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:

Rescue of hearing and vestibular function by antisense oligonucleotides in a mouse model of human deafness

Abstract

Hearing impairment is the most common sensory disorder, with congenital hearing impairment present in approximately 1 in 1,000 newborns 1 . Hereditary deafness is often mediated by the improper development or degeneration of cochlear hair cells 2 . Until now, it was not known whether such congenital failures could be mitigated by therapeutic intervention 3, 4, 5 . Here we show that hearing and vestibular function can be rescued in a mouse model of human hereditary deafness. An antisense oligonucleotide (ASO) was used to correct defective pre-mRNA splicing of transcripts from the USH1C gene with the c.216G>A mutation, which causes human Usher syndrome, the leading genetic cause of combined deafness and blindness 6, 7 . Treatment of neonatal mice with a single systemic dose of ASO partially corrects Ush1c c.216G>A splicing, increases protein expression, improves stereocilia organization in the cochlea, and rescues cochlear hair cells, vestibular function and low-frequency hearing in mice. These effects were sustained for several months, providing evidence that congenital deafness can be effectively overcome by treatment early in development to correct gene expression and demonstrating the therapeutic potential of ASOs in the treatment of deafness.

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: Correction of USH1C 216A splicing using ASOs.
Figure 2: ASOs correct vestibular function and rescue hearing in 216AA mice.
Figure 3: ASO-29 treatment corrects mRNA splicing and harmonin protein expression and prevents cochlear hair cell loss in 216AA mice.
Figure 4: Restoration of hair cell stereocilia bundle shape in mice.

Similar content being viewed by others

References

  1. Morton, C.C. & Nance, W.E. Newborn hearing screening—a silent revolution. N. Engl. J. Med. 354, 2151–2164 (2006).

    Article  CAS  Google Scholar 

  2. Dror, A.A. & Avraham, K.B. Hearing loss: mechanisms revealed by genetics and cell biology. Annu. Rev. Genet. 43, 411–437 (2009).

    Article  CAS  Google Scholar 

  3. Conde de Felipe, M.M., Feijoo Redondo, A.F., Garcia-Sancho, J., Schimmang, T. & Alonso, M.B. Cell- and gene-therapy approaches to inner ear repair. Histol. Histopathol. 26, 923–940 (2011).

    CAS  PubMed  Google Scholar 

  4. Di Domenico, M. et al. Towards gene therapy for deafness. J. Cell. Physiol. 226, 2494–2499 (2011).

    Article  CAS  Google Scholar 

  5. Bermingham-McDonogh, O. & Reh, T.A. Regulated reprogramming in the regeneration of sensory receptor cells. Neuron 71, 389–405 (2011).

    Article  CAS  Google Scholar 

  6. Bitner-Glindzicz, M. et al. A recessive contiguous gene deletion causing infantile hyperinsulinism, enteropathy and deafness identifies the Usher type 1C gene. Nat. Genet. 26, 56–60 (2000).

    Article  CAS  Google Scholar 

  7. Verpy, E. et al. A defect in Harmonin, a PDZ domain-containing protein expressed in the inner ear sensory hair cells, underlies Usher syndrome type 1C. Nat. Genet. 26, 51–55 (2000).

    Article  CAS  Google Scholar 

  8. Kimberling, W.J. et al. Frequency of Usher syndrome in two pediatric populations: Implications for genetic screening of deaf and hard of hearing children. Genet. Med. 12, 512–516 (2010).

    Article  CAS  Google Scholar 

  9. Ouyang, X.M. et al. Characterization of Usher syndrome type I gene mutations in an Usher syndrome patient population. Hum. Genet. 116, 292–299 (2005).

    Article  CAS  Google Scholar 

  10. Ebermann, I. et al. Deafblindness in French Canadians from Quebec: a predominant founder mutation in the USH1C gene provides the first genetic link with the Acadian population. Genome Biol. 8, R47 (2007).

    Article  Google Scholar 

  11. Ouyang, X.M. et al. USH1C: a rare cause of USH1 in a non-Acadian population and a founder effect of the Acadian allele. Clin. Genet. 63, 150–153 (2003).

    Article  CAS  Google Scholar 

  12. Lentz, J. et al. The USH1C 216G→A splice-site mutation results in a 35-base-pair deletion. Hum. Genet. 116, 225–227 (2005).

