Scholar 

  7. Geng, R. et al. The mechanosensory structure of the hair cell requires clarin-1, a protein encoded by Usher syndrome III causative gene. J. Neurosci. 32, 9485–9498 (2012).

    Article  CAS  Google Scholar 

  8. Gopal, S.R. et al. Zebrafish models for the mechanosensory hair cell dysfunction in Usher Syndrome 3 reveal that clarin-1 is an essential hair bundle protein. J. Neurosci. 35, 10188–10201 (2015).

    Article  CAS  Google Scholar 

  9. Zhang, J.H., Chung, T.D. & Oldenburg, K.R. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J. Biomol. Screen. 4, 67–73 (1999).

    Article  CAS  Google Scholar 

  10. Baell, J.B. & Holloway, G.A. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 53, 2719–2740 (2010).

    Article  CAS  Google Scholar 

  11. Cumming, J.G., Davis, A.M., Muresan, S., Haeberlein, M. & Chen, H. Chemical predictive modelling to improve compound quality. Nat. Rev. Drug Discov. 12, 948–962 (2013).

    Article  CAS  Google Scholar 

  12. Li, X. et al. Generation of destabilized green fluorescent protein as a transcription reporter. J. Biol. Chem. 273, 34970–34975 (1998).

    Article  CAS  Google Scholar 

  13. Kisselev, A.F., van der Linden, W.A. & Overkleeft, H.S. Proteasome inhibitors: an expanding army attacking a unique target. Chem. Biol. 19, 99–115 (2012).

    Article  CAS  Google Scholar 

  14. Polli, J.W. et al. Rational use of in vitro P-glycoprotein assays in drug discovery. J. Pharmacol. Exp. Ther. 299, 620–628 (2001).

    CAS  PubMed  Google Scholar 

  15. Schinkel, A.H. et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell 77, 491–502 (1994).

    Article  CAS  Google Scholar 

  16. Schulte, T.W. & Neckers, L.M. The benzoquinone ansamycin 17-allylamino-17-demethoxygeldanamycin binds to HSP90 and shares important biologic activities with geldanamycin. Cancer Chemother. Pharmacol. 42, 273–279 (1998).

    Article  CAS  Google Scholar 

  17. Sakami, S. et al. Probing mechanisms of photoreceptor degeneration in a new mouse model of the common form of autosomal dominant retinitis pigmentosa due to P23H opsin mutations. J. Biol. Chem. 286, 10551–10567 (2011).

    Article  CAS  Google Scholar 

  18. Körschen, H.G. et al. A 240 kDa protein represents the complete beta subunit of the cyclic nucleotide-gated channel from rod photoreceptor. Neuron 15, 627–636 (1995).

    Article  Google Scholar 

  19. Chen, Y. et al. A high-throughput drug screening strategy for detecting rhodopsin P23H mutant rescue and degradation. Invest. Ophthalmol. Vis. Sci. 56, 2553–2567 (2015).

    Article  CAS  Google Scholar 

  20. Sakami, S., Kolesnikov, A.V., Kefalov, V.J. & Palczewski, K. P23H opsin knock-in mice reveal a novel step in retinal rod disc morphogenesis. Hum. Mol. Genet. 23, 1723–1741 (2014).

    Article  CAS  Google Scholar 

  21. Chiang, W.C. et al. Robust endoplasmic reticulum-associated degradation of rhodopsin precedes retinal degeneration. Mol. Neurobiol. 52, 679–695 (2015).

    Article  CAS  Google Scholar 

  22. Nemet, I., Tian, G. & Imanishi, Y. Organization of cGMP sensing structures on the rod photoreceptor outer segment plasma membrane. Channels 8, 528–535 (2014).

    Article  Google Scholar 

  23. Lumpkin, E.A. et al. Math1-driven GFP expression in the developing nervous system of transgenic mice. Gene Expr. Patterns 3, 389–395 (2003).

    Article  CAS  Google Scholar 

  24. Zhou, X., Jen, P.H., Seburn, K.L., Frankel, W.N. & Zheng, Q.Y. Auditory brainstem responses in 10 inbred strains of mice. Brain Res. 1091, 16–26 (2006).

    Article  CAS  Google Scholar 

  25. Burkard, R.F., Manuel, D. & Eggermont, J.J. (eds.) Auditory Evoked Potentials: Basic Principles and Clinical Application (Lippincott Williams & Wilkins, 2007).

