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.

  • Review Article
  • Published:

Bioengineering strategies for restoring vision

Abstract

Late-stage retinal degenerative disease involving photoreceptor loss can be treated by optogenetic therapy, cell transplantation and retinal prostheses. These approaches aim to restore light sensitivity to the retina as well as visual perception by integrating neuronal responses for transmission to the cortex. In age-related macular degeneration, some cell-based therapies also aim to restore photoreceptor-supporting tissue to prevent complete photoreceptor loss. In the earlier stages of degeneration, gene-replacement therapy could attenuate retinal-disease progression and reverse loss of function. And gene-editing strategies aim to correct the underlying genetic defects. In this Review, we highlight the most promising gene therapies, cell therapies and retinal prostheses for the treatment of retinal disease, discuss the benefits and drawbacks of each treatment strategy and the factors influencing whether functional tissue is reconstructed and repaired or replaced with an electronic device, and summarize upcoming technologies for enhancing the restoration of vision.

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

Fig. 1: Treatments in vision restoration according to retinal-degeneration stage, and structural remodelling in typical rod–cone dystrophy.
Fig. 2: Normal retinal phenotype and three different types of retinal degeneration.
Fig. 3: Delivery of human rhodopsin in rd1 mice via an AAV vector using a ubiquitous chicken-β-actin promoter.
Fig. 4: Subretinal transplantation of Nrl–GFP donor cells into rd1–DsRed mice with advanced photoreceptor-layer atrophy.
Fig. 5: The visual pathway and locations of different retinal implants147.
Fig. 6: Electronic subretinal implant.

Similar content being viewed by others

References

  1. Sohocki, M. M. et al. Prevalence of mutations causing retinitis pigmentosa and other inherited retinopathies. Hum. Mutat. 17, 42–51 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Liew, G., Michaelides, M. & Bunce, C. A comparison of the causes of blindness certifications in England and Wales in working age adults (16–64 years), 1999–2000 with 2009–2010. BMJ Open. 4, e004015 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Wong, W. L. et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob. Health 2, e106–e116 (2014).

    Article  PubMed  Google Scholar 

  4. Russell, S. et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet 390, 849–860 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Xue, K. et al. Beneficial effects on vision in patients undergoing retinal gene therapy for choroideremia. Nat. Med. 24, 1507–1512 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cukras, C. et al. Retinal AAV8-RS1 gene therapy for X-linked retinoschisis: initial findings from a phase I/IIa trial by intravitreal delivery. Mol. Ther. 26, 2282–2294 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cehajic-Kapetanovic, J. et al. Initial results from a first-in-human gene therapy trial on X-linked retinitis pigmentosa caused by mutations in RPGR. Nat. Med. 26, 354–359 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kantor, A. et al. CRISPR genome engineering for retinal diseases. Prog. Mol. Biol. Transl. Sci. 182, 29–79 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Quinn, J. et al. Genome-editing strategies for treating human retinal degenerations. Hum. Gene Ther. 32, 247–259 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Maeder, M. L. et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat. Med. 25, 229–233 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Jonas, J. B. et al. Glaucoma. Lancet 390, 2183–2193 (2017).

    Article  PubMed  Google Scholar 

  12. Guy, J. et al. Gene therapy for Leber hereditary optic neuropathy: low- and medium-dose visual results. Ophthalmology 124, 1621–1634 (2017).

    Article  PubMed  Google Scholar 

  13. Vignal, C. et al. Safety of rAAV2/2-ND4 gene therapy for Leber hereditary optic neuropathy. Ophthalmology 125, 945–947 (2018).

    Article  PubMed  Google Scholar 

  14. Wu, S., Chang, K. C., Nahmou, M. & Goldberg, J. L. Induced pluripotent stem cells promote retinal ganglion cell survival after transplant. Invest. Ophthalmol. Vis. Sci. 59, 1571–1576 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhang, X. et al. Cell transplantation of retinal ganglion cells derived from hESCs. Restor. Neurol. Neurosci. 38, 131–140 (2020).

    PubMed  Google Scholar 

  16. Suen, H. C. et al. Transplantation of retinal ganglion cells derived from male germline stem cell as a potential treatment to glaucoma. Stem Cells Dev. 28, 1365–1375 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Jones, B. W. et al. Retinal remodeling triggered by photoreceptor degenerations. J. Comp. Neurol. 464, 1–16 (2003).

    Article  PubMed  Google Scholar 

  18. Marc, R. E. & Jones, B. W. Retinal remodeling in inherited photoreceptor degenerations. Mol. Neurobiol. 28, 139–147 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Marc, R. E., Jones, B. W., Watt, C. B. & Strettoi, E. Neural remodeling in retinal degeneration. Prog. Retin. Eye Res. 22, 607–655 (2003).

    Article  PubMed  Google Scholar 

  20. Jones, B. W. & Marc, R. E. Retinal remodeling during retinal degeneration. Exp. Eye Res. 81, 123–137 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Marc, R. E. et al. Neural reprogramming in retinal degeneration. Invest. Ophthalmol. Vis. Sci. 48, 3364–3371 (2007).

    Article  PubMed  Google Scholar 

  22. Cuenca, N. et al. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog. Retin. Eye Res. 43, 17–75 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Jones, B. W. et al. Retinal remodeling in human retinitis pigmentosa. Exp. Eye Res. 150, 149–165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Krishnamoorthy, V. et al. Retinal remodeling: concerns, emerging remedies and future prospects. Front. Cell Neurosci. 10, 38 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Pfeiffer, R. L., Marc, R. E. & Jones, B. W. Persistent remodeling and neurodegeneration in late-stage retinal degeneration. Prog. Retin. Eye Res. 74, 100771 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Eleftheriou, C. G. et al. Meclofenamic acid improves the signal to noise ratio for visual responses produced by ectopic expression of human rod opsin. Mol. Vis. 23, 334–345 (2017).

