Previous Article in Journal
MIGS, Cataract Surgery, or Both? An Analysis of Clinical Trial Data to Compare Efficacy and Outcomes on Glaucoma Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Optogenetics as a Novel Therapeutic Approach for Ocular Disease

Unit of Ophtalmology, Sense Organs Department, University La Sapienza of Rome, Polo Pontino-Ospedale A. Fiorini, 04019 Terracina, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Transl. Ophthalmol. 2025, 3(4), 21; https://doi.org/10.3390/jcto3040021
Submission received: 20 March 2025 / Revised: 6 August 2025 / Accepted: 15 October 2025 / Published: 20 October 2025

Abstract

Optogenetics is a field that emerged with the goal of studying the physiology of nerve cells by selectively expressing opsins—channel proteins that can be activated by light exposure. Once the methodology was established, several research groups sought to express these proteins in damaged nerve tissue to restore proper signal transmission. Over the years, numerous efforts have been made to restore vision in patients with chronic degenerative diseases, particularly retinitis pigmentosa, with clinical trials yielding encouraging results. However, significant challenges remain, such as the difficulty of delivering the signal to specific retinal cells and the complexity of replicating the physiological activation of the target cells. As research continues, optogenetics remains a promising yet evolving field. This review aims to highlight the therapeutic advantages of optogenetics over currently available strategies and to promote further scientific exploration of this emerging discipline.

1. Introduction

Optogenetics is a groundbreaking technique that enables the precise control of specific cell types in living tissues using light. Optogenetics originated in 2006 from the studies of Karl Deisseroth, with the ambition of making any neuronal cell capable of activating or inhibiting itself under light stimulation thanks to gene vectors encoding light-sensitive channel proteins [1]. In attempting to apply these techniques, difficulties have emerged in reproducing actual physiological functions due to deregulation of the extent of activation or inhibition and structural alterations due to vector transport techniques and the proteins themselves being over-expressed within the cell [2]. At the basis of this technology are ‘opsins’, a family of seven-tailed transmembrane proteins that are able to modulate their action with light exposure. Opsins are divided into type I and type II. Type I opsins are present in prokaryotes, algae, and fungi; they have been associated with 11-trans-retinal, which upon light exposure isomerizes to 11-cis-retinal, whereupon a conformational change in the protein allows for the passage of ions. Then, 11-cis-retinal is reconverted to 11-trans-retinal, while still remaining bound to the protein [3]. Type II opsins, found in eukaryotes, on the other hand, have an associated G protein, are in the 11-cis-retinal conformation, and upon light exposure become 11-trans-retinal; this step is followed by dissociation of the retinal from the protein. This is why type I opsins, which have faster activation and inhibition kinetics, are preferred in optogenetic studies, as they are independent of the 11-cis-retinal reassociation time and the G-protein-associated pathway [4].
The first opsins to be applied are ChannelRhodopsin-1 (ChR-1), followed by ChR-2, isolated from the green alga Chlamydomonas reinhardtii; exposure to blue light (470 nm) results in the opening and passage of cations, which leads to depolarization and cell activation [5,6]. Halorhodopsins were then introduced, first isolated from the archaea Natronomonas pharaonis (NpHR); exposure to yellow light (580 nm) results in its opening and passage of chlorine anions, which results in hyper-polarization and neuron inactivation (Han & Boyden, 2007) [7]. Finally, the ArchaeRhodopsins isolated from Halorubrum sodomense act as proton pumps to the outside of the cell, capable of hyperpolarising and inhibiting neurons; this is in the green-yellow range (566 nm) [7].
The first applications in the field of ophthalmology were proposed in 2006 by Anding’s Bi et al. study group; they demonstrated in retinal tissues with damaged photoreceptors that it is possible to restore retinal signals and in the visual cortex, thanks to the functional restoration of ganglion cells, by using ChR transported by adenoviral-associated vectors (AAV); the limitations were due to the lack of selectivity of ON and OFF cell activation, thus making bipolar cells the ideal target to restore in the case of photoreceptor damage [8]. In fact, studies by Legali et al. demonstrated how activation of ON bipolar cells by ChR introduced by electroporation could evoke responses similar to physiological ones [9]. However, as electroporation was not very applicable in vivo, adenoviral vectors were then proposed, among the most recently developed being those by ‘direct evolution’, capable of penetrating the layers of the retina with greater efficiency, up to the bipolar cells and then the photoreceptors and retinal pigment epithelium; this was done by intravitreal injection, sparing the more invasive sub-retinal injection [10].

2. Materials and Methods

We conducted a search of the literature, including all publication years up to January 2024, using MEDLINE (PubMed). The database was first searched using the following keywords: “optogenetic, retinitis pigmentosa, adenovirus, genetic therapy, inherited retinal disease, glaucoma”. The publication types included were reviews and clinical trials. We considered only studies in English and those with an abstract, thus reducing the count to 281 papers. The reference lists of all retrieved articles were assessed to identify additional relevant studies. The research of articles was performed using PubMed (https://pubmed.ncbi.nlm.nih.gov, accessed on 1 October 2023) and Reference Citation Analysis (https://www.referencecitationanalysis.com, accessed on 1 October 2023).

