Volume: 3 | Issue: 3 | View as PDF | Cite
  • OPEN ACCESS

Decoding Retinal Degeneration Diseases: From Molecular Pathways to Cutting-edge Therapies

  • Youyuan Zhuang1,2,3,
  • Jiuyi Lu1,2,3,
  • Jiawen Li1,2,3,
  • Chong Chen1,2,3,*  and
  • Xinting Liu1,2,3,* 
 Author information
Nature Cell and Science   2025;3(3):e00021

doi: 10.61474/ncs.2025.00021

Abstract

Retinal degenerative diseases (RDDs) represent a broad group of disorders characterized by the progressive loss of retinal structure and function, most notably the death of photoreceptors and retinal pigment epithelial cells. These disorders encompass hereditary conditions caused by monogenic or polygenic mutations, such as retinitis pigmentosa, Stargardt disease, and Leber congenital amaurosis, as well as non-hereditary forms caused mainly by environmental factors and aging, the most notable of which is age-related macular degeneration. RDDs are complex disorders driven by a convergence of genetic, epigenetic, inflammatory, and proteostatic mechanisms. This review explores the multifaceted molecular underpinnings of retinal degeneration, with particular emphasis on the dynamic crosstalk between DNA methylation, non-coding RNAs, oxidative stress, mitochondrial dysfunction, chronic inflammation, and protein misfolding. We also highlight the pathogenic roles of microglial activation, endoplasmic reticulum stress, and impaired autophagy, in addition to vascular and extracellular matrix disruptions. Current therapeutic strategies focus on gene editing, epigenetic modulators, antiangiogenic therapies, and stem cell transplantation, all of which have shown promise in clinical trials. However, challenges such as disease heterogeneity, limitations in preclinical models, and immune rejection continue to hinder the translation of these therapies to clinical practice. Future research must integrate single-cell multi-omics, organoid models, and artificial intelligence to better understand the cellular heterogeneity and mechanistic interplay of disease processes. This will be crucial for advancing precision medicine and developing multi-target combinatorial interventions. The objective of this review is to summarize recent advances in the molecular mechanisms of RDDs and to highlight emerging therapeutic strategies that may guide future research and clinical translation.

Keywords

Retinal degenerative diseases, Photoreceptors, Gene therapy, Cell-based therapy, Oxidative stress, Mitochondrial dysfunction, Epigenetic regulation, Non-coding RNAs, Inflammation, Srotein misfolding, Optogenetic therapy, Organoid models

Introduction

Retinal degenerative diseases (RDDs) involve the gradual deterioration of vision due to the progressive loss of retinal neurons, photoreceptors, and the underlying retinal pigment epithelium (RPE) (Fig. 1). Given the limited regenerative potential of the adult mammalian retina, the death of these cells often leads to irreversible visual deficits. Prominent RDDs include age-related macular degeneration (AMD),1 retinitis pigmentosa (RP),2 and Stargardt’s disease.3 To develop effective therapeutic approaches, significant efforts have been devoted to elucidating the genetic and molecular mechanisms underlying these pathological processes. These efforts highlight that the progression of these diseases results from complex interactions among genetic, epigenetic, metabolic, and microenvironmental factors.

Retinal structure and degeneration in retinal degenerative diseases (RDDs).
Fig. 1  Retinal structure and degeneration in retinal degenerative diseases (RDDs).

(a) Conceptual diagram of cell types and their distribution in the normal human retina. (b) Schematic representation of end-stage inherited retinal degeneration, depicting complete photoreceptor loss with relative preservation of the inner retinal layers. IRD, Inherited retinal diseases; RPE, retinal pigment epithelium.