    PubMed  PubMed Central  Google Scholar 

  27. Telias, M. et al. Retinoic acid induces hyperactivity, and blocking its receptor unmasks light responses and augments vision in retinal degeneration. Neuron 102, 574–586.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Acland, G. M. et al. Gene therapy restores vision in a canine model of childhood blindness. Nat. Genet. 28, 92–95 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Bainbridge, J. W. B. et al. Effect of gene therapy on visual function in Leber’s congenital amaurosis. N. Engl. J. Med. 358, 2231–2239 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Maguire, A. M. et al. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N. Engl. J. Med. 358, 2240–2248 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cideciyan, A. V. et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc. Natl Acad. Sci. USA 105, 15112–15117 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Fischer, M. D. et al. Safety and vision outcomes of subretinal gene therapy targeting cone photoreceptors in achromatopsia: a nonrandomized controlled trial. JAMA Ophthalmol. 138, 643–651 (2020).

    Article  PubMed  Google Scholar 

  33. Josan, A. S. et al. Microperimetry Hill of vision and volumetric measures of retinal sensitivity. Transl. Vis. Sci. Technol. 10, 12 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  34. MacDonald, I. M. et al. Perspectives on gene therapy: choroideremia represents a challenging model for the treatment of other inherited retinal degenerations. Trans. Vis. Sci. Tech. 9, 17 (2020).

    Article  Google Scholar 

  35. McClements, M. E. et al. An AAV dual vector strategy ameliorates the Stargardt phenotype in adult Abca4-/- mice. Hum. Gene Ther. 30, 590–600 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hirsch, M. L., Wolf, S. J. & Samulski, R. J. Delivering transgenic DNA exceeding the carrying capacity of AAV vectors. Methods Mol. Biol. 1382, 21–39 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Trapani, I. et al. Improved dual AAV vectors with reduced expression of truncated proteins are safe and effective in the retina of a mouse model of Stargardt disease. Hum. Mol. Genet. 24, 6811–6825 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. T Trapani, I. et al. Effective delivery of large genes to the retina by dual AAV vectors. EMBO Mol. Med. 6, 194–211 (2014).

    Article  Google Scholar 

  39. Yi-Ting, T. et al. Clustered regularly interspaced short palindromic repeats-based genome surgery for the treatment of autosomal dominant retinitis pigmentosa. Ophthalmology 125, 1421–1430 (2018).

    Article  Google Scholar 

  40. Diakatou, M., Manes, G., Bocquet, B., Meunier, I. & Kalatzis, V. Genome editing as a treatment for the most prevalent causative genes of autosomal dominant retinitis pigmentosa. Int. J. Mol. Sci. 20, 2542 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C. & Doudna, J. A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 507, 62–67 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sander, J. D. & Joung, J. K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347–355 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cai, Y. et al. In vivo genome editing rescues photoreceptor degeneration via a Cas9/RecA-mediated homology-directed repair pathway. Sci. Adv. 5, eaav3335 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bakondi, B. et al. In vivo CRISPR/Cas9 Gene editing corrects retinal dystrophy in the S334ter-3 rat model of autosomal dominant retinitis pigmentosa. Mol. Ther. 24, 556–563 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tsai, Y. et al. CRISPR-based genome surgery for the treatment of autosomal dominant retinitis. Ophthalmology 125, 1421–1430 (2018).

    Article  PubMed  Google Scholar 

  46. McClements, M. E., Staurenghi, F., MacLaren, R. E. & Cehajic-Kapetanovic, J. Optogenetic gene therapy for the degenerate retina: recent advances. Front. Neurosci. 11, 570909 (2020).

    Article  Google Scholar 

  47. Bi, A. et al. Ectopic expression of a microbial-type rhodopsin restores visual responses in mice with photoreceptor degeneration. Neuron 50, 23–33 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang, Y., Ivanova, E., Bi, A. & Pan, Z. H. Ectopic expression of multiple microbial rhodopsins restores ON and OFF light responses in retinas with photoreceptor degeneration. J. Neurosci. 29, 9186–9196 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Thyagarajan, S. et al. Visual function in mice with photoreceptor degeneration and transgenic expression of channelrhodopsin 2 in ganglion cells. J. Neurosci. 30, 8745–8758 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Doroudchi, M. M. et al. Virally delivered channelrhodopsin-2 safely and effectively restores visual function in multiple mouse models of blindness. Mol. Ther. 19, 1220–1229 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sengupta, A. et al. Red-shifted channelrhodopsin stimulation restores light responses in blind mice, macaque retina, and human retina. EMBO Mol. Med. 8, 1248–1264 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lagali, P. S. et al. Light-activated channels targeted to ON bipolar cells restore visual function in retinal degeneration. Nat. Neurosci. 11, 667–675 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Macé, E. et al. Targeting channelrhodopsin-2 to ON-bipolar cells with vitreally administered AAV restores ON and OFF visual responses in blind mice. Mol. Ther. 23, 7–16 (2015).

    Article  PubMed  Google Scholar 

  54. Busskamp, V. et al. Genetic rectivation of cone photoreceptors restores visual responses in retinitis pigmentosa. Science 329, 413–417 (2010).

