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:

Gene therapy restores auditory and vestibular function in a mouse model of Usher syndrome type 1c

Abstract

Because there are currently no biological treatments for hearing loss, we sought to advance gene therapy approaches to treat genetic deafness. We focused on Usher syndrome, a devastating genetic disorder that causes blindness, balance disorders and profound deafness, and studied a knock-in mouse model, Ush1c c.216G>A, for Usher syndrome type IC (USH1C). As restoration of complex auditory and balance function is likely to require gene delivery systems that target auditory and vestibular sensory cells with high efficiency, we delivered wild-type Ush1c into the inner ear of Ush1c c.216G>A mice using a synthetic adeno-associated viral vector, Anc80L65, shown to transduce 80–90% of sensory hair cells. We demonstrate recovery of gene and protein expression, restoration of sensory cell function, rescue of complex auditory function and recovery of hearing and balance behavior to near wild-type levels. The data represent unprecedented recovery of inner ear function and suggest that biological therapies to treat deafness may be suitable for translation to humans with genetic inner ear 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: SEM of the organ of Corti in Ush1c c.216G>A mice at P8.
Figure 2: Mechanotransduction in hair cells of Ush1c c.216G>A neonatal mice.
Figure 3: Expression and localization of fluorescently labeled harmonin in tissues exposed to adeno-associated viral vectors in vitro and in vivo.
Figure 4: Recovery of mechanotransduction in hair cells of mice injected with Anc80L65 harmonin vectors.
Figure 5: ABR and DPOAE threshold recovery in mice injected at P1 with AAV2/Anc80L65. CMV.harmonin-b1.
Figure 6: Startle response, rotarod performance and open field behavior recovery in mice injected at P1 with AAV2/Anc80L65. CMV.harmonin-a1 and AAV2/Anc80L65.CMV.harmonin-b1.
Figure 7: SEM of the organ of Corti in mice treated with harmonin-b1 and untreated mice.

Similar content being viewed by others

References

  1. Chien, W.W. et al. Gene therapy restores hair cell stereocilia morphology in inner ears of deaf whirler mice. Mol. Ther. 24, 17–25 (2016).

    Article  CAS  Google Scholar 

  2. Askew, C. et al. Tmc gene therapy restores auditory function in deaf mice. Sci. Transl. Med. 7, 295ra108 (2015).

    Article  Google Scholar 

  3. Akil, O. et al. Restoration of hearing in the VGLUT3 knockout mouse using virally mediated gene therapy. Neuron 75, 283–293 (2012).

    Article  CAS  Google Scholar 

  4. Boughman, J.A., Vernon, M. & Shaver, K.A. Usher syndrome: definition and estimate of prevalence from two high-risk populations. J. Chronic Dis. 36, 595–603 (1983).

    Article  CAS  Google Scholar 

  5. Smith, R.J. et al. Clinical diagnosis of the Usher syndromes. Am. J. Med. Genet. 50, 32–38 (1994).

    Article  CAS  Google Scholar 

  6. Vernon, M. Usher's syndrome--deafness and progressive blindness. Clinical cases, prevention, theory and literature survey. J. Chronic Dis. 22, 133–151 (1969).

    Article  CAS  Google Scholar 

  7. Weil, D. et al. Defective myosin VIIA gene responsible for Usher syndrome type 1B. Nature 374, 60–61 (1995).

    Article  CAS  Google Scholar 

  8. Weston, M.D. et al. Myosin VIIA mutation screening in 189 Usher syndrome type 1 patients. Am. J. Hum. Genet. 59, 1074–1083 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Adato, A. et al. Mutation profile of all 49 exons of the human myosin VIIA gene, and haplotype analysis, in Usher 1B families from diverse origins. Am. J. Hum. Genet. 61, 813–821 (1997).

    Article  CAS  Google Scholar 

  10. Liu, X.Z., Newton, V.E., Steel, K.P. & Brown, S.D. Identification of a new mutation of the myosin VII head region in Usher syndrome type 1. Hum. Mutat. 10, 168–170 (1997).