Genetic mutations and epigenetic changes jointly disrupt retinal homeostasis, leading to progressive visual impairment. To date, more than 330 disease-causing genes have been identified in multiple RDD phenotypes (RETNET; https://retnet.org , accessed on June 28, 2025). The proper function of retinal cells relies on genes such as UBAP1L, ALG6, and IDH3G.4–6 In addition to monogenic mutations, polygenic influences play a substantial role, with the cumulative effect of multiple risk alleles modulating disease onset and progression.7–9 Epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNA regulation, add another layer of complexity.10 These heritable yet reversible changes in gene expression offer deeper insights into the dynamic epigenetic landscape of the retina. Furthermore, genetic predispositions may combine with environmental stressors such as smoking, ultraviolet radiation, and air pollution-induced oxidative stress to increase the risk of disease development.11

Oxidative stress and mitochondrial dysfunction are central to retinal degeneration.12,13 The high metabolic activity of the retina makes it susceptible to the accumulation of reactive oxygen species (ROS), which can trigger cellular damage like mitochondrial dysfunction. Mitochondrial dysfunction exacerbates ROS production, creating a feedback loop that accelerates photoreceptor cell death.13 Inflammation and immune dysregulation also play pivotal roles, with microglial activation, peripheral immune cell infiltration, and cytokine network dysregulation disrupting retinal homeostasis.14 Additionally, protein misfolding and endoplasmic reticulum (ER) stress activate the unfolded protein response (UPR), which can turn from protective to pro-apoptotic under chronic stress.15–17 Impaired autophagy and ubiquitination further contribute to the accumulation of misfolded proteins and cellular stress.18,19 Vascular abnormalities and extracellular matrix (ECM) changes are also key factors, with studies showing significant microvascular degeneration and ECM protein expression changes in RDDs.20,21 Overall, these molecular mechanisms highlight the multifactorial nature of RDDs and provide potential targets for therapeutic intervention.

In recent years, significant progress has been made in cell-based therapies, pharmacological interventions, gene therapy, and regenerative medicine. Pharmacological interventions play a crucial role in managing retinal degeneration by targeting the underlying molecular mechanisms. Major developments include the evolution of anti-vascular endothelial growth factor (anti-VEGF) agents, the investigation of repurposed drugs with neuroprotective potential, and the optimization of targeted intraocular drug delivery systems.22–24 Advances in gene therapy vectors and delivery methods have opened new possibilities for treating and potentially curing RDDs.25 Furthermore, advances in stem cell technologies and bioengineering have brought cellular therapies closer to clinical application,26–29 although challenges such as immune compatibility and functional integration still persist.30,31 Overall, these therapeutic approaches offer promising avenues for addressing previously untreatable retinal disorders.

A central objective of this review is to scrutinize the latest fundamental investigations and clinical trials related to retinal degeneration published in recent years, reflecting the field’s rapid progress. We investigated the molecular mechanisms underlying retinal degeneration (Fig. 2), assessed current and emerging treatment approaches (Fig. 3), and analyzed the obstacles to translating these therapies from the laboratory to clinical practice. This review aims to provide a comprehensive and clear overview of the current status of RDDs.

Schematic illustration of the molecular mechanisms underlying retinal degenerative diseases (RDDs).
Fig. 2  Schematic illustration of the molecular mechanisms underlying retinal degenerative diseases (RDDs).

The diagram summarizes the principal molecular pathways implicated RDDs, including genetic and epigenetic alterations, oxidative stress and mitochondrial dysfunction, dysregulated inflammation and immunity, protein misfolding with endoplasmic reticulum (ER) stress and autophagy imbalance, and vascular and extracellular matrix abnormalities. ERS, endoplasmic reticulum stress; ROS, reactive oxygen species.

Schematic representation of therapeutic strategies for retinal degenerative diseases (RDDs).
Fig. 3  Schematic representation of therapeutic strategies for retinal degenerative diseases (RDDs).

The diagram depicts the primary treatment modalities for RDDs, including pharmacological treatments, gene- and molecular-based strategies, cell therapy approaches, retinal prosthetic devices, optogenetic interventions, and complementary therapeutic measures.