  26. Lobanova, E.S., Finkelstein, S., Skiba, N.P. & Arshavsky, V.Y. Proteasome overload is a common stress factor in multiple forms of inherited retinal degeneration. Proc. Natl. Acad. Sci. USA 110, 9986–9991 (2013).

    Article  CAS  Google Scholar 

  27. Lee, J.N. et al. Proteasome inhibitors induce auditory hair cell death through peroxisome dysfunction. Biochem. Biophys. Res. Commun. 456, 269–274 (2015).

    Article  CAS  Google Scholar 

  28. Shearer, A.E. et al. Comprehensive genetic testing for hereditary hearing loss using massively parallel sequencing. Proc. Natl. Acad. Sci. USA 107, 21104–21109 (2010).

    Article  CAS  Google Scholar 

  29. Wiseman, R.L., Haynes, C.M. & Ron, D. SnapShot: The unfolded protein response. Cell 140, e592 (2010).

    Article  Google Scholar 

  30. Schröder, M. & Kaufman, R.J. The mammalian unfolded protein response. Annu. Rev. Biochem. 74, 739–789 (2005).

    Article  Google Scholar 

  31. Claessen, J.H., Kundrat, L. & Ploegh, H.L. Protein quality control in the ER: balancing the ubiquitin checkbook. Trends Cell Biol. 22, 22–32 (2012).

    Article  CAS  Google Scholar 

  32. Ward, C.L., Omura, S. & Kopito, R.R. Degradation of CFTR by the ubiquitin-proteasome pathway. Cell 83, 121–127 (1995).

    Article  CAS  Google Scholar 

  33. Luo, Y., McDonald, K. & Hanrahan, J.W. Trafficking of immature DeltaF508-CFTR to the plasma membrane and its detection by biotinylation. Biochem. J. 419, 211–219 (2009).

    Article  CAS  Google Scholar 

  34. Gee, H.Y., Noh, S.H., Tang, B.L., Kim, K.H. & Lee, M.G. Rescue of ΔF508-CFTR trafficking via a GRASP-dependent unconventional secretion pathway. Cell 146, 746–760 (2011).

    Article  CAS  Google Scholar 

  35. Geng, R. et al. Usher syndrome IIIA gene clarin-1 is essential for hair cell function and associated neural activation. Hum. Mol. Genet. 18, 2748–2760 (2009).

    Article  CAS  Google Scholar 

  36. Geller, S.F. et al. CLRN1 is nonessential in the mouse retina but is required for cochlear hair cell development. PLoS Genet. 5, e1000607 (2009).

    Article  Google Scholar 

  37. Tian, G., Lee, R., Ropelewski, P. & Imanishi, Y. Impairment of vision in a mouse model of Usher syndrome type III. Invest. Ophthalmol. Vis. Sci. 57, 866–875 (2016).

    Article  CAS  Google Scholar 

  38. Wang, X. et al. Hsp90 cochaperone Aha1 downregulation rescues misfolding of CFTR in cystic fibrosis. Cell 127, 803–815 (2006).

    Article  CAS  Google Scholar 

  39. Petersen, T.N., Brunak, S., von Heijne, G. & Nielsen, H. SignalP 4.0: discriminating signal peptides from transmembrane regions. Nat. Methods 8, 785–786 (2011).

    Article  CAS  Google Scholar 

  40. Nagai, T. et al. A variant of yellow fluorescent protein with fast and efficient maturation for cell-biological applications. Nat. Biotechnol. 20, 87–90 (2002).

    Article  CAS  Google Scholar 

  41. MacKenzie, D., Arendt, A., Hargrave, P., McDowell, J.H. & Molday, R.S. Localization of binding sites for carboxyl terminal specific anti-rhodopsin monoclonal antibodies using synthetic peptides. Biochemistry 23, 6544–6549 (1984).

    Article  CAS  Google Scholar 

  42. Hajkova, D. et al. Proteomic changes in the photoreceptor outer segment upon intense light exposure. J. Proteome Res. 9, 1173–1181 (2010).

    Article  CAS  Google Scholar 

  43. Guo, Y., Miyagi, M., Zeng, R. & Sheng, Q. O18Quant: a semiautomatic strategy for quantitative analysis of high-resolution 16O/18O labeled data. BioMed Res. Int. 2014, 971857 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Santhoshkumar, P. & Sharma, K.K. Phe71 is essential for chaperone-like function in alpha A-crystallin. J. Biol. Chem. 276, 47094–47099 (2001).