    Article  CAS  Google Scholar 

  11. 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 

  12. 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 

  13. Bolz, H. et al. Mutation of CDH23, encoding a new member of the cadherin gene family, causes Usher syndrome type 1D. Nat. Genet. 27, 108–112 (2001).

    Article  CAS  Google Scholar 

  14. Bork, J.M. et al. Usher syndrome 1D and nonsyndromic autosomal recessive deafness DFNB12 are caused by allelic mutations of the novel cadherin-like gene CDH23. Am. J. Hum. Genet. 68, 26–37 (2001).

    Article  CAS  Google Scholar 

  15. Ahmed, Z.M. et al. Mutations of the protocadherin gene PCDH15 cause Usher syndrome type 1F. Am. J. Hum. Genet. 69, 25–34 (2001).

    Article  CAS  Google Scholar 

  16. Alagramam, K.N. et al. The mouse Ames waltzer hearing-loss mutant is caused by mutation of Pcdh15, a novel protocadherin gene. Nat. Genet. 27, 99–102 (2001a).

    Article  CAS  Google Scholar 

  17. Alagramam, K.N. et al. Mutations in the novel protocadherin PCDH15 cause Usher syndrome type 1F. Hum. Mol. Genet. 10, 1709–1718 (2001b).

    Article  CAS  Google Scholar 

  18. Weil, D. et al. Usher syndrome type I G (USH1G) is caused by mutations in the gene encoding SANS, a protein that associates with the USH1C protein, harmonin. Hum. Mol. Genet. 12, 463–471 (2003).

    Article  CAS  Google Scholar 

  19. Riazuddin, S. et al. Alterations of the CIB2 calcium- and integrin-binding protein cause Usher syndrome type 1J and nonsyndromic deafness DFNB48. Nat. Genet. 44, 1265–1271 (2012).

    Article  CAS  Google Scholar 

  20. 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 

  21. 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 

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

    Article  CAS  Google Scholar 

  23. 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 

  24. Gregory, F.D. et al. Harmonin inhibits presynaptic Cav1.3 Ca2 channels in mouse inner hair cells. Nat. Neurosci. 14, 1109–1111 (2011).

    Article  CAS  Google Scholar 

  25. Gregory, F.D., Pangrsic, T., Calin-Jageman, I.E., Moser, T. & Lee, A. Harmonin enhances voltage-dependent facilitation of Cav1.3 channels and synchronous exocytosis in mouse inner hair cells. J. Physiol. (Lond.) 591, 3253–3269 (2013).

    Article  CAS  Google Scholar 

  26. 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 

  27. 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 

  28. 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 

  29. Landegger, L. et al. A synthetic AAV vector enables safe and efficient gene transfer to the mammalian inner ear. Nat. Biotechnol. http://dx.doi.org/10.1038/nbt.3781 (2017).

  30. Lentz, J.J. et al. Rescue of hearing and vestibular function by antisense oligonucleotides in a mouse model of human deafness. Nat. Med. 19, 345–350 (2013).

    Article  CAS  Google Scholar 

  31. Gale, J.E., Marcotti, W., Kennedy, H.J., Kros, C.J. & Richardson, G.P. FM1-43 dye behaves as a permeant blocker of the hair-cell mechanotransducer channel. J. Neurosci. 21, 7013–7025 (2001).

    Article  CAS  Google Scholar 

  32. Meyers, J.R. et al. Lighting up the senses: FM1-43 loading of sensory cells through nonselective ion channels. J. Neurosci. 23, 4054–4065 (2003).

    Article  CAS  Google Scholar 

  33. Géléoc, G.S. & Holt, J.R. Developmental acquisition of sensory transduction in hair cells of the mouse inner ear. Nat. Neurosci. 6, 1019–1020 (2003).

    Article  Google Scholar 

  34. Zinn, E. et al. In silico reconstruction of the viral evolutionary lineage yields a potent gene therapy vector. Cell Rep. 12, 1056–1068 (2015).

    Article  CAS  Google Scholar 

  35. Kho, S.T., Pettis, R.M., Mhatre, A.N. & Lalwani, A.K. Safety of adeno-associated virus as cochlear gene transfer vector: analysis of distant spread beyond injected cochleae. Mol. Ther. 4, 368–373 (2000).