    Article  CAS  Google Scholar 

  13. Lentz, J., Pan, F., Ng, S.S., Deininger, P. & Keats, B. Ush1c216A knock-in mouse survives Katrina. Mutat. Res. 616, 139–144 (2007).

    Article  CAS  Google Scholar 

  14. Lentz, J.J. et al. Deafness and retinal degeneration in a novel USH1C knock-in mouse model. Dev. Neurobiol. 70, 253–267 (2010).

    Article  CAS  Google Scholar 

  15. Hardisty-Hughes, R.E., Parker, A. & Brown, S.D. A hearing and vestibular phenotyping pipeline to identify mouse mutants with hearing impairment. Nat. Protoc. 5, 177–190 (2010).

    Article  CAS  Google Scholar 

  16. Michalski, N. et al. Harmonin-b, an actin-binding scaffold protein, is involved in the adaptation of mechanoelectrical transduction by sensory hair cells. Pflugers Arch. 459, 115–130 (2009).

    Article  CAS  Google Scholar 

  17. Grillet, N. et al. Harmonin mutations cause mechanotransduction defects in cochlear hair cells. Neuron 62, 375–387 (2009).

    Article  CAS  Google Scholar 

  18. Lefèvre, G. et al. A core cochlear phenotype in USH1 mouse mutants implicates fibrous links of the hair bundle in its cohesion, orientation and differential growth. Development 135, 1427–1437 (2008).

    Article  Google Scholar 

  19. Boëda, B. et al. Myosin VIIa, harmonin and cadherin 23, three Usher I gene products that cooperate to shape the sensory hair cell bundle. EMBO J. 21, 6689–6699 (2002).

    Article  Google Scholar 

  20. Peng, A.W., Salles, F.T., Pan, B. & Ricci, A.J. Integrating the biophysical and molecular mechanisms of auditory hair cell mechanotransduction. Nat. Commun. 2, 523 (2011).

    Article  Google Scholar 

  21. Müller, M., von Hunerbein, K., Hoidis, S. & Smolders, J.W. A physiological place-frequency map of the cochlea in the CBA/J mouse. Hear. Res. 202, 63–73 (2005).

    Article  Google Scholar 

  22. Greenwood, D.D. A cochlear frequency-position function for several species—29 years later. J. Acoust. Soc. Am. 87, 2592–2605 (1990).

    Article  CAS  Google Scholar 

  23. Hua, Y. et al. Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model. Nature 478, 123–126 (2011).

    Article  CAS  Google Scholar 

  24. Wheeler, T.M. et al. Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature 488, 111–115 (2012).

    Article  CAS  Google Scholar 

  25. El-Amraoui, A. & Petit, C. Usher I syndrome: unravelling the mechanisms that underlie the cohesion of the growing hair bundle in inner ear sensory cells. J. Cell Sci. 118, 4593–4603 (2005).

    Article  CAS  Google Scholar 

  26. Petit, C. & Richardson, G.P. Linking genes underlying deafness to hair-bundle development and function. Nat. Neurosci. 12, 703–710 (2009).

    Article  CAS  Google Scholar 

  27. Hall, J.W. III. Development of the ear and hearing. J. Perinatol. 20, S12–S20 (2000).

    Article  Google Scholar 

  28. Uhlmann, R.A., Taylor, M., Meyer, N.L. & Mari, G. Fetal transfusion: the spectrum of clinical research in the past year. Curr. Opin. Obstet. Gynecol. 22, 155–158 (2010).

    Article  Google Scholar 

  29. Baker, B.F. et al. 2′-O-(2-methoxy)ethyl–modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 mRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. J. Biol. Chem. 272, 11994–12000 (1997).

    Article  CAS  Google Scholar 

  30. Hastings, M.L. et al. Tetracyclines that promote SMN2 exon 7 splicing as therapeutics for spinal muscular atrophy. Sci. Transl. Med. 1, 5ra12 (2009).

    Article  Google Scholar 

  31. Hardie, N.A., MacDonald, G. & Rubel, E.W. A new method for imaging and 3D reconstruction of mammalian cochlea by fluorescent confocal microscopy. Brain Res. 1000, 200–210 (2004).