    Article  CAS  PubMed  Google Scholar 

  55. Gauvain, G. et al. Optogenetic therapy: high spatiotemporal resolution and pattern discrimination compatible with vision restoration in non-human primates. Commun. Biol. 4, 125 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sahel, J. A. et al. Partial recovery of visual function in a blind patient after optogenetic therapy. Nat. Med. 27, 1223–1229 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Cehajic-Kapetanovic, J. et al. Restoration of vision with ectopic expression of human rod opsin. Curr. Biol. 25, 2111–2122 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Gaub, B. M., Berry, M. H., Holt, A. E., Isacoff, E. Y. & Flannery, J. G. Optogenetic vision restoration using rhodopsin for enhanced sensitivity. Mol. Ther. 23, 1562–1571 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lin, B., Koizumi, A., Tanaka, N., Panda, S. & Masland, R. H. Restoration of visual function in retinal degeneration mice by ectopic expression of melanopsin. Proc. Natl Acad. Sci. USA 105, 16009–16014 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. De Silva, S. R. et al. Long-term restoration of visual function in end-stage retinal degeneration using subretinal human melanopsin gene therapy. Proc. Natl Acad. Sci. USA 114, 11211–11216 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  61. van Wyk, M., Pielecka-Fortuna, J., Löwel, S. & Kleinlogel, S. Restoring the ON switch in blind retinas: opto-mGluR6, a next-generation, cell-tailored optogenetic tool. PLoS Biol. 13, e1002143 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Berry, M. H. et al. Restoration of high-sensitivity and adapting vision with a cone opsin. Nat. Commun. 10, 1221 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Gaub, B. M. et al. Restoration of visual function by expression of a light-gated mammalian ion channel in retinal ganglion cells or ON-bipolar cells. Proc. Natl Acad. Sci. USA 111, E5574–E5583 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Berry, M. H. et al. Restoration of patterned vision with an engineered photoactivatable G protein-coupled receptor. Nat. Commun. 8, 1862 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Tochitsky, I. et al. Restoring visual function to blind mice with a photoswitch that exploits electrophysiological remodeling of retinal ganglion cells. Neuron 81, 800–813 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Tochitsky, I., Trautman, J., Gallerani, N., Malis, J. G. & Kramer, R. H. Restoring visual function to the blind retina with a potent, safe and long-lasting photoswitch. Sci. Rep. 7, 45487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cehajic-Kapetanovic, J. et al. Glycosidic enzymes enhance retinal transduction following intravitreal delivery of AAV2. Mol. Vis. 17, 1771–1783 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Cehajic-Kapetanovic, J., Milosavljevic, N., Bedford, R. A., Lucas, R. J. & Bishop, P. N. Efficacy and safety of glycosidic enzymes for improved gene delivery to the retina following intravitreal injection in mice. Mol. Ther. Methods Clin. Dev. 9, 192–202 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Fick, A. ‘On liquid diffusion’. Ann. Phys. Chem. 94, 59 (1855).

    Article  Google Scholar 

  70. Dalkara, D. et al. In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. Sci. Transl. Med. 5, 189ra76 (2013).

    Article  PubMed  Google Scholar 

  71. Kotterman, M. A. et al. Antibody neutralization poses a barrier to intravitreal adeno-associated viral vector gene delivery to non-human primates. Gene Ther. 22, 116–126 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Cehajic-Kapetanovic, J. et al. Highest reported visual acuity after electronic retinal implantation. Acta Ophthalmol. 98, 736–740 (2020).

    Article  PubMed  Google Scholar 

  73. McClements, M. E. et al. AAV induced expression of human rod and cone opsin in bipolar cells of a mouse model of retinal degeneration, BioMed. Res. Int. https://doi.org/10.1155/2021/4014797 (2021).

  74. Thanos, C. G. et al. Sustained secretion of ciliary neurotrophic factor to the vitreous, using the encapsulated cell therapy-based NT-501 intraocular device. Tissue Eng. 10, 1617–1622 (2004).

    Article  CAS  PubMed  Google Scholar 

  75. Lund, R. D. et al. Cells isolated from umbilical cord tissue rescue photoreceptors and visual functions in a rodent model of retinal disease. Stem Cells 25, 602–611 (2007).

    Article  CAS  PubMed  Google Scholar 

  76. Ho, A. C. et al. Experience with a subretinal cell-based therapy in patients with geographic atrophy secondary to age-related macular degeneration. Am. J. Ophthalmol. 179, 67–80 (2017).

    Article  PubMed  Google Scholar 

  77. Park, S. S. Cell therapy applications for retinal vascular diseases: diabetic retinopathy and retinal vein occlusion. Invest. Ophthalmol. Vis. Sci. 57, ORSFj1–ORSFj10 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Park, S. S. et al. Intravitreal autologous bone marrow CD34+ cell therapy for ischemic and degenerative retinal disorders: preliminary phase 1 clinical trial findings. Invest. Ophthalmol. Vis. Sci. 56, 81–89 (2014).