    Article  Google Scholar 

  36. Bedrosian, J.C. et al. In vivo delivery of recombinant viruses to the fetal murine cochlea: transduction characteristics and long-term effects on auditory function. Mol. Ther. 14, 328–335 (2006).

    Article  CAS  Google Scholar 

  37. Wang, L., Jiang, H. & Brigande, J.V. Gene transfer to the developing mouse inner ear by in vivo electroporation. J. Vis. Exp. 64, 3653 (2012).

    Google Scholar 

  38. Depreux, F.F. et al. Antisense oligonucleotides delivered to the amniotic cavity in utero modulate gene expression in the postnatal mouse. Nucleic Acids Res. 44, 9519–9529 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Clark, K.R., Sferra, T.J. & Johnson, P.R. Recombinant adeno-associated viral vectors mediate long-term transgene expression in muscle. Hum. Gene Ther. 8, 659–669 (1997).

    Article  CAS  Google Scholar 

  40. Duan, D. et al. Circular intermediates of recombinant adeno-associated virus have defined structural characteristics responsible for long-term episomal persistence in muscle tissue. J. Virol. 72, 8568–8577 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Noben-Trauth, K., Zheng, Q.Y. & Johnson, K.R. Association of cadherin 23 with polygenic inheritance and genetic modification of sensorineural hearing loss. Nat. Genet. 35, 21–23 (2003).

    Article  CAS  Google Scholar 

  42. Kane, K.L. et al. Genetic background effects on age-related hearing loss associated with Cdh23 variants in mice. Hear. Res. 283, 80–88 (2012).

    Article  CAS  Google Scholar 

  43. Lelli, A., Asai, Y., Forge, A., Holt, J.R. & Géléoc, G.S. Tonotopic gradient in the developmental acquisition of sensory transduction in outer hair cells of the mouse cochlea. J. Neurophysiol. 101, 2961–2973 (2009).

    Article  Google Scholar 

  44. Stauffer, E.A. & Holt, J.R. Sensory transduction and adaptation in inner and outer hair cells of the mouse auditory system. J. Neurophysiol. 98, 3360–3369 (2007).

    Article  Google Scholar 

  45. Zheng, L., Zheng, J., Whitlon, D.S., García-Añoveros, J. & Bartles, J.R. Targeting of the hair cell proteins cadherin 23, harmonin, myosin XVa, espin, and prestin in an epithelial cell model. J. Neurosci. 30, 7187–7201 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Manton Center for Orphan Disease Pilot Award 2011 to G.S.G. (Boston Children's Hospital), the Bertarelli Foundation, Program in Translational Neuroscience and Neuroengineering, Kidz-b-Kidz Foundation (now Arts for USH), the Jeff and Kimberly Barber Gene Therapy Research Fund and a consortium agreement under a primary award from the Foundation Fighting Blindness, Cost Center #7 5794 (P.I. L. Vandenberghe). A.A.I. received support from R01DC000304 (DP Corey). S.H.-A. was the recipient of the diversity faculty fellowship award from Harvard Medical School. Behavior and Viral Cores at Boston Children's Hospital are supported by the Boston Children's Hospital Intellectual and Developmental Disabilities Research Center (BCH IDDRC), P30 HD18655. M. Hastings' contribution was supported by R01DC012596. We thank M. Valero (EPL, MEEI) for assistance with ABRs, S. Xu (BCH core) for help with the behavior work, C. Wang (BCH viral core) for AAV production, and C. Nist-Lund (BCH) for technical assistance. L. Zheng and J. Bartles45 (Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL) graciously provided EGFP-tagged, labeled constructs for harmonin-a1 and harmonin-b1 plasmids.