3. Optogenetics in Retinal Degenerative Disease

Optogenetics has emerged as a promising therapeutic approach for treating retinal diseases, particularly those involving degenerative conditions. By selectively targeting specific retinal cells, optogenetics aims to restore visual perception in patients with damaged photoreceptors, offering the potential for functional vision recovery.

3.1. Retinal Degenerative Disease

3.1.1. Retinitis Pigmentosa

Among hereditary degenerative diseases of the retina, Retinitis pigmentosa (RP) is a spectrum of clinically similar and genetically heterogeneous phenotypes characterized by primary degeneration of rod and cone photoreceptors [11]. They are divided into syndromic and non-syndromic RP subtypes. The clinical manifestations of non-syndromic RP (being the most frequent subtype of the disease) include a progressive pattern of night blindness occurring mostly in the first three decades of life and a subsequent narrowing of the visual field due to the loss of the rods. The advanced degeneration also involves the cones, resulting in central vision loss. RP is a leading cause of visual disability, with a worldwide prevalence of 1:4000 [12]. With the progression of the disease and the occurrence of complete loss of optic rod and cone cells, gene- and cell-replacement therapies become ineffective, and it is necessary to consider other approaches aimed at treating advanced retinal degeneration. The abovementioned new approach can be represented by optogenetics, a field that is still under development [13].

3.1.2. Stargardt Disease

Another progressive inherited retinal disorder is Stargardt disease, primarily affecting the macula, the central part of the retina responsible for detailed vision. It is caused by mutations in the ABCA4 gene, which encodes a protein involved in the transport of retinoids in photoreceptor cells. This malfunction leads to the accumulation of toxic substances within retinal cells, particularly in the cones, causing their degeneration and subsequent vision loss. Early symptoms typically include blurred vision, difficulty seeing in low light, and central vision loss, often manifesting in childhood or adolescence. Similarly other retinal degenerations, once cone cells are significantly damaged, treatments targeting the underlying causes may become ineffective. However, gene therapy and optogenetic approaches are being explored as promising strategies for restoring vision. Recent studies have shown the potential of optogenetic tools to restore light sensitivity in surviving retinal cells, such as retinal ganglion cells or bipolar cells, offering hope for vision recovery in Stargardt disease patients. Research by Piotter et al. (2021) has explored various therapeutic approaches, including gene therapy and optogenetics, with encouraging results in preclinical studies [14]. In this context, the work by Huang et al. (2022) provides insight into the therapeutic strategies under investigation for Stargardt, highlighting the potential for optogenetic approaches to restore some functional vision by targeting surviving retinal cells [15].

3.1.3. Age-Related Macular Degeneration

Age-related macular degeneration (AMD is one of the leading causes of vision impairment in older adults, primarily affecting those over the age of 55. Unlike RP or Stargardt disease, it is not strictly an inherited condition, although it possesses a strong genetic susceptibility. AMD is considered a multifactorial disease, where both genetic predisposition and environmental factors, such as aging and exposure to oxidative stress and inflammation, play significant roles. It is a degenerative disease of the retina that primarily affects the macula and presents, according to the Ferris 2014 classification [16], as intermediate AMD in subjects with large drusen (greater than ≥125 μm) or with pigmentary abnormalities associated with drusen and late AMD. Late AMD exists in two forms: dry (atrophic) AMD, characterized by gradual retinal cell breakdown, and wet (neovascular) AMD, in which abnormal blood vessels grow beneath the retina and leak fluid, leading to further damage. Genetic alterations in biochemical pathways such as angiogenesis (VEGFA), lipid transport and metabolism (APOE), modulation of extracellular matrix (COL101A, MMP9, and TIMP3), metabolism of all-trans-retinaldehyde (ABCA4), and complement cascade (CFH, CFD, C3, C5, C7) are involved in the pathogenesis of the disease [17]. Many of these genes are also implicated in the regulation of oxidative stress. For instance, ABCA4 dysfunction leads to the accumulation of toxic bisretinoids such as A2E, which generate reactive oxygen species (ROS) under light exposure. Similarly, dysregulation of APOE and CFH has been associated with impaired antioxidant defense mechanisms and increased susceptibility to oxidative damage in retinal cells. These alterations exacerbate mitochondrial dysfunction and chronic inflammation, both of which are key contributors to oxidative stress and photoreceptor degeneration. The degeneration of retinal cells, particularly the photoreceptors in the macula, leads to significant central vision loss, severely affecting daily activities like reading and recognizing faces.
For the treatment of AMD, anti-VEGF therapies target vascular endothelial growth factor (VEGF) to inhibit the formation of abnormal blood vessels. They have become a mainstay for wet AMD. These therapies have demonstrated considerable efficacy in preventing further vision loss, but they do not restore lost vision. On the other hand, gene therapy approaches are being explored as a means of addressing the underlying cellular degeneration. Researchers are working on gene-editing tools and viral vectors to deliver healthy copies of genes or to silence deleterious genes, with the aim of halting or even reversing the degenerative process [18,19]. Stem cell-based therapies, while holding promise for replacing damaged retinal pigment epithelium (RPE) cells, currently face several limitations. These include challenges related to cell survival and integration, immune rejection, and the risk of uncontrolled cell proliferation. Moreover, variability in differentiation efficiency and delivery methods hampers the reproducibility and safety of these treatments in clinical settings [11]. In addition to these treatments, optogenetics is emerging as a promising approach for restoring vision in advanced stages of AMD. Unlike gene therapy, which aims to repair or replace damaged genes, optogenetics involves introducing light-sensitive proteins into surviving retinal cells (ganglion or bipolar cells) in order to restore light sensitivity and enable these cells to relay visual information to the brain. This approach is particularly relevant for patients who have lost their photoreceptors but still have some remaining retinal cell function. Studies suggest that optogenetic treatments could help bypass the damaged photoreceptors and restore partial vision, especially in patients with geographic atrophy, a severe form of dry AMD [20].