    Article  CAS  Google Scholar 

  32. Sage, C., Venteo, S., Jeromin, A., Roder, J. & Dechesne, C.J. Distribution of frequenin in the mouse inner ear during development, comparison with other calcium-binding proteins and synaptophysin. Hear. Res. 150, 70–82 (2000).

    Article  CAS  Google Scholar 

  33. Milliken, G.A. & Johnson, D.E. Analysis of Messy Data Volume I: Designed Experiments Ch. 30, 413–423 (Lifetime Learning Publications, Belmont, California, 1984).

    Google Scholar 

  34. Edwards, D. & Berry, J.J. The efficiency of simulation-based multiple comparisons. Biometrics 43, 913–928 (1987).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge support from the Hearing Health Foundation, Midwest Eye-Banks, the National Organization for Hearing Research Foundation, Capita Foundation and the US National Institutes of Health. We thank D. Cunningham and E. Rubel for assistance with scanning electron microscopy analysis; A. Rosenkranz, R. Marr and M. Oblinger for use of equipment; J. Huang for assistance with open-field analysis, L. Ochoa for assistance with ABR analysis computer support, G. MacDonald for assistance with confocal imaging and deconvolution analysis, U. Wolfrum (Johannes Gutenberg University of Mainz) for harmonin b–specific antibodies, H. Thompson for statistical analysis, and A. Case, B. Keats and M. Havens for discussions and comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The project was conceived of by M.L.H. Experiments were designed and performed by A.J.H., F.M.J., J.J.L., K.E.M., M.L.H. and D.M.D., and were analyzed by M.L.H., J.J.L., F.R., F.M.J., A.J.H. and K.E.M. Animal work was carried out by M.L.H., D.M.D., K.E.M., A.J.H., F.M.J., J.J.L. and M.J.S. Molecular experiments were performed by A.J.H., F.M.J., K.E.M. and M.L.H. J.J.L. carried out the immunofluorescence analysis. J.J.L., M.J.S. and H.E.F. performed auditory brainstem response experiments, and J.J.L. and N.G.B. interpreted the results. M.L.H. and J.J.L. wrote the paper.

Corresponding authors

Correspondence to Jennifer J Lentz or Michelle L Hastings.

Ethics declarations

Competing interests

F.R. may materially benefit financially through stock options in Isis Pharmaceuticals. M.L.H. and F.R. have patents pending with the United States Patent and Trademark Office for the ASOs and the targeting approach.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Table 1 (PDF 3338 kb)

Supplementary Video 1

Ush1c.216AA mice treated with ASO-29. This video shows four 1-month-old mice. Two representative heterozygous (Ush1c.216GA) mice (top row) and homozygous mutant mice (Ush1c.216AA) (bottom row), one of each group treated with ASO-C (left) and one with ASO-29 (right). The ASO-C treated mutant exhibits characteristic circling behavior and hyperactivity (bottom left), whereas the ASO-29-treated mouse (bottom right) is indistinguishable from the heterozygous mice (top). (MOV 2547 kb)

Supplementary Video 2

Behavioral response to acoustic stimuli in Ush1c.216AA mice treated with ASO-29. This video shows four 2-month-old mice. Two representative heterozygous (Ush1c.216GA) mice (top row) and homozygous mutant mice (Ush1c.216AA) (bottom row), one of each group treated with ASO-C (left) and one with ASO-29 (right). Opaque dividers were inserted between the cages to blind the mice to each other. A whistle blow (3-16 kHz and 90-110 dB SPL; Supplementary Fig. 6) was activated after acclimation to the environment. This acoustic stimulus occurs 2.9 s into the video and persists for 0.9 s. A startle response, indicated by a rapid body movement, followed by freezing (a period of watchful immobility), suggests, qualitatively, that the heterozygote mice (GA) and the mutant 216AA mouse treated with ASO-29 could hear the acoustic stimulation. The 216AA mutant mouse treated with ASO-C showed no visible response to the acoustic stimulus. (MOV 2776 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lentz, J., Jodelka, F., Hinrich, A. et al. Rescue of hearing and vestibular function by antisense oligonucleotides in a mouse model of human deafness. Nat Med 19, 345–350 (2013). https://doi.org/10.1038/nm.3106

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3106

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