Molecular mechanisms

Genetic and epigenetic factors

RDDs, including AMD and RP, are multifactorial disorders in which genetic mutations and epigenetic dysregulation jointly disrupt retinal homeostasis and drive progressive visual impairment.32,33 Extensive genetic studies have pinpointed key genes associated with these conditions. For instance, Ullah et al.6 demonstrated that biallelic loss-of-function variants in UBAP1L are implicated in human nonsyndromic retinal dystrophies. Monson et al.5 reported that the ALG6 variant is correlated with increased severity of macular cone dysfunction but milder peripheral rod involvement. Bianco et al.4 reported that IDH3G, which encodes the γ-subunit of mitochondrial isocitrate dehydrogenase and is expressed in photoreceptor inner segments, is a new candidate for X-linked RP. In addition to these monogenic mutations, polygenic influences play a substantial role in retinal degeneration. The cumulative impact of multiple risk alleles can modulate both the onset and progression of the disease, emphasizing the importance of rare and common variants alike.7–9 Gorman et al.7 combined data from the Million Veteran Program and five other cohorts to conduct pioneering multi-ancestry AMD genome-wide association studies, discovering 63 loci in total, 30 of which were novel.

Epigenetic modifications are equally significant in the pathogenic process. These are heritable yet reversible changes in gene expression that occur without altering the underlying DNA sequence. Such modifications include DNA methylation, histone modifications, and regulation by noncoding RNAs. Wahlin et al.34 initially reported irregular DNA methylation levels in the rd1 mouse model of RP. At timepoints corresponding to the peak of rod cell death, both rod and cone photoreceptors in the rd1 retina displayed heightened immunoreactivity for 5mC and 5hmC relative to wild-type controls. Recent progress in high-throughput sequencing and epigenomic profiling has provided a more profound understanding of the dynamic epigenetic landscape of the retina. Advani et al.35 carried out integrated RNA sequencing and DNA methylation array analyses on 160 human retinal samples, identifying 37,453 methylation QTLs and 13,747 DNA methylation sites that influence gene expression. Summary data-based Mendelian randomization and colocalization analyses revealed 87 target genes whose methylation and gene expression changes likely affect the genotype of AMD. Histone modifications present an extra regulatory tier by adjusting chromatin structure and swaying transcription factor access to gene promoters. For example, dysregulation of EZH2 (a catalytic component of polycomb repressive complex 2) activity is correlated with abnormal cell cycle progression and apoptosis in retinal cells. Moreover, changes in histone acetylation patterns are connected to the repression of neuroprotective genes and the aggravation of inflammatory pathways.36 Moreover, the homeostasis of non-coding RNAs is vital for the maturation and survival of different retinal cells, as well as for maintaining the normal structure and function of the retina. For example, miR-20b restrains photoreceptor cell proliferation and development and promotes apoptosis by targeting fibroblast growth factor 2 and growth factor receptor-bound protein 2 through the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway.37 MiR-210 is essential for retinal cell integrity and survival, and its absence triggers retinal degeneration.38 MiR-210 also regulates lipid metabolism by targeting acetyl-CoA synthase, thereby preventing neurodegeneration in the Drosophila retina.39 Let-7a/miR-125b,40 miR-20b/106a, and miR-204/211 play roles in promoting the maturation and differentiation of RPE cells.41

The interaction between genetic mutations and epigenetic modifications is an emerging concept of great importance. Environmental stressors, such as smoking, ultraviolet radiation, and air pollution-induced oxidative stress, can work together with genetic predispositions to increase the risk of disease onset and development.11 For example, mutations in photoreceptor-specific genes are often associated with altered methylation patterns that further inhibit gene expression, thereby accelerating degeneration.42,43 On the other hand, preclinical models have shown that epigenetic therapies aimed at reversing these maladaptive modifications hold promise, highlighting the therapeutic potential of targeting epigenetic dysregulation in retinal degeneration.44

Oxidative stress and mitochondrial dysfunction

Retinal degeneration arises from a complex interplay between oxidative stress and mitochondrial dysfunction, both of which critically compromise retinal cell survival (Fig. 4). The retina, characterized by high metabolic activity and oxygen consumption, is especially susceptible to the accumulation of ROS, which can trigger lipid peroxidation, protein oxidation, and DNA damage.13 When pathological conditions cause ROS production to exceed the capacity of the retina’s endogenous antioxidant defenses, retinal apoptosis is initiated.45 Mitochondria, which are central to both adenosine triphosphate generation via oxidative phosphorylation and ROS production, play a key role in this process.46 Under normal conditions, a small amount of electron leakage from the electron transport chain leads to low-level ROS formation.47 However, in the context of retinal degeneration, mitochondrial dysfunction—characterized by impaired electron transport, diminished adenosine triphosphate production, and increased electron leakage—significantly elevates ROS generation.48,49 Additionally, chronic oxidative stress upregulates the expression of the fission protein dynamin-related protein 1 and downregulates the fusion-related protein optic atrophy 1, leading to mitochondrial fragmentation, leading to mitochondrial fragmentation. This disruption of the mitochondrial network integrity facilitates the release of cytochrome c and other proapoptotic factors, thereby increasing the likelihood of apoptosis.50