3.2. Preclinical Studies

Although optogenetics is an evolving science, numerous preclinical and clinical studies have been conducted, with preclinical studies having shown very encouraging results on visual recovery in animal models. One of the first preclinical studies demonstrated that a Chop2-GFP chimera transfected through AAV into the retinal ganglion cells of rd1 mice enabled the retinal ganglion cells to encode light signals into action potentials capable of reaching the visual cortex [8]. Hiroshi Tomita et al. studied the green algae rhodopsin archaeotype, called channelrhodopsin-2 (ChR2), as an optogenetic tool enabling retinal ganglion cells type ON to send electrical signals to the rodent cortex in response to light. Rats with photoreceptor degeneration, after the expression of ChR2 in retinal ganglion cells (RGCs) and regulation by Thy-1.2 promoter, have demonstrated head-tracking behavior during light–dark reticles. Moreover, a change in the electrophysiology of RGCs was found. This highlights ChR2’s potential for application in patients with retinitis pigmentosa, aiming to restore their vision [21].
In the clinical course of retinal diseases, as demonstrated in the rd1 and rd10 mice, ganglion and bipolar cells are relatively preserved, but due to the loss of photoreceptors and their synapses, they undergo regressive remodeling. Furthermore, under normal conditions, ganglion cells show a high degree of variability in response to light stimulus: OFF ganglion cells reduce their activity, while ON cells increase it. Based on these notions, targeting ganglion cells in optogenetic therapy can be complex. Moreover, being at a more extreme level of the visual information processing system, a loss of electrical–visual information may occur due to the absence of processing by bipolar cells, amacrine cells, and horizontal cells. However, access to these cells is complex due to their internal location in the retinal layers. Since the retina consists of well-separated layers and two limiting membranes, access to the outermost layers from the vitreous chamber is difficult for vectors. For this reason, intravitreal injection may require either peeling of the ILM, a highly invasive procedure, or the use of high concentrations of adenoviruses, which could cause unexpected inflammatory responses. The alternative sub-retinal injection, however, exposes patients to high risks, such as rhegmatogenous retinal detachment [22]. Furthermore, it is not easy to develop vectors with selective promoters for bipolar cells cause Adenoviral vectors can carry small gene packages, approximately 5 kilobases (kb); therefore, it is not possible to use large, highly specific promoters, such as L7 for bipolar cells, requiring the use of more compressed promoters [22]. Numerous preclinical studies on the use of opsins in animal models are currently underway, as summarized in Table 1.
Pamela S Lagali et al. [9] highlighted critical issues regarding the use of retinal ganglion cells as targets for optogenetic therapy. The authors transfected the bipolar ON cells with ChR2 in mice rd1, achieving a recovery of light sensitivity. That finding revealed the photosensitivity of bipolar ON cells, alongside their ability to induce light-evoked spiking activity in RGCs. Moreover, the authors observed transient responses and maintenance of central organization of ganglion cells. These characteristics enabled the creation of information in the visual cortex of rodents, enabling them to successfully perform r [9]. However, Chr2 has a low photosensitivity to blue light, require signal amplification devices that must be highly precise in terms of time, space, and intensity to produce activation similar to physiological activation. Furthermore, the amplification of wavelengths such as blue can generate phototoxicity that can damage the residual cells of the retina. This is because photons in the blue spectrum have greater energy than those in the red spectrum; this energy is first absorbed by endogenous photosensitizers such as NADP and FMN and then transferred by photosensitization to oxygen, generating free radicals [23]. Furthermore, chronic exposure to blue light could lead to excessive activation of intrinsically photosensitive retinal ganglion cells (ipRGCs), cells capable of regulating the release of melatonin, resulting in possible disruption of the circadian rhythm [24].
To overcome this issue, Berry et al. transferred a gene of medium wavelength cone opsin (MW-opsin) in ON and OFF retinal ganglion cells of rd1 mice and achieved improved recognition of both two-dimensional spatial patterns on LCD screens and three-dimensional object recognition with internal light [25]. Weldon W. Wright et al. transfected bipolar ON cells of rd10 mice in vivo by intravitreal administration of adeno-associated virus (vMCO1) with a multi-characteristic opsin capable of generating transmembrane current in response to environmental light stimulation [26]. Their study involved analyzing visuospatial behavior in a radial visual water maze, which was faster in the mice transfected with MCO1 than in the rd10 control arm without MCO1. MCO1 demonstrated greater advantages over medium wavelength cone opsin (MW-opsin) with regard to spectral sensitivity, a fast response time, and repeatability over multiple stimulations [27]. Gregory Gauvain et al. tested AAV2.7m8—ChR-tdT (ChrimsonR-tdTomato) vector in perifoveal retinal ganglion cells of non-human primates. In their study, they calculated the infectious dose required to generate sufficient numbers of cells expressing the optogenic construct. They used a type of opsin that reacts to red light with a safer exposure/response dose profile. Their observation was that this type of opsin only reacted to an intensity of light that is not present in everyday environments. Therefore, they used an external stimulation device capable of amplifying photons from an image and triggering the activation of cells expressing [28]. However, understanding visual quality in primates is complex, as until now the restoration of visual abilities has been evaluated by focusing on contrast sensitivity and horizontal movement, excluding more complex functions such as facial recognition or visual memory activation [29].
With current technologies, optogenetics is capable of restoring a level of vision no greater than legal blindness, which raises the ethical issue of subjecting patients to invasive procedures with long-term effects that are still unknown, without producing benefits that could positively transform their quality of life [30].
Table 1. Summary of ongoing preclinical studies of optogenetic therapy.
Table 1. Summary of ongoing preclinical studies of optogenetic therapy.
ResearchersOpsinPopulationTargetYear
Pamela S Lagali et al. [9]ChR2Rd1Retinal bipolar ON cells2008
Hiroshi Tomita et al. [21]ChR2-Thy-1.2 promoterW-TChR2VGanglion cells type ON2009
Chow et al. [8]Chop2-GFP chimeraRd1Retinal ganglion cells2010
Weldon W. Wright et al. [23]vMCO1rd10Retinal bipolar ON cells2017
Berry et al. [22]MW-opsinRd1ON and OFF retinal ganglion cells2019
Gregory Gauvain et al. [25]ChR-tdTnon-human primatesPerifoveal retinal ganglion cells2021
Yan et al. [28]ChronosFPRd1Retinal ganglion cells2023