Author information

Authors and Affiliations

Authors

Contributions

The project was conceived and designed by G.S.G. C.A. performed RWM injections with AAV2/1 vectors, electrophysiological recordings and analysis of OHCs, FM1-43 imaging of the organ of Corti; B.P. carried out RWM injections with AAV2/Anc80L65 vectors, electrophysiological recordings and analysis of IHCs mechanotransduction currents. G.S.G. performed electrophysiological recordings of vestibular hair cells. A.G., S.H.-A. and G.S.G. performed ABR experiments. Semi-quantitative radiolabeled PCR and RT-PCR was performed by F.M.J. and M.L.H. ABR and behavioral data analysis was performed by A.G. and G.S.G. Y.A. designed and prepared the plasmid constructs. SEM was performed by J.J.L. (P18), and A.A.I. (P8 and 6 weeks). DIC, immunostaining and confocal imaging was performed by G.S.G. L.H.V. assisted with production of AAV vectors. J.R.H assisted with the design of the experiments, preparation of figures and manuscript. G.S.G. analyzed, interpreted the results and wrote the manuscript.

Corresponding author

Correspondence to Gwenaëlle S Géléoc.

Ethics declarations

Competing interests

A patent #00633-0203P01 on “Materials and methods for delivering nucleic acids to cochlear and vestibular cells” has been deposited by L.H.V., G.S.G. and J.R.H. The Anc80L65 vector is patented by L.H.V., patent #WO/2015/054653 – “Methods of predicting ancestral virus sequence and uses thereof.” L.H.V. is co-founder and SAB member of GenSight Biologics and consultant to various gene therapy companies. L.H.V. receives research support from Selecta Biosciences and Lonza Houston on Anc-AAV development and discovery.

Integrated supplementary information

Supplementary Figure 1 Analysis of hair bundle morphology in Ush1c c.216G>A mice at P18 by SEM.

(a-c) Heterozygous c.216GA mice displayed normal hair bundle morphology at P18. (d-i) Disorganized hair bundles were observed along the organ of P18 Homozygous c.216AA mice. (j-l) IHCs hair bundle were mildly disrupted (arrows) in c.216AA mice. Distance measured from apex tip: Base 3.5-4 mm; Mid 1.8-2.2 mm; Apex 0.6-0.8 mm. Scale bar a-f; j-l: 5 μm; g-i: 1 μm.

Supplementary Figure 2 Mechanotranduction properties in c.216AA mice.

(a-e) Analysis of mechanotransduction in neonatal OHCs from middle and mid-apical turns of the cochlea, P3-P6. Representative current traces from ~Po= 0.5 were fit with a double exponential decay function to assess adaptation in c.216GA and c.216AA mutant (a). Fits were used to generate fast (c) and slow (d) time constants as well as the extent of adaptation (e). The 10-90% operating range was estimated from the second-order Boltzmann fits and was not significantly (NS) altered (b). Extent of adaptation, measured at Popen= 0.5, was significantly less in c.216AA mice than for heterozygous OHCs as shown in this scatter plot (e). (f-j) Analysis of mechanotransduction in neonatal IHCs. 10-90% operating range values were smaller in c.216GA versus c.216AA IHCs (g). Adaptation was always present albeit slightly slower and with a significant lesser extent in c.216AA IHCs (h-j). Statistical analysis is indicated in each plot: *P<0.05, **P<0.01 and ***P<0.001, one-way ANOVA.

Supplementary Figure 3 Expression of fluorescently labeled harmonin-a1 and harmonin-b1 at 6 weeks in c.216AA organ of Corti after P1 dual vector injection.

(a-c) Confocal images of the basal turn in 6 weeks old c.216AA mice after P1 co-injection of AAV2/Anc80L65.CMV.tdTomato::harmonin-a1 (0.5 μl; 4.1 × 1012 gc/ml) and AAV2/Anc80L65.CMV.EGFP::harmonin-b1 (0.5 μl; 3.0 × 1012 gc/ml). 69% and 74% of the total number of cells (IHCs and OHCs) expressed EGFP (a) and tdTomato (c), respectively and 65% expressed both markers demonstrating successful co-transduction. Total hair cells were counted based on the phalloidin image (b, blue). EGFP+ (a) and tdTomato+ (c) hair cells were counted based on the confocal images shown. Scale bar: 20 μm.

Supplementary Figure 4 Analysis of ABR response in 6 weeks old control c.216GA and injected rescued c.216AA mice.