Oxidative stress and mitochondrial dysfunction compromise retinal cell survival.
Fig. 4  Oxidative stress and mitochondrial dysfunction compromise retinal cell survival.

The conceptual diagram depicts the intricate crosstalk between oxidative stress and mitochondrial dysfunction during retinal degeneration, which collectively compromises cell survival. ATP, adenosine triphosphate; ETC, electron transport chain; mtDNA, mitochondrial DNA; ROS, reactive oxygen species.

Mitophagy, the selective removal of damaged mitochondria via autophagy, is essential for mitigating mitochondrial dysfunction.12 Under physiological conditions, mitophagy preserves mitochondrial quality and function. However, in retinal degeneration, excessive ROS and mitochondrial damage impair this protective process. As dysfunctional mitochondria accumulate, ROS production further increases, and energy metabolism becomes increasingly disrupted. These factors create a deleterious feedback loop that accelerates photoreceptor cell death.51,52 Moreover, the build-up of mitochondrial DNA (mtDNA) mutations is another key factor. Human induced pluripotent stem cell–derived RPE cells harboring mitochondrial DNA (mtDNA) mutations associated with mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes exhibit high heteroplasmy, resulting in deficits in mitochondrial function and mitophagy. These findings implicate mtDNA mutations in the disruption of mitochondrial quality control and in the promotion of AMD-like pathology.53

Oxidative stress activates various signaling cascades that amplify retinal damage. ROS can stimulate the MAPK pathway, leading to the phosphorylation of transcription factors that upregulate pro-apoptotic and pro-inflammatory genes.54,55 In parallel, oxidative stress disrupts calcium homeostasis; elevated intracellular calcium levels can trigger the opening of the mitochondrial permeability transition pore, resulting in mitochondrial swelling, depolarization, and the activation of cell death pathways.56 This calcium dysregulation further compromises mitochondrial function and increases cellular susceptibility to oxidative damage. Excessive ROS also triggers adaptive metabolic reprogramming in retinal cells. Under high oxidative conditions, cells may shift their metabolism from oxidative phosphorylation toward glycolysis and the pentose phosphate pathway to generate additional reducing equivalents such as nicotinamide adenine dinucleotide phosphate in an effort to combat ROS.48,57 Despite these compensatory shifts, persistent oxidative stress continues to compromise cellular viability, ultimately leading to irreversible retinal damage.

Inflammation and immune dysregulation

Inflammation serves as a pivotal molecular mechanism in retinal degeneration, originating from intricate interactions between resident immune cells and retinal tissue that ultimately lead to photoreceptor and RPE cell dysfunction.14 In the context of retinal degeneration, chronic inflammatory responses are characterized by abnormal microglial activation, peripheral immune cell infiltration, and cytokine network dysregulation. These factors collectively disrupt retinal homeostasis and expedite cell death.58,59 Recently, Yu et al.59 unveiled a distinctive microglial profile, marked by galectin-3 upregulation at atrophic sites in both mouse models and human AMD. These findings demonstrated that deletion of microglial galectin-3 resulted in phagocytosis defects, increased photoreceptor death, RPE damage, and vision loss, highlighting its protective role. Furthermore, Trem2 signaling was shown to direct microglial migration to atrophic sites and induce galectin-3 expression, with pharmacological Trem2 activation protecting in a galectin-3-dependent manner.59 Another study indicated that nascent RPE inflammation cascades to involve microglial activation and photoreceptor degeneration with monocyte infiltration and that inflammation drives severe, early-onset photoreceptor degeneration associated with Mertk loss of function.60