3.3. Clinical Applications

The results of Gregory Gauvin et al. [28] contributed to the initiation of PIONEER, a Phase 1/2a open-label, dose-escalation study sponsored by GenSight Biologics (Paris, France), designed to evaluate the safety and tolerability of GS030 in patients with advanced non-syndromic retinitis pigmentosa (RP). This approach targets retinal ganglion cells using the light-sensitive opsin ChrimsonR (ClinicalTrials.gov Identifier: NCT03326336). In addition to this study, other clinical trials are currently underway, as summarized in Table 2.
Sahel et al. [31] described a notable case of partial visual recovery in a 58-year-old male participant from the PIONEER study, affected by advanced non-syndromic RP with only light perception. Following a single intravitreal injection of the optogenetic vector GS030-DP (ChrimsonR-tdTomato delivered via an AAV2.7m8 vector), combined with a visual training program using light-stimulating goggles (GS030-MD), the patient experienced improvements in visual perception, visuomotor coordination, and EEG responses. The study is ongoing, with an estimated completion date in December 2025.
In parallel, RESTORE (NCT04945772) is a completed Phase 2b, multicenter, randomized, double-masked, sham-controlled, dose-ranging study sponsored by Nanoscope Therapeutics. It aimed to assess the efficacy and safety of a single intravitreal injection of MCO-010, a virally delivered multi-characteristic opsin targeting ON-bipolar cells, in adults with RP. While the study has concluded, the final results have not yet been published (ClinicalTrials.gov, accessed 9 February 2025).
Nanoscope Therapeutics has also conducted STARLIGHT (NCT05417126), a clinical trial evaluating the safety and efficacy of a single intravitreal injection of vMCO-010 in patients with Stargardt disease. This therapy also targets ON-bipolar cells. During a 48-week follow-up, participants with macular involvement demonstrated progressive improvements in visual acuity [32]. The results have been submitted and are undergoing quality control review. Furthermore, Nanoscope Therapeutics has announced plans to initiate a Phase 3 clinical trial for MCO-010 in Stargardt macular degeneration. Notably, MCO-010 has received FDA fast-track designation and orphan drug designation for both RP and Stargardt disease.
Another promising approach is BS01, a Phase 1/2 clinical trial evaluating the safety and efficacy of an intravitreal injection of BS01, an AAV2-based vector encoding ChronosFP—a light-sensitive opsin with high light sensitivity and fast kinetics. ChronosFP targets retinal ganglion cells and is coupled with an intraocular encoder–transducer system that converts visual input into retinal neural signals, effectively simulating a functional preganglionic retina [33]. The trial is currently in the recruitment phase (ClinicalTrials.gov Identifier: NCT04278131; accessed 9 February 2025).