    Article  PubMed  Google Scholar 

  79. Otani, A. et al. Rescue of retinal degeneration by intravitreally injected adult bone marrow-derived lineage-negative hematopoietic stem cells. J. Clin. Invest. 114, 765–774 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Koh, S. et al. Subretinal human umbilical tissue-derived cell transplantation preserves retinal synaptic connectivity and attenuates Müller glial reactivity. J. Neurosci. 38, 2923–2943 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Koh, S. et al. Human umbilical tissue-derived cells promote synapse formation and neurite outgrowth via thrombospondin family proteins. J. Neurosci. 35, 15649–15665 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Börger, V. et al. Mesenchymal stem/stromal cell-derived extracellular vesicles and their potential as novel immunomodulatory therapeutic agents. Int. J. Mol. Sci. 18, 1450 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Zhou, J. et al. Retinal progenitor cells release extracellular vesicles containing developmental transcription factors, microRNA and membrane proteins. Sci. Rep. 8, 2823 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Knickelbein, J. E. et al. Modulation of immune responses by extracellular vesicles from retinal pigment epithelium. Invest. Ophthalmol. Vis. Sci. 57, 4101–4107 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kashani, A. H. et al. A bioengineered retinal pigment epithelial monolayer for advanced, dry age-related macular degeneration. Sci. Transl. Med. 10, eaao4097 (2018).

    Article  PubMed  Google Scholar 

  86. Mandai, M. et al. Autologous induced stem-cell-derived retinal cells for macular degeneration. N. Engl. J. Med. 376, 1038–1046 (2017).

    Article  CAS  PubMed  Google Scholar 

  87. da Cruz, L. et al. Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat. Biotechnol. 36, 328–337 (2018).

    Article  PubMed  Google Scholar 

  88. Schwartz, S. D. et al. Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet 379, 713–720 (2012).

    Article  CAS  PubMed  Google Scholar 

  89. Schwartz, S. D., Tan, G., Hosseini, H. & Nagiel, A. Subretinal transplantation of embryonic stem cell–derived retinal pigment epithelium for the treatment of macular degeneration: an assessment at 4 years. Invest. Ophthalmol. Vis. Sci. 57, ORSFc1–ORSFc9 (2016).

    Article  CAS  PubMed  Google Scholar 

  90. Sharma, R. et al. Clinical-grade stem cell-derived retinal pigment epithelium patch rescues retinal degeneration in rodents and pigs. Sci. Transl. Med. 11, eaat5580 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Ben M’Barek, K. et al. Human ESC–derived retinal epithelial cell sheets potentiate rescue of photoreceptor cell loss in rats with retinal degeneration. Sci. Transl. Med. 9, eaai7471 (2019).

    Article  Google Scholar 

  92. Kuppermann, B. D., Boyer, D. S., Mills, B., Yang, J. & Klassen, H. J. Safety and activity of a single, intravitreal injection of human retinal progenitor cells (jCell) for treatment of retinitis pigmentosa (RP). Invest. Ophthalmol. Vis. Sci. 59, 2987 (2018).

    Google Scholar 

  93. Yang, J. et al. Translational development of human retinal progenitor cells for treatment of retinitis pigmentosa. Invest. Ophthalmol. Vis. Sci. 54, 2237 (2013).

    Google Scholar 

  94. Warfvinge, K. et al. Xenotransplantation of human neural progenitor cells to the subretinal space of nonimmunosuppressed pigs. J. Transplant. 2011, 948740 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Aftab, U. et al. Growth kinetics and transplantation of human retinal progenitor cells. Exp. Eye Res. 89, 301–310 (2009).

    Article  CAS  PubMed  Google Scholar 

  96. Redenti, S. et al. Retinal tissue engineering using mouse retinal progenitor cells and a novel biodegradable, thin-film poly(e-caprolactone) nanowire scaffold. J. Ocul. Biol. Dis. Infor. 1, 19–29 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Singh, M. S. et al. Reversal of end-stage retinal degeneration and restoration of visual function by photoreceptor transplantation. Proc. Natl Acad. Sci. USA 110, 1101–1106 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Garita-Hernandez, M. et al. Restoration of visual function by transplantation of optogenetically engineered photoreceptors. Nat. Commun. 10, 4524 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Gust, J. & Reh, T. A. Adult donor rod photoreceptors integrate into the mature mouse retina. Invest. Ophthalmol. Vis. Sci. 52, 5266–5272 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Singh, M. S. et al. Transplanted photoreceptor precursors transfer proteins to host photoreceptors by a mechanism of cytoplasmic fusion. Nat. Commun. 7, 13537 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Santos-Ferreira, T. et al. Retinal transplantation of photoreceptors results in donor-host cytoplasmic exchange. Nat. Commun. 7, 13028 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Pearson, R. A. et al. Donor and host photoreceptors engage in material transfer following transplantation of post-mitotic photoreceptor precursors. Nat. Commun. 7, 13029 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Ortin-Martinez, A. et al. A reinterpretation of cell transplantation: GFP transfer from donor to host photoreceptors. Stem Cells 35, 932–939 (2017).

    Article  CAS  PubMed  Google Scholar 

  104. Garweg, J. G., Tappeiner, C. & Halberstadt, M. Pathophysiology of proliferative vitreoretinopathy in retinal detachment. Surv. Ophthalmol. 58, 321–329 (2013).

    Article  PubMed  Google Scholar 

  105. Li, L. & Turner, J. E. Transplantation of retinal pigment epithelial cells to immature and adult rat hosts: short- and long-term survival characteristics. Exp. Eye Res. 47, 771–785 (1988).

    Article  CAS  PubMed  Google Scholar 

  106. Royo, P. E. & Quay, W. B. Retinal transplantation from fetal to maternal mammalian eye. Growth 23, 313–336 (1959).

    CAS  PubMed  Google Scholar 

  107. Kador, K. E. et al. Retinal ganglion cell polarization using immobilized guidance cues on a tissue-engineered scaffold. Acta Biomater. 10, 4939–4946 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Gandhi, J. K. et al. Fibrin hydrogels are safe, degradable scaffolds for sub-retinal implantation. PLoS ONE 15, e0227641 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Ott, H. C. et al. Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nat. Med. 14, 213–221 (2008).