Neutrophils have been implicated in chronic retinal inflammation, according to recent research. In a retrospective case-control study, He et al.61 observed a significantly greater neutrophil-to-lymphocyte ratio (NLR) in the peripheral blood of RP patients than in that of control patients with only age-related cataracts. Moreover, the NLR was positively correlated with the degree of visual function impairment, implying that systemic neutrophil-mediated inflammation contributes to RP progression. Additionally, Fan et al.62 reported that neutrophils cocultured with the adult retinal pigment epithelial cell line-19 in a laser-induced choroidal neovascularization (CNV) mouse model markedly increased the secretion of various pro-inflammatory cytokines and induced DNA double-strand breaks, leading to S-phase arrest in RPE cells and facilitating CNV formation, thus revealing the crucial pro-inflammatory role of neutrophils in wet AMD pathogenesis. In addition to microglia and neutrophils, the RPE also plays a dual role. It supports photoreceptor function and modulates local immune responses. Under physiological conditions, the RPE serves as an immunoregulatory barrier. However, under conditions of oxidative stress and ER dysfunction, the expression of key immunomodulatory molecules such as interleukin-1 receptor-associated kinase M in the RPE decreases, resulting in increased inflammatory cytokine secretion and aggravated outer retinal degeneration.63

In addition to cellular players, complex signaling pathways integrate environmental stress signals with intrinsic immune responses. The cyclic GMP-AMP synthase (cGAS)-STING pathway, which is activated by cytosolic DNA fragments, has emerged as a critical mediator of innate immune responses. In models of light-induced retinal degeneration, aberrant cGAS-STING activation in microglia and infiltrating macrophages is correlated with elevated proinflammatory cytokine expression and accelerated photoreceptor apoptosis.64,65 Moreover, prolonged ER stress can potentiate inflammatory responses by activating nuclear factor kappa B and other transcription factors that upregulate cytokine production, linking protein misfolding with immune dysregulation in a self-perpetuating cycle.17,66

Protein misfolding, ER stress, and autophagy

High metabolic activity and rapid protein turnover render retinal neurons and RPE cells exceptionally susceptible to proteostatic disturbances. Aberrant protein folding and aggregation trigger ER stress, activating the UPR as an initial protective mechanism to restore cellular homeostasis.67 However, chronic ER stress converts sustained UPR signaling from cytoprotective to pro-apoptotic, significantly contributing to retinal degeneration in diseases such as AMD and RP.15–17 The accumulation of misfolded proteins in the ER, particularly in RPE cells, has emerged as a key molecular event in retinal degeneration. The UPR is mediated through key transducers, including inositol-requiring enzyme 1 alpha, protein kinase R-like endoplasmic reticulum kinase, and activating transcription factor 6. Recent conditional knockout studies have shown that deletion of inositol-requiring enzyme 1 alpha in rod photoreceptors does not affect early retinal development but leads to significant photoreceptor loss and functional decline in aged retinas, underscoring the importance of properly regulated ER stress signaling in maintaining retinal integrity.68 These data emphasize that while the UPR initially plays a protective role, its prolonged activation due to persistent accumulation of misfolded proteins may ultimately trigger cell death.

In addition to UPR activation, changes in autophagy, which is key for breaking down misfolded proteins and damaged cell parts, are crucial in retinal degeneration. Impaired autophagy often occurs early in disease. For example, in the rd10 mouse model of RP, clear changes in autophagy markers such as p62 and LC3 are observed even before photoreceptor degeneration is obvious. These findings suggest that defective autophagy contributes to the formation of harmful protein aggregates and exacerbates ER stress.18 Additionally, abnormal aggregation of RNA-binding proteins (RBPs) and stress granule formation are closely linked to disrupted autophagy and ER stress in the degenerating retina.18,19 Chaperone-mediated autophagy (CMA) is also vital for retinal proteostasis. Orally bioavailable small molecules selectively activate CMA in vivo by stabilizing the retinoic acid receptor α–NCOR1 complex, fine-tuning retinoic acid receptor alpha-dependent transcription.69 These activators preserve CMA activity during aging and markedly reduce photoreceptor degeneration in an RP mouse model, highlighting a novel therapeutic strategy for retinal diseases.