4. Optogenetics in Other Ophthalmic Diseases

One of the most promising, yet less explored, areas is the application of optogenetics in optic nerve disorders, particularly glaucoma and ischemic optic neuropathies. These conditions are major causes of irreversible blindness, largely due to the degeneration of retinal ganglion cells (RGCs) and their axons. Traditional treatments focus primarily on slowing disease progression (e.g., reducing intraocular pressure in glaucoma), but fail to restore lost neuronal function. Optogenetics offers potential not only for neuroprotection but also for functional restoration in these optic nerve pathologies. A significant portion of optogenetic research in glaucoma focuses on modulating intraocular pressure (IOP), the primary modifiable risk factor in the disease. Traditional pharmacological and surgical interventions often provide inconsistent outcomes, particularly in steroid-induced glaucoma, where trabecular meshwork (TM) dysfunction leads to increased IOP. Recent optogenetic studies have demonstrated the potential for precise control of TM function through targeted light activation. In a groundbreaking study, Kowal et al. utilized the CRY2-CIBN optogenetic system to regulate the activity of OCRL 5-phosphatase, an enzyme involved in maintaining cytoskeletal dynamics in TM cells [33]. By delivering CRY2-OCRL constructs via AAV2 vectors into the anterior chamber and applying blue light stimulation, they successfully reversed glucocorticoid-induced TM dysfunction in a mouse model. This optogenetic approach led to enhanced aqueous humor outflow and a corresponding reduction in IOP, highlighting a novel non-invasive strategy for managing steroid-induced ocular hypertension. In parallel, optogenetics has shown promise as a neuroprotective tool by targeting not only the eye but also the visual centers in the brain. One of the most innovative studies in this area, conducted by Geeraerts et al., investigated the neuroprotective effects of optogenetic stimulation of the superior colliculus (SC) in a glaucoma mouse model [34]. The SC, a primary projection target of RGCs, plays a crucial role in visual processing and motor responses to visual stimuli. The researchers used a stabilized step-function opsin (SSFO) to achieve prolonged activation of SC neurons with minimal light exposure. This targeted stimulation resulted in a 63% reduction in RGC loss, suggesting that enhancing neuronal activity in the SC promotes retrograde neuroprotective signaling to the retina. These findings highlight the potential of brain-targeted optogenetic therapies as a complementary approach to traditional ocular treatments in glaucoma. While neuroprotection is critical for halting disease progression, the ultimate goal in treating optic nerve disorders is the restoration of visual function. Optogenetics offers a promising avenue for this by rendering surviving RGCs light-sensitive, thus bypassing damaged photoreceptors and optic nerve fibers. In a comprehensive review, Prosseda et al. detailed various optogenetic strategies aimed at restoring visual function in optic neuropathies [35]. One approach involves the use of ChR2 and melanopsin (OPN4), introduced into RGCs via AAV-mediated gene delivery. These opsins confer light sensitivity to the remaining retinal neurons, allowing for the partial restoration of visual perception even in cases of advanced optic nerve damage. This technique is particularly advantageous because it is independent of the underlying genetic cause of the optic nerve disorder, making it applicable across a wide range of conditions, from glaucoma to ischemic optic neuropathies. Preliminary clinical trials have already shown encouraging results. Patients with advanced optic nerve damage have demonstrated partial recovery of light perception following optogenetic treatment [36], marking a significant milestone in the field of vision restoration. In addition, recent preclinical studies focused specifically on optic nerve injury have demonstrated promising outcomes: for example, in animal models of optic nerve crush (ONC), AAV-mediated delivery of F-iTrkB led to significant retinal ganglion cell protection, axonal regeneration, and improvement in visual function as measured by optokinetic response and visual evoked potentials (VEPs) [37].
However, the functional outcomes are still limited by factors such as light intensity requirements and the spatial resolution of the reactivated neurons. To address these challenges, researchers are developing red-shifted opsins, which respond to longer wavelengths of light, allowing for deeper tissue penetration and more naturalistic visual experiences.

5. Conclusions

Optogenetics represents a significant advancement in the field of ophthalmology, offering promising therapeutic strategies for the treatment of retinal degenerative diseases, including retinitis pigmentosa, Stargardt disease, and age-related macular degeneration. The ability to restore light sensitivity in surviving retinal cells, such as ganglion and bipolar cells, offers hope for patients with advanced retinal degeneration, who would otherwise be untreatable by traditional gene and cell replacement therapies. Through the use of light-sensitive opsins, optogenetic techniques have shown considerable potential to bypass damaged photoreceptors, enabling the restoration of functional vision.
Clinical trials, such as those exploring the use of MCO and ChR2, are gradually translating these preclinical successes into human applications, demonstrating partial recovery of visual function in patients with advanced retinal diseases. Additionally, optogenetics holds promise for treating optic nerve disorders, such as glaucoma and ischemic optic neuropathies, where traditional treatments focus primarily on slowing disease progression but do not restore lost visual function. By leveraging the power of light to modulate neuronal activity, optogenetics could offer a new avenue for neuroprotection and the restoration of vision in conditions that were previously deemed irreversible.
Despite these encouraging results, several challenges remain in refining optogenetic therapies for widespread clinical use. Issues such as the required light intensity, potential retinal cell damage, and the need for precise targeting of opsin expression must be addressed. However, with continued advancements in opsin technology, viral vector delivery systems, and personalized treatment strategies, optogenetics is poised to revolutionize the treatment of retinal degenerative diseases and optic nerve disorders, offering hope for millions of patients worldwide. Future research and clinical trials will be critical in optimizing these techniques, ensuring safety, efficacy, and long-term benefits for individuals suffering from vision loss due to retinal and optic nerve pathologies.