    Article  CAS  PubMed  Google Scholar 

  110. Karczewski, M. & Malkiewicz, T. Scaffolds from surgically removed kidneys as a potential source of organ transplantation. Biomed. Res. Int. 2015, 325029 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Kundu, J. et al. Decellularized retinal matrix: natural platforms for human retinal progenitor cell culture. Acta Biomater. 31, 61–70 (2016).

  112. García Delgado, A. B. et al. Subretinal transplant of induced pluripotent stem cell-derived retinal pigment epithelium on nanostructured fibrin-agarose. Tissue Eng. Part A 25, 799–808 (2019).

    Article  PubMed  Google Scholar 

  113. Krishna, Y. et al. Expanded polytetrafluoroethylene as a substrate for retinal pigment epithelial cell growth and transplantation in age-related macular degeneration. Br. J. Ophthalmol. 95, 569–573 (2011).

    Article  CAS  PubMed  Google Scholar 

  114. Chedly, J. et al. Physical chitosan microhydrogels as scaffolds for spinal cord injury restoration and axon regeneration. Biomaterials 138, 91–107 (2017).

    Article  CAS  PubMed  Google Scholar 

  115. Caron, I. et al. A new three-dimensional biomimetic hydrogel to deliver factors secreted by human mesenchymal stem cells in spinal cord injury. Biomaterials 75, 135–147 (2016).

    Article  CAS  PubMed  Google Scholar 

  116. Sakiyama-Elbert, S., Johnson, P. J., Hodgetts, S. I., Plant, G. W. & Harvey, A. R. Scaffolds to promote spinal cord regeneration. Handb. Clin. Neurol. 109, 575–594 (2012).

    Article  CAS  PubMed  Google Scholar 

  117. Hong, L. T. A. et al. An injectable hydrogel enhances tissue repair after spinal cord injury by promoting extracellular matrix remodeling. Nat. Commun. 8, 533 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Masaeli, E. et al. Tissue engineering of retina through high resolution 3-dimensional inkjet bioprinting. Biofabrication 12, 025006 (2020).

    Article  CAS  PubMed  Google Scholar 

  119. Singh, D. et al. A biodegradable scaffold enhances differentiation of embryonic stem cells into a thick sheet of retinal cells. Biomaterials 154, 158–168 (2018).

    Article  CAS  PubMed  Google Scholar 

  120. Akiba, R. et al. Quantitative and qualitative evaluation of photoreceptor synapses in developing, degenerating and regenerating retinas. Front. Cell Neurosci. 13, 16 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Omori, Y. et al. Presynaptic dystroglycan-pikachurin complex regulates the proper synaptic connection between retinal photoreceptor and bipolar cells. J. Neurosci. 32, 6126–6137 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Nandrot, E. F., Chang, Y. & Finnemann, S. C. Alphavbeta5 integrin receptors at the apical surface of the RPE: one receptor, two functions. Adv. Exp. Med. Biol. 613, 369–375 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Dorgau, B. et al. Decellularised extracellular matrix-derived peptides from neural retina and retinal pigment epithelium enhance the expression of synaptic markers and light responsiveness of human pluripotent stem cell derived retinal organoids. Biomaterials 199, 63–75 (2019).

    Article  CAS  PubMed  Google Scholar 

  124. Jung, Y. H. et al. 3D microstructured scaffolds to support photoreceptor polarization and maturation. Adv. Mater. 30, e1803550 (2018).

    Article  PubMed  Google Scholar 

  125. Bernardos, R. L., Barthel, L. K., Meyers, J. R. & Raymond, P. A. Late-stage neuronal progenitors in the retina are radial Müller glia that function as retinal stem cells. J. Neurosci. 27, 7028–7040 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Thummel, R., Kassen, S. C., Montgomery, J. E., Enright, J. M. & Hyde, D. R. Inhibition of Müller glial cell division blocks regeneration of the light-damaged zebrafish retina. Dev. Neurobiol. 68, 392–408 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Johns, P. R. & Fernald, R. D. Genesis of rods in teleost fish retina. Nature 293, 141–142 (1981).

    Article  CAS  PubMed  Google Scholar 

  128. Fischer, A. J. & Reh, T. A. Müller glia are a potential source of neural regeneration in the postnatal chicken retina. Nat. Neurosci. 4, 247–252 (2001).

    Article  CAS  PubMed  Google Scholar 

  129. Garber, K. RIKEN suspends first clinical trial involving induced pluripotent stem cells. Nat. Biotech. 33, 890–891 (2015).

    Article  CAS  Google Scholar 

  130. Spencer, R., Fisher, S., Lewis, G. P. & Malone, T. Epiretinal membrane in a subject after transvitreal delivery of palucorcel (CNTO 2476). Clin. Ophthalmol. 11, 1797–1803 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Oishi, A. et al. Retinal nerve fiber layer thickness in patients with retinitis pigmentosa. Eye 23, 561–566 (2009).

    Article  CAS  PubMed  Google Scholar 

  132. Hood, D. C. et al. Thickness of receptor and post-receptor retinal layers in patients with retinitis pigmentosa measured with frequency-domain optical coherence tomography. Invest. Ophthalmol. Vis. Sci. 50, 2328–2336 (2009).

    Article  PubMed  Google Scholar 

  133. Punzo, C. & Cepko, C. Cellular responses to photoreceptor death in the rd1 mouse model of retinal degeneration. Invest. Ophthalmol. Vis. Sci. 48, 849–857 (2007).