(a,d) Example of ABR responses at 8 and 16 kHz for control c.216GA and rescued c.216AA mice, 90dB stimulus. (b,c, e-f) Average wave 1 amplitude (b,e) and latency (c,f) at 8-11.3 and 16 kHz in 6 weeks old mice with comparable thresholds (n=8 c.216GA, n=5 c.216AA + harmonin-b1 RWM P1). Mean ± S.E. While wave 1 amplitude did not differ significantly between each group (t-test: P>0.1 for all sound levels, b and e), peak latency differed significantly for 8-11.3 kHz responses (t-test: P<0.05 for all sound levels, c) and 16 kHz (t-test: P<0.001, f).

Supplementary Figure 5 USH1C expression in the inner ear of mice injected with AAV2/Anc80L65.CMV.harmonin-b1.

Expression was assessed in six-week old c.216GA and AAV2/Anc80L65.CMV.harmonin-b1 (0.8 μl; 1.9 × 1012 gc/ml) injected and non-injected c.216AA mice (a) Semi-quantitative RT-PCR of correctly spliced (450 bp) and truncated (415 bp) mRNA expression revealed vector-derived expression of correctly spliced Ush1c in injected (“I”) and contralateral ears (“C”) of c.216AA rescued mice #1 and #2 (thresholds ≤ 35 dB response at 11.3 kHz from injected ears). Mouse #3 with poor ABR response (thresholds ≥90 dB at 11.3 kHz) showed modest recovery of correct mRNA expression and mouse #4 (thresholds ≥100 dB at 11.3 kHz) showed none. While the correctly spliced form is not detected in uninjected left (“L”) and right (“R”) ears of c.216AA mice (mice #5, 6), both the correct and truncated splice forms were detected in c.216GA mice (mice #7, 8, 9). Corresponding mouse Gapdh shown in the bottom panel was amplified to confirm a consistent amount of material was used. (b) Semi-quantitative radiolabeled PCR analysis confirmed the presence AAV-mUsh1c DNA in injected and contralateral ears of Ush1c.216AA mice. AAV-Ush1c DNA was present but reduced in mice #3 and #4. (c) The relative amount of viral DNA correlates with ABR thresholds. The 11.3 and 16 kHz responses from injected and contralateral cochleas of the four injected mice (#1-4) are illustrated. Linear regressions show high correlation between the amount of viral DNA and ABR threshold responses. Tissues were collected at 6 weeks of age. (d) Immunostaining of harmonin confirmed restored expression of harmonin protein in stereocilia of c.216AA mice that recovered hearing following Anc80L65.CMV.harmonin-b1 P1 RWM injection. Panels show immunofluorescence of harmonin (green) and F-actin (red) in outer hair cell (OHC) bundles in mid apical regions of wild type (WT, 6 weeks old), homozygous c.216AA (4 weeks), injected c.216AA mice with no rescue (4 weeks, no ABR response at 110 dB at any frequency tested), and injected c.216AA mice with rescue (4 weeks, 30 dB at 11 kHz). Scale bar: 10 μm.

Supplementary Figure 6 Long term ABR threshold recovery correlates with OHCs survival in the mid to apical region of the auditory organ.

Hair cell count across the entire Organ of Corti was performed post-mortem in left ears of three uninjected c.216AA and five injected c.216AA (P1 RWM injection, 0.8 μl AAV2/Anc80L65.CMV.harmonin-b1) at 6 months of age. Insert: While two of the mice (#1 and #2) showed poor ABR response (PR) thresholds across the entire range tested (≥95 dB), three (#3-5) responded (R) with thresholds ranging 35 and 55 dB for sound stimuli between 5.6 and 16 kHz. The total number of IHC and OHCs hair cells was increased in injected mice. Comparison of rescued injected mice with those injected that had poor rescue shows that the number of IHCs was not different but a significant number of OHCs were noted in the rescued mice. Analysis across the entire length of the organ showed the difference can be accounted for as an increase in hair cell survival from the mid to apical regions of the organ.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 1221 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pan, B., Askew, C., Galvin, A. et al. Gene therapy restores auditory and vestibular function in a mouse model of Usher syndrome type 1c. Nat Biotechnol 35, 264–272 (2017). https://doi.org/10.1038/nbt.3801

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3801

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