Author Contributions

Conceptualization, E.M.V. and S.M.; methodology, E.M.V. and S.M.; software, S.M., F.M. and M.C.; validation, E.M.V.; formal analysis S.M., M.B. and F.M.; investigation, M.C., M.B. and F.M.; resources, S.M., F.M., M.B. and M.C.; data curation, S.M.; writing—original draft preparation, E.M.V. and S.M.; writing—review and editing, S.M., F.M., M.B. and M.C.; visualization, M.B., M.C. and F.M.; supervision, E.M.V.; project administration, E.M.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions of this study are included in the article. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Deisseroth, K.; Feng, G.; Majewska, A.K.; Miesenböck, G.; Ting, A.; Schnitzer, M.J. Next-generation optical technologies for illuminating genetically targeted brain circuits. J. Neurosci. Off. J. Soc. Neurosci. 2006, 26, 10380–10386. [Google Scholar] [CrossRef]
  2. Häusser, M. Optogenetics: The age of light. Nat. Methods 2014, 11, 1012–1014. [Google Scholar] [CrossRef] [PubMed]
  3. Haupts, U.; Tittor, J.; Bamberg, E.; Oesterhelt, D. General Concept for Ion Translocation by Halobacterial Retinal Proteins: The Isomerization/Switch/Transfer (IST) Model. Biochemistry 1997, 36, 2–7. [Google Scholar] [CrossRef] [PubMed]
  4. Boyden, E.S.; Zhang, F.; Bamberg, E.; Nagel, G.; Deisseroth, K. Millisecond-timescale, genetically targeted optical control of neural activity. Nat. Neurosci. 2005, 8, 1263–1268. [Google Scholar] [CrossRef] [PubMed]
  5. Nagel, G.; Ollig, D.; Fuhrmann, M.; Kateriya, S.; Musti, A.M.; Bamberg, E.; Hegemann, P. Channelrhodopsin-1: A Light-Gated Proton Channel in Green Algae. Science 2002, 296, 2395–2398. [Google Scholar] [CrossRef]
  6. Nagel, G.; Szellas, T.; Huhn, W.; Kateriya, S.; Adeishvili, N.; Berthold, P.; Ollig, D.; Hegemann, P.; Bamberg, E. Channelrhodopsin-2, a directly light-gated cation-selective membrane channel. Proc. Natl. Acad. Sci. USA 2003, 100, 13940–13945. [Google Scholar] [CrossRef]
  7. Chow, B.Y.; Han, X.; Dobry, A.S.; Qian, X.; Chuong, A.S.; Li, M.; Henninger, M.A.; Belfort, G.M.; Lin, Y.; Monahan, P.E.; et al. High-performance genetically targetable optical neural silencing by light-driven proton pumps. Nature 2010, 463, 98–102. [Google Scholar] [CrossRef]
  8. Bi, A.; Cui, J.; Ma, Y.-P.; Olshevskaya, E.; Pu, M.; Dizhoor, A.M.; Pan, Z.-H. Ectopic expression of a microbial-type rhodopsin restores visual responses in mice with photoreceptor degeneration. Neuron 2006, 50, 23–33. [Google Scholar] [CrossRef]
  9. Lagali, P.S.; Balya, D.; Awatramani, G.B.; Münch, T.A.; Kim, D.S.; Busskamp, V.; Cepko, C.L.; Roska, B. Light-activated channels targeted to ON bipolar cells restore visual function in retinal degeneration. Nat. Neurosci. 2008, 11, 667–675. [Google Scholar] [CrossRef]
  10. Dalkara, D.; Byrne, L.C.; Klimczak, R.R.; Visel, M.; Yin, L.; Merigan, W.H.; Flannery, J.G.; Schaffer, D.V. In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. Sci. Transl. Med. 2013, 5, 189ra76. [Google Scholar] [CrossRef]
  11. Vingolo, E.M.; Mascolo, S.; Miccichè, F.; Manco, G. Retinitis Pigmentosa: From Pathomolecular Mechanisms to Therapeutic Strategies. Medicina 2024, 60, 189. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Verbakel, S.K.; van Huet, R.A.C.; Boon, C.J.F.; den Hollander, A.I.; Collin, R.W.J.; Klaver, C.C.W.; Hoyng, C.B.; Roepman, R.; Jeroen Klevering, B. Non-syndromic retinitis pigmentosa. Prog. Retin. Eye Res. 2018, 66, 157–186. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, W.; Liu, S.; Li, P.; Yao, K. Retinitis Pigmentosa: Progress in Molecular Pathology and Biotherapeutical Strategies. Int. J. Mol. Sci. 2022, 23, 4883. [Google Scholar] [CrossRef] [PubMed]