Ubiquitination, a critical posttranslational modification, regulates protein degradation via both the ubiquitin-proteasome system (UPS) and autophagy. Disruptions in ubiquitin-mediated proteolysis have been implicated in retinal degeneration. In such cases, impaired clearance of misfolded proteins exacerbates cellular stress and promotes neurodegenerative cascades.70 Therefore, a finely tuned balance between ubiquitination and autophagic degradation is essential for maintaining retinal protein homeostasis. Any imbalance may increase the susceptibility of retinal cells to dysfunction and death.70,71 Importantly, cellular stress responses in retinal cells are not confined solely to the ER. For example, ER stress-induced disruption of autophagy can adversely affect mitochondrial quality control, thereby further amplifying the degenerative process.72,73

Vascular abnormalities and changes in the extracellular matrix

Vascular anomalies are crucial in driving retinal degeneration. A recent study revealed that patients with advanced RP exhibit reduced macular vessel density, with notable differences observed in all four quadrants of the deep capillary plexus and three quadrants of the superficial capillary plexus.21 Another retrospective study employed optical coherence tomography angiography to investigate macular vascular abnormalities in patients with macular dystrophies and RP compared to healthy controls. The findings revealed that patients without edema presented minimal or no alterations in foveal avascular zone (FAZ). In contrast, RP patients with edema exhibited significantly reduced FAZ dimensions—both vertically and horizontally—as well as a smaller FAZ surface area in the superficial vascular complex. Meanwhile, the FAZ in the intermediate capillary complex was markedly enlarged.74 Cross-sectional research by Overbey et al.75 provided a comprehensive database for choriocapillaris flow deficit percentage (CCFD%) across dry AMD stages via swept-source optical coherence tomography angiography. The CCFD% increases with the severity of AMD, an incomplete RPE, outer retinal atrophy, and subretinal drusenoid deposits (SDDs), particularly in the early and intermediate stages, as well as with the size of RPE atrophy.75 These findings support CCFD% as a valuable clinical and research biomarker and underscore the need for longitudinal studies to confirm its prognostic value.75 Abdolrahimzadeh et al.76 demonstrated significant choriocapillaris damage in early AMD, particularly in eyes with SDDs. The central macular choriocapillaris flow area in the SDD group was significantly lower (p ≤ 0.001) than that in the healthy control group, and there was a trend toward reduced vessel density in the superficial capillary plexus and deep capillary plexus in the SDD and conventional drusen groups.76

ECM changes are key factors driving retinal degeneration. DiCesare et al.20 demonstrated that glycogen synthase kinase 3 inhibitors significantly decrease ECM protein expression in the outer retina and RPE basement membrane, reducing basal deposits and inhibiting AMD-like pathology in STZ-induced mice and in vitro RPE cells. These findings highlight the glycogen synthase kinase 3-ECM axis as a potential therapeutic target for AMD. Obasanmi et al.77 identified granzyme B (GzmB) as a novel therapeutic target for neovascular AMD (nAMD). GzmB, a serine protease, is increased in the RPE and choroidal mast cells of aging and nAMD eyes, promoting ECM degradation, inflammation, and angiogenesis. In vitro and in vivo experiments demonstrated that inhibiting GzmB or preventing mast-cell degranulation reduced choroidal angiogenesis and CNV lesions. Thus, targeting GzmB could be a new approach to suppress CNV in nAMD. Navneet et al.78 highlighted the role of elastase enzymes in AMD progression. These enzymes degrade elastin in the ECM, compromising Bruch’s membrane contributing to CNV. Elevated elastase activity was observed in both AMD models and patient cells. Treatment with A1AT, an elastase inhibitor, reduced CNV lesions and restored RPE integrity in mice, suggesting that it could modify AMD progression by stabilizing the ECM.