    Article  PubMed  Google Scholar 

  134. MacLaren, R. E. Development and role of retinal glia in regeneration of ganglion cells following retinal injury. Br. J. Ophthalmol. 80, 458–464 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Chua, J. et al. Early remodeling of Müller cells in the rd/rd mouse model of retinal dystrophy. J. Comp. Neurol. 521, 2439–2453 (2013).

    Article  CAS  PubMed  Google Scholar 

  136. Kinouchi, R. et al. Robust neural integration from retinal transplants in mice deficient in GFAP and vimentin. Nat. Neurosci. 6, 863–868 (2003).

    Article  CAS  PubMed  Google Scholar 

  137. Lee, W. et al. The external limiting membrane in early-onset Stargardt disease. Invest. Ophthalmol. Vis. Sci. 55, 6139–6149 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Pearson, R. A. et al. Targeted disruption of outer limiting membrane junctional proteins (Crb1 and ZO-1) increases integration of transplanted photoreceptor precursors into the adult wild-type and degenerating retina. Cell Transplant. 19, 487–503 (2010).

    Article  CAS  PubMed  Google Scholar 

  139. Lao, L. L., Peppas, N. A., Boey, F. Y. & Venkatraman, S. S. Modeling of drug release from bulk-degrading polymers. Int. J. Pharm. 418, 28–41 (2011).

    Article  CAS  PubMed  Google Scholar 

  140. Singh, R. K., Kolandaivelu, S. & Ramamurthy, V. Early alteration of retinal neurons in Aipl1−/− animals. Invest. Ophthalmol. Vis. Sci. 55, 3081–3092 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Tansley, K. The development of the rat eye in graft. J. Exp. Biol. 22, 221–223 (1946).

    Article  CAS  PubMed  Google Scholar 

  142. Humayun, M. S. et al. Human neural retinal transplantation. Invest. Ophthalmol. Vis. Sci. 41, 3100–3106 (2000).

    CAS  PubMed  Google Scholar 

  143. Tomita, M. et al. Bone marrow-derived stem cells can differentiate into retinal cells in injured rat retina. Stem Cells 20, 279–283 (2002).

    Article  CAS  PubMed  Google Scholar 

  144. MacLaren, R. E. et al. Retinal repair by transplantation of photoreceptor precursors. Nature 444, 203–207 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Lamba, D. A., Gust, J. & Reh, T. A. Transplantation of human embryonic stem cell derived photoreceptors restores some visual function in Crx-deficient mice. Cell Stem Cell 4, 73–79 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Lamba, D. A. et al. Generation, purification and transplantation of photoreceptors derived from human induced pluripotent stem cells. PLoS ONE 5, e8763 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Eiraku, M. et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 472, 51–56 (2011).

    Article  CAS  PubMed  Google Scholar 

  148. Pearson, R. A. et al. Restoration of vision after transplantation of photoreceptors. Nature 485, 99–103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Howden, S. E. et al. Genetic correction and analysis of induced pluripotent stem cells from a patient with gyrate atrophy. Proc. Natl Acad. Sci. USA 108, 6537–6542 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Barnea-Cramer, A. O. et al. Repair of retinal degeneration following ex vivo minicircle DNA gene therapy and transplantation of corrected photoreceptor progenitors. Mol. Ther. 28, 830–844 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Naldini, L. Ex vivo gene transfer and correction for cell-based therapies. Nat. Rev. Genet. 12, 301–315 (2011).

    Article  CAS  PubMed  Google Scholar 

  152. Chow, A. Y. The artificial silicon retina microchip for the treatment of vision loss from retinitis pigmentosa. Arch. Ophthalmol. 122, 460–469 (2004).

    Article  PubMed  Google Scholar 

  153. Guenther, T., Lovell, N. H. & Suaning, G. J. Bionic vision: system architectures: a review. Expert Rev. Med. Devices 9, 33–48 (2012).

    Article  PubMed  Google Scholar 

  154. Garg, S. J. & Federman, J. Optogenetics, visual prosthesis and electrostimulation for retinal dystrophies. Curr. Opin. Ophthalmol. 24, 407–414 (2013).

    Article  PubMed  Google Scholar 

  155. Luo, Y. H. & da Cruz, L. A review and update on the current status of retinal prostheses (bionic eye). Br. Med. Bull. 109, 31–44 (2014).

    Article  PubMed  Google Scholar 

  156. Zrenner, E. Fighting blindness with microelectronics. Sci. Transl. Med. 5, 210ps16 (2013).

    Article  PubMed  Google Scholar 

  157. Humayun, M. S. et al. Visual perception in a blind subject with a chronic microelectronic retinal prosthesis. Vis. Res. 43, 2573–2581 (2003).

    Article  PubMed  Google Scholar 

  158. Yanai, D. et al. Visual performance using a retinal prosthesis in three subjects with retinitis pigmentosa. Am. J. Ophthalmol. 143, 820–827 (2007).

    Article  PubMed  Google Scholar 

  159. Rizzo, J. F. III, Wyatt, J., Loewenstein, J., Kelly, S. & Shire, D. Perceptual efficacy of electrical stimulation of human retina with a microelectrode array during short-term surgical trials. Invest. Ophthalmol. Vis. Sci. 44, 5362–5369 (2003).

    Article  PubMed  Google Scholar 

  160. Humayun, M. S. et al. Preliminary 6 month results from the Argus II epiretinal prosthesis feasibility study. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 4566–4568 (2009).

  161. Ho, A. C. et al. Argus II Study Group. LONG-term results from an epiretinal prosthesis to restore sight to the blind. Ophthalmology 122, 1547–1554 (2015).