  14. Piotter, E.; McClements, M.E.; MacLaren, R.E. Therapy Approaches for Stargardt Disease. Biomolecules 2021, 11, 1179. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. Huang, D.; Heath Jeffery, R.C.; Aung-Htut, M.T.; McLenachan, S.; Fletcher, S.; Wilton, S.D.; Chen, F.K. Stargardt disease and progress in therapeutic strategies. Ophthalmic Genet. 2022, 43, 1–26. [Google Scholar] [CrossRef] [PubMed]
  16. Ferris, F.L., 3rd; Wilkinson, C.P.; Bird, A.; Chakravarthy, U.; Chew, E.; Csaky, K.; Sadda, S.R. Beckman Initiative for Macular Research Classification Committee. Clinical classification of age-related macular degeneration. Ophthalmology 2013, 120, 844–851. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  17. Fritsche, L.G.; Igl, W.; Bailey, J.N.C.; Grassmann, F.; Sengupta, S.; Bragg-Gresham, J.L.; Burdon, K.P.; Hebbring, S.J.; Wen, C.; Gorski, M.; et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat. Genet. 2016, 48, 134–143. [Google Scholar] [CrossRef]
  18. van Lookeren Campagne, M.; LeCouter, J.; Yaspan, B.L.; Ye, W. Mechanisms of age-related macular degeneration and therapeutic opportunities. J. Pathol. 2014, 232, 151–164. [Google Scholar] [CrossRef] [PubMed]
  19. Fleckenstein, M.; Keenan, T.D.L.; Guymer, R.H.; Chakravarthy, U.; Schmitz-Valckenberg, S.; Klaver, C.C.; Wong, W.T.; Chew, E.Y. Age-related macular degeneration. Nat. Rev. Dis. Primers 2021, 7, 31. [Google Scholar] [CrossRef] [PubMed]
  20. Borchert, G.A.; Shamsnajafabadi, H.; Ng, B.W.J.; Xue, K.; De Silva, S.R.; Downes, S.M.; MacLaren, R.E.; Cehajic-Kapetanovic, J. Age-related macular degeneration: Suitability of optogenetic therapy for geographic atrophy. Front. Neurosci. 2024, 18, 1415575. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Tomita, H.; Sugano, E.; Fukazawa, Y.; Isago, H.; Sugiyama, Y.; Hiroi, T.; Ishizuka, T.; Mushiake, H.; Kato, M.; Hirabayashi, M.; et al. Visual Properties of Transgenic Rats Harboring the Channelrhodopsin-2 Gene Regulated by the Thy-1.2 Promoter. PLoS ONE 2009, 4, e7679. [Google Scholar] [CrossRef]
  22. Wu, Z.; Yang, H.; Colosi, P. Effect of genome size on AAV vector packaging. Mol. Ther. J. Am. Soc. Gene Ther. 2010, 18, 80–86. [Google Scholar] [CrossRef]
  23. Feldbauer, K.; Zimmermann, D.; Pintschovius, V.; Spitz, J.; Bamann, C.; Bamberg, E. Channelrhodopsin-2 is a leaky proton pump. Proc. Natl. Acad. Sci. USA 2009, 106, 12317–12322. [Google Scholar] [CrossRef] [PubMed]
  24. Salceda, R. Light Pollution and Oxidative Stress: Effects on Retina and Human Health. Antioxidants 2024, 13, 362. [Google Scholar] [CrossRef] [PubMed]
  25. Berry, M.H.; Holt, A.; Salari, A.; Veit, J.; Visel, M.; Levitz, J.; Aghi, K.; Gaub, B.M.; Sivyer, B.; Flannery, J.G.; et al. Restoration of high-sensitivity and adapting vision with a cone opsin. Nat. Commun. 2019, 10, 1221. [Google Scholar] [CrossRef] [PubMed]
  26. Wright, W.; Gajjeraman, S.; Batabyal, S.; Pradhan, S.; Bhattacharya, S.; Mahapatra, V.; Tripathy, A.; Mohanty, S. Restoring vision in mice with retinal degeneration using multicharacteristic opsin. Neurophotonics 2017, 4, 041505. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  27. Batabyal, S.; Gajjeraman, S.; Pradhan, S.; Bhattacharya, S.; Wright, W.; Mohanty, S. Sensitization of ON-bipolar cells with ambient light activatable multi-characteristic opsin rescues vision in mice. Gene Ther. 2021, 28, 162–176. [Google Scholar] [CrossRef]
  28. Gauvain, G.; Akolkar, H.; Chaffiol, A.; Arcizet, F.; Khoei, M.A.; Desrosiers, M.; Jaillard, C.; Caplette, R.; Marre, O.; Bertin, S.; et al. Optogenetic therapy: High spatiotemporal resolution and pattern discrimination compatible with vision restoration in non-human primates. Commun. Biol. 2021, 4, 125. [Google Scholar] [CrossRef]