    Article  CAS  Google Scholar 

  45. Lodowski, K.H. et al. Signals governing the trafficking and mistrafficking of a ciliary GPCR, rhodopsin. J. Neurosci. 33, 13621–13638 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Usher III Initiative (Y.I., K.N.A. and BioFocus group), the Maniglia Endowed Funds (K.N.A.), Hope for Vision (Y.I.), the Prince Family Foundation (Y.I.), start-up funding from Case Western Reserve University (CWRU) (Y.I.), the US National Institutes of Health (NIH) (grants R01-DC010816 (to K.N.A.), R01-EY020826 (to Y.I.) and R24-EY021126 (to K.P.)), and the Arnold and Mabel Beckman Foundation (K.P.). We thank the Elden family and the Prince Family Foundation for their generous support of our research via the Usher III Initiative, D. Saperstein for his leadership of the Initiative's research efforts, and A.R. Moise for useful suggestions and discussions before the inception of this project. We thank R. Papoian, S. Nelson and W. Seibel at the Drug Discovery Center (University of Cincinnati) for conducting the HTS. The monoclonal antibody E7 developed by M. Klymkowsky was obtained from the Developmental Studies Hybridoma Bank created by the National Institute of Child Health and Human Development (NICHD) of the NIH, and maintained at The University of Iowa, Iowa City, USA. Fluorescence microscopy was conducted to optimize the HTS assay at the light microscopy imaging core facility of CWRU. We thank Y. Chen at CWRU for providing the RHOP23H NIH/3T3 cell line, and L.T. Webster Jr. for insightful comments on this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Y.I. and K.P. conceived the small-molecule screening experiments and designed the small-molecule HTS assay, and G.T. and R.L. contributed to the establishment of the assay; G.T., I.N. and R.L. conducted the experiments characterizing small molecules; I.N. and M.M. performed mass spectroscopy and associated experiments; K.N.A. conceived the transgenic mouse models TgAC1 and TgAC1;KI/KI; K.N.A., S.R.G., R.G. and D.H.-C.C. were involved in the generation of TgAC1;KI/KI mice; S.R.G. characterized the hearing loss profile in TgAC1;KI/KI mice; K.N.A., S.R.G. and D.H.-C.C. conceived the dosing scheme for regimen II; S.R.G. and D.H.-C.C. performed efficacy testing associated with regimens I and II; K.F.M., C.J.L., W.R.K.E. and A.P.O. performed the medicinal chemistry experiments; N.A.L. accommodated the small-molecule screening assay at BioFocus; K.O. and F.A. conducted the small-molecule screening assay at BioFocus; K.F.M., N.A.L., D.F.F., R.W.B., A.M.M. and W.E.H. provided project supervision at BioFocus; Y.I. and K.N.A. provided project supervision at CWRU; I.N. and K.F.M. wrote the results section associated with medicinal chemistry and target identification; and K.N.A., K.P. and Y.I. wrote the manuscript.

Corresponding authors

Correspondence to Kumar N Alagramam or Yoshikazu Imanishi.

Ethics declarations

Competing interests

Usher III Initiative contracted K.F.M., C.J.L., W.R.K.E., A.P.O., N.A.L., K.O., F.A., D.F.F., R.W.B. and A.M.M. from BioFocus to carry out hit-to-lead optimization of small molecules described in this report. W.E.H. serves as the director of pharmaceutical development for the Usher III Initiative and as chief translational officer for the School of Medicine at CWRU. W.E.H., R.W.B., K.F.M., C.J.L., W.R.K.E. and A.P.O. are named on patents WO 2014066835, WO 2014066836 and WO 2012148994. The mouse Clrn1 cDNA used to generate transgenic mouse TgAC1 and the human ortholog are protected by provisional US patent 62/076,114 to K.N.A.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Figures 1–13 and Supplementary Tables 1–4. (PDF 3712 kb)

Supplementary Note

Synthetic Procedures (PDF 3546 kb)

Supplementary Data Set 1

The 90 prioritized screening hits and 48 (highlighted structures) selected for further characterization. (PDF 1699 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alagramam, K., Gopal, S., Geng, R. et al. A small molecule mitigates hearing loss in a mouse model of Usher syndrome III. Nat Chem Biol 12, 444–451 (2016). https://doi.org/10.1038/nchembio.2069

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2069

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