    Article  PubMed  Google Scholar 

  162. Humayun, M. S. et al. Argus II Study Group. Interim results from the international trial of Second Sight’s visual prosthesis. Ophthalmology 119, 779–788 (2012).

  163. Richard, G. et al. Multicenter study on acute electrical stimulation of the human retina with an epiretinal implant: clinical results in 20 patients. Invest. Ophthalmol. Vis. Sci. 46, 1143 (2005).

    Google Scholar 

  164. Keserü, M. et al. Acute electrical stimulation of the human retina with an epiretinal electrode array. Acta Ophthalmol. 90, e1–e8 (2012).

    Article  PubMed  Google Scholar 

  165. da Cruz, L. et al. Five-year safety and performance results from the Argus II retinal prosthesis system clinical trial. Argus II Study Group. Ophthalmology 123, 2248–2254 (2016).

    Article  PubMed  Google Scholar 

  166. Zrenner, E. et al. Subretinal electronic chips allow blind patients to read letters and combine them to words. Proc. Biol. Sci. 278, 1489–1497 (2011).

    PubMed  Google Scholar 

  167. Roessler, G. et al. Implantation and explantation of a wireless epiretinal retina implant device in blind RP patients. Invest. Ophthalmol. Vis. Sci. 50, 3003–3008 (2009).

    Article  PubMed  Google Scholar 

  168. Stingl, K. et al. Subretinal visual implant Alpha IMS–clinical trial interim report. Vis. Res. 111, 149–160 (2015).

    Article  PubMed  Google Scholar 

  169. Stingl, K. et al. Artificial vision with wirelessly powered subretinal electronic implant alpha-IMS. Proc. Biol. Sci. 280, 20130077 (2013).

    PubMed  PubMed Central  Google Scholar 

  170. Stingl, K. et al. Interim results of a multicenter trial with the new electronic subretinal implant Alpha AMS in 15 patients blind from inherited retinal degenerations. Front. Neurosci. 11, 445 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  171. Edwards, T. L. et al. Assessment of the electronic retinal implant Alpha AMS in restoring vision to blind patients with end-stage retinitis pigmentosa. Ophthalmology 125, 432–443 (2018).

    Article  PubMed  Google Scholar 

  172. Ayton, L. N. et al. Bionic Vision Australia Research Consortium. First-in-human trial of a novel suprachoroidal retinal prosthesis. PLoS ONE 9, e115239 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  173. Fujikado, T. et al. Testing of semichronically implanted retinal prosthesis by suprachoroidal-transretinal stimulation in patients with retinitis pigmentosa. Invest. Ophthalmol. Vis. Sci. 52, 4726–4733 (2011).

    Article  PubMed  Google Scholar 

  174. Abbott, C. J. et al. Safety studies for a 44-channel suprachoroidal retinal prosthesis: a chronic passive study. Invest. Ophthalmol. Vis. Sci. 59, 1410–1424 (2018).

    Article  CAS  PubMed  Google Scholar 

  175. MacLaren, R. E. Electronic retinal implant surgery. Eye 31, 191–195 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Mathieson, K. et al. Photovoltaic retinal prosthesis with high pixel density. Nat. Photonics 6, 391–397 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Perez Fornos, A., Sommerhalder, J., Pittard, A., Safran, A. B. & Pelizzone, M. Simulation of artificial vision: IV. Visual information required to achieve simple pointing and manipulation tasks. Vis. Res. 48, 1705–1718 (2008).

    Article  PubMed  Google Scholar 

  178. Palanker, D., Le Mer, Y., Mohand-Said, S., Muqit, M. & Sahel, J. A. Photovoltaic restoration of central vision in atrophic age-related macular degeneration. Ophthalmology 127, 1097–1104 (2020).

  179. Duncan, J. L. et al. Improvements in vision-related quality of life in blind patients implanted with the Argus II epiretinal prosthesis. Clin. Exp. Optom. 100, 144–150 (2017).

    Article  PubMed  Google Scholar 

  180. Dagnelie, G. et al. Argus® II Study Group. Performance of real-world functional vision tasks by blind subjects improves after implantation with the Argus® II retinal prosthesis system. Clin. Exp. Ophthalmol. 45, 152–159 (2017).

    Article  PubMed  Google Scholar 

  181. da Cruz, L. et al. Argus II Study Group. Five-year safety and performance results from the Argus II retinal prosthesis system clinical trial. Ophthalmology 123, 2248–2254 (2016).

    Article  PubMed  Google Scholar 

  182. Luo, Y. H., Zhong, J. J. & da Cruz, L. The use of Argus® II retinal prosthesis by blind subjects to achieve localisation and prehension of objects in 3-dimensional space. Graefes Arch. Clin. Exp. Ophthalmol. 253, 1907–1914 (2015).

    Article  PubMed  Google Scholar 

  183. Kotecha, A., Zhong, J., Stewart, D. & da Cruz, L. The Argus II prosthesis facilitates reaching and grasping tasks: a case series. BMC Ophthalmol. 14, 71 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  184. da Cruz, L. et al. Argus II Study Group. The Argus II epiretinal prosthesis system allows letter and word reading and long-term function in patients with profound vision loss. Br. J. Ophthalmol. 97, 632–636 (2013).