  29. Ferrari, U.; Deny, S.; Sengupta, A.; Caplette, R.; Trapani, F.; Sahel, J.A.; Dalkara, D.; Picaud, S.; Duebel, J.; Marre, O. Towards optogenetic vision restoration with high resolution. PLoS Comput. Biol. 2020, 16, e1007857. [Google Scholar] [CrossRef]
  30. Harris, A.R.; Gilbert, F. Restoring vision using optogenetics without being blind to the risks. Graefe’s Arch. Clin. Exp. Ophthalmol. 2022, 260, 41–45. [Google Scholar] [CrossRef]
  31. Sahel, J.A.; Boulanger-Scemama, E.; Pagot, C.; Arleo, A.; Galluppi, F.; Martel, J.N.; Esposti, S.D.; Delaux, A.; de Saint Aubert, J.B.; de Montleau, C.; et al. Partial recovery of visual function in a blind patient after optogenetic therapy. Nat. Med. 2021, 27, 1223–1229. [Google Scholar] [CrossRef] [PubMed]
  32. Mahajan, V.B. Longitudinal BCVA Analysis of Patients with Stargardt Disease and Macular Degeneration Treated with MCO-010, a Mutation-Agnostic Optogenetic Therapy: 48-Week Results from a Phase 2a Clinical Trial (STARLIGHT). Investig. Ophthalmol. Vis. Sci. 2024, 65, 5266. [Google Scholar]
  33. Yan, B.; Viswanathan, S.; Brodie, S.E.; Deng, W.T.; Coleman, K.E.; Hauswirth, W.W.; Nirenberg, S. A clinically viable approach to restoring visual function using optogenetic gene therapy. Mol. Ther. Methods Clin. Dev. 2023, 29, 406–417. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Kowal, D.R.; Prosseda, P.P.; Ning, K.; Wang, B.; Alvarado, J.; Sendayen, B.E.; Jabbehdari, S.; Stamer, W.D.; Hu, Y.; Sun, Y. Optogenetic control of intraocular pressure in a glucocorticoid-induced ocular hypertension mouse model. Biomolecules 2021, 12, 269. [Google Scholar]
  35. Geeraerts, E.; Claes, M.; Dekeyster, E.; Salinas-Navarro, M.; De Groef, L.; Van den Haute, C.; Scheyltjens, I.; Baekelandt, V.; Arckens, L.; Moons, L. Optogenetic stimulation of the superior colliculus confers retinal neuroprotection in a mouse glaucoma model. Neurobiol. Dis. 2019, 39, 2313–2325. [Google Scholar] [CrossRef]
  36. Prosseda, P.P.; Tran, M.; Kowal, T.; Wang, B.; Sun, Y. Advances in Ophthalmic Optogenetics: Approaches and Applications. Biomolecules 2022, 12, 269. [Google Scholar] [CrossRef]
  37. Harada, C.; Guo, X.; Harada, T. Monogenic gene therapy for glaucoma and optic nerve injury. Neural Regen. Res. 2025, 20, 815–816. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Table 2. Summary of ongoing clinical trials of optogenetic therapy.
Table 2. Summary of ongoing clinical trials of optogenetic therapy.
Clinical Trial IdentifierCompanyDiseaseInterventionTrial Stage
NCT03326336GenSight BiologicsRetinitis PigmentosaDrug: GS030-DP Medical device: GS030-MDPhase 1/2a
NCT04945772Nanoscope TherapeuticsRetinitis PigmentosaDrug: MCO-010Phase 2b
NCT05417126Nanoscope TherapeuticsStargardtDrug: vMCO-010Phase 2b
NCT04278131Bionic Sight LLCRetinitis PigmentosaDrug: BS01Phase 1/2
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vingolo, E.M.; Mascolo, S.; Calabro, M.; Miccichè, F.; Barresi, M. Optogenetics as a Novel Therapeutic Approach for Ocular Disease. J. Clin. Transl. Ophthalmol. 2025, 3, 21. https://doi.org/10.3390/jcto3040021

AMA Style

Vingolo EM, Mascolo S, Calabro M, Miccichè F, Barresi M. Optogenetics as a Novel Therapeutic Approach for Ocular Disease. Journal of Clinical & Translational Ophthalmology. 2025; 3(4):21. https://doi.org/10.3390/jcto3040021

Chicago/Turabian Style

Vingolo, Enzo Maria, Simona Mascolo, Mattia Calabro, Filippo Miccichè, and Mirko Barresi. 2025. "Optogenetics as a Novel Therapeutic Approach for Ocular Disease" Journal of Clinical & Translational Ophthalmology 3, no. 4: 21. https://doi.org/10.3390/jcto3040021

APA Style

Vingolo, E. M., Mascolo, S., Calabro, M., Miccichè, F., & Barresi, M. (2025). Optogenetics as a Novel Therapeutic Approach for Ocular Disease. Journal of Clinical & Translational Ophthalmology, 3(4), 21. https://doi.org/10.3390/jcto3040021

Article Metrics

Back to TopTop