Lipid metabolic dysregulation

Lipid metabolism is not only the energetic foundation for maintaining retinal structure and function but also a key driver in the pathogenesis and progression of various RDDs.79,80 As one of the most metabolically active tissues in the body, retinal homeostasis relies heavily on precise metabolic regulation. Photoreceptors are particularly dependent on lipid metabolism: their outer segments contain high levels of polyunsaturated fatty acids (PUFAs), especially docosahexaenoic acid (DHA), which are crucial for membrane fluidity, visual signal transduction, and synaptic function.81 Lipid metabolism supports the continuous renewal of photoreceptor outer segment membranes, which undergo daily turnover through a coordinated cycle of synthesis by photoreceptors and phagocytosis by the RPE.82 Cholesterol, phospholipids, and sphingolipids are also tightly regulated to preserve photoreceptor integrity, intercellular signaling, and visual cycle function.83

Defects in lipid transport or cholesterol/phospholipid metabolism can result in abnormal lipid accumulation within cells and in extracellular matrices, such as Bruch’s membrane and drusen/subretinal drusenoid deposits, leading to RPE stress, complement activation, inflammatory cascades, and ultimately photoreceptor degeneration.84–86 For example, ABCA4 loss-of-function, associated with Stargardt disease, has been shown in recent lipidomic studies to cause intracellular and extracellular accumulation of A2E and retinoid intermediates in RPE cells. This is accompanied by broader lipidome remodeling and lipid droplet deposition, which exacerbates phototoxicity, oxidative stress, and RPE/photoreceptor cell death, thereby mechanistically linking visual cycle metabolism with cellular toxicity.87 Similarly, Bietti crystalline dystrophy—caused by mutations in CYP4V2, an enzyme critical for PUFA metabolism and membrane lipid turnover—leads to disrupted lipid metabolism and local crystalline deposits, driving progressive RPE and retinal atrophy.88

Photoreceptor membranes, rich in PUFAs, are highly susceptible to lipid peroxidation under conditions of high metabolic demand and light exposure. This generates reactive aldehydes such as 4-hydroxynonenal and malondialdehyde, which damage membrane proteins and mitochondria, thereby amplifying inflammatory responses.89 Concurrently, iron dyshomeostasis accelerates lipid peroxidation via Fenton chemistry, promoting ferroptosis, a lipid peroxidation-dependent mode of cell death, as a central mechanism underlying damage to the RPE and photoreceptors.90 Recent cellular and animal studies have directly linked ferroptosis to AMD and blue light/A2E-induced RPE degeneration, providing a molecular basis for targeted interventions.91

Moreover, dysregulation of specific lipid classes, such as sphingolipids and ceramides, directly impacts cell fate by inducing ER stress, mitochondrial dysfunction, and apoptosis or necrosis-like pathways.92 Tahia et al.93 demonstrated that in a BALB/c mouse model of light-induced retinal damage, systemic administration of L-cycloserine—an inhibitor of ceramide synthesis—after a 30-minute pretreatment significantly reduced pro-apoptotic gene expression, and protected photoreceptors from cell death. These findings support its potential as a novel therapeutic approach for treating RDDs.93

Therapeutic strategies

Cell-based therapies and regenerative medicine hold immense potential for the treatment of RDDs. Advances in stem cell technologies, transplantation techniques, and bioengineering approaches have brought these therapies closer to clinical application. While significant hurdles remain, ongoing research is steadily addressing key challenges related to immune compatibility, cell differentiation, and functional integration. Future developments in gene editing, biomaterials, and personalized medicine may further increase the feasibility and effectiveness of cell-based approaches for retinal regeneration.29,94,95 As research continues to refine these strategies, the goal of restoring vision for patients with currently untreatable retinal disorders is becoming increasingly attainable. Currently available treatments for RDDs are summarized in Table 1, providing an overview of pharmacological, gene-based, cellular, and device-assisted therapeutic options.

Table 1

Therapeutic strategies for retinal degenerative diseases (RDDs)

CategorySubcategoryRepresentative methods/drugs/technologiesKey findings/therapeutic effects
Pharmacological interventionsAnti-inflammatory & immunomodulationCorticosteroids (triamcinolone, dexamethasone)Effective for cystoid macular edema in RP
Anti-angiogenesisAnti-VEGF agents (bevacizumab, ranibizumab, etc.)Systematic review confirms efficacy in DME, RVO-ME, and nAMD; improves BCVA and reduces CMT
Dual-targeted therapyFaricimab (anti-VEGF + anti-Ang-2)Comparable BCVA to anti-VEGF, but better anatomical outcomes
Conventional/repurposed drugsIsopropyl unoprostone, nilvadipine, valproic acid, growth factorsIncrease retinal sensitivity or slow IRD progression
Drug repurposingMetforminAssociated with reduced AMD risk in non-diabetic patients; neuroprotective, antioxidant, mitochondrial-supportive
AntioxidantsN-acetylcysteineImproves BCVA and cone function in RP patients
Nutritional supplementsDHA, lutein, zeaxanthin, curcumin, vitamin A, zinc, manganese, saffron, safranal, coenzyme QSupport photoreceptor survival, improve ERG, increase ONL thickness
Gene therapy & molecular approachesGene replacementLuxturna (AAV2-RPE65)FDA-approved for LCA2; 65% patients show functional vision improvement
Gene silencingsiRNA/miRNA (e.g., anti-VEGF)Under investigation in AMD, glaucoma
Gene editingCRISPR/Cas9, TALENs, ZFNsCorrect RP/IRD mutations; reverse phenotypes in cells/animal models
Modifier gene therapyNr2e3, RORαEnhance transcriptional networks, retinal homeostasis, slow degeneration
Delivery systemsAAV (RGX-314, AAVv128), liposomes, nanoparticlesImprove transduction efficiency; reduce VEGF injection burden; suppress CNV
Cell-based approachesEmbryonic stem cells (ESCs)ESC-derived RPE/photoreceptorsRestore partial visual function in animals; limited by rejection and ethics
Induced pluripotent stem cells (iPSCs)iPSC-derived RPE grafts, RPE + progenitor cell co-transplantationLong-term survival and benefit in rodent, porcine, primate models
Mesenchymal stem cells (MSCs)MSC transplantation, MSC-derived exosomes/miRNAsAnti-inflammatory, maintain blood–retina barrier, promote visual recovery
Retinal prostheses & nanotechnologyElectronic prosthesesRetinal implantsRestore basic vision in late-stage RP/AMD
Nanomaterial-based prosthesesGold nanoparticle–titania nanowires, tellurium nanowire networksRestore light responses in blind mice and primates; stable long-term effects
Optogenetic therapyMicrobial opsinsChannelrhodopsins, ReaChRConfer photosensitivity to inner retinal neurons, partial vision restoration
Hybrid stimulationOptogenetics + electrical stimulationEnhanced neural responsiveness, reduced latency
Complementary therapiesElectrical stimulationTranscorneal or transorbital stimulationImprove blood flow, oxygen consumption; delay visual field loss
AcupunctureElectro-acupunctureIncrease ocular blood flow, reduce RGC injury
Physical activity & lifestyleExercise, yogaEnhance photoreceptor survival, reduce inflammation, improve quality of life
Psychosocial supportPsychological counseling, socioeconomic supportReduce anxiety/depression, improve vision-related quality of life

Pharmacological interventions

Pharmacological interventions play crucial roles in managing retinal degeneration, offering ways to slow disease progression and preserve visual function by targeting underlying molecular mechanisms. Many studies have focused on agents that address key pathogenic processes, such as abnormal angiogenesis, inflammation, oxidative stress, and neurodegeneration.

Anti-inflammatory therapies, along with immunomodulation, are critically important in managing the inflammatory processes associated with retinal degeneration. Intravitreal injections of corticosteroids such as triamcinolone and dexamethasone have proven effective in treating cystoid macular edema, which is a common complication in patients with RP.96 Similarly, anti-VEGF therapy has become a cornerstone for managing diabetic macular edema, macular edema related to retinal vein occlusion (RVO), and nAMD. The systematic review by Aldokhail et al.,97 which included 18 studies (ranging from randomized controlled trials to prospective studies, retrospective analyses, and observational studies), demonstrated that anti-VEGF therapy was effective across all three conditions. Different proportions of patients experienced improvements in best-corrected visual acuity (BCVA) and reductions in central macular