    Article  PubMed  Google Scholar 

  185. Dorn, J. D. et al. Argus II Study Group. The detection of motion by blind subjects with the epiretinal 60-electrode (Argus II) retinal prosthesis. JAMA Ophthalmol. 131, 183–189 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  186. Geruschat, D. R. et al. An analysis of observer-rated functional vision in patients implanted with the Argus II retinal prosthesis system at three years. Clin. Exp. Optom. 99, 227–232 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  187. Jensen, R. J., Rizzo, J. F. III, Ziv, O. R., Grumet, A. & Wyatt, J. Thresholds for activation of rabbit retinal ganglion cells with an ultrafine, extracellular microelectrode. Invest. Ophthalmol. Vis. Sci. 44, 3533–3543 (2003).

    Article  PubMed  Google Scholar 

  188. Brindley, G. S. & Lewin, W. S. The visual sensations produced by electrical stimulation of the medial occipital cortex. J. Physiol. 194, 54-5P (1968).

  189. Dobelle, W. H., Mladejovsky, M. G. & Girvin, J. P. Artifical vision for the blind: electrical stimulation of visual cortex offers hope for a functional prosthesis. Science 183, 440–444 (1974).

    Article  CAS  PubMed  Google Scholar 

  190. Lewis, P. M. & Rosenfeld, J. V. Electrical stimulation of the brain and the development of cortical visual prostheses: an historical perspective. Brain Res. 1630, 208–224 (2016).

    Article  CAS  PubMed  Google Scholar 

  191. Bradley, D. C. et al. Visuotopic mapping through a multichannel stimulating implant in primate V1. J. Neurophys. 93, 1659–1670 (2005).

    Article  CAS  Google Scholar 

  192. Niketeghad, S. & Pouratian, N. Brain machine interfaces for vision restoration: the current state of cortical visual prosthetics. Neurotherapeutics 16, 134–143 (2019).

    Article  CAS  PubMed  Google Scholar 

  193. Rosenfeld, J. V. et al. Tissue response to a chronically implantable wireless intracortical visual prosthesis (Gennaris array). J. Neural Eng. 17, 046001 (2020).

    PubMed  Google Scholar 

  194. Fernández, E & Normann, R. in Artificial Vision (ed. Gabel, V. P.) 191–201 (Springer, 2017).

  195. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Zhu, J. et al. Gene and mutation independent therapy via CRISPR-Cas9 mediated cellular reprogramming in rod photoreceptors. Cell Res. 27, 830–833 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Edwards, T. L. et al. First-in-human study of the safety and viability of intraocular robotic surgery. Nat. Biomed. Eng. 2, 649–656 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Cehajic-Kapetanovic, J. et al. First-in-human robot-assisted subretinal drug delivery under local anaesthesia: a randomisedclinical trial. Am. J. Ophthalmol. https://doi.org/10.1016/j.ajo.2021.11.011 (2021).

  199. Mayoral, S. R., Etxeberria, A., Shen, Y. A. & Chan, J. R. Initiation of CNS myelination in the optic nerve is dependent on axon caliber. Cell Rep. 25, 544–550.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Ayton, L. N. et al. Harmonization of outcomes and vision endpoints in vision restoration trials: recommendations from the International HOVER Taskforce. Transl. Vis. Sci. Technol. 9, 25 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  201. Maya-Vetencourt, J. F. et al. A fully organic retinal prosthesis restores vision in a rat model of degenerative blindness. Nat. Mater. 16, 681–689 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Ha, S. et al. Towards high-resolution retinal prostheses with direct optical addressing and inductive telemetry. J. Neural Eng. 13, 056008 (2016).

    Article  PubMed  Google Scholar 

  203. Samba, R., Herrmann, T. & Zeck, G. PEDOT-CNT coated electrodes stimulate retinal neurons at low voltage amplitudes and low charge densities. J. Neural Eng. 12, 016014 (2015).

    Article  CAS  PubMed  Google Scholar 

  204. Jones, P. D. & Stelzle, M. Can nanofluidic chemical release enable fast, high resolution neurotransmitter-based neurostimulation? Front. Neurosci. 10, 138 (2016).

    PubMed  PubMed Central  Google Scholar 

  205. Albert, E. S. et al. TRPV4 channels mediate the infrared laser-evoked response in sensory neurons. J. Neurophysiol. 107, 3227–3234 (2012).

    Article  CAS  PubMed  Google Scholar 

  206. Bonmassar, G. et al. Microscopic magnetic stimulation of neural tissue. Nat. Commun. 3, 921 (2012).

    Article  PubMed  Google Scholar 

  207. Holladay, J. T. Visual acuity measurements. J. Cataract Refract. Surg. 30, 287–290 (2004).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Brett (BA Arts and Graphics) and T. Buckley (BM BCh) from Eye Research Group Oxford for their help with the images and figure design. We acknowledge funding from the University of Oxford NIHR Biomedical Research Centre. J.C.-K. was also funded by the Global Ophthalmology Fellowship Awards Programme, Bayer, Switzerland. E.Z. was supported by the DFG excellence center EXC 307 (CIN Tübingen). R.E.M. was funded by an MRC i4i Award for clinical trials of retinal implants.

Author information

Authors and Affiliations

Authors

Contributions

J.C.-K. wrote and revised the manuscript, with input from E.Z. on section ‘Retinal and cortical prostheses’, M.S.S. on section ‘Cell therapy’, and R.E.M. on the overall manuscript. All authors approved the final version of the manuscript.

Corresponding author

Correspondence to Jasmina Cehajic-Kapetanovic.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Budd Tucker and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cehajic-Kapetanovic, J., Singh, M.S., Zrenner, E. et al. Bioengineering strategies for restoring vision. Nat. Biomed. Eng 7, 387–404 (2023). https://doi.org/10.1038/s41551-021-00836-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-021-00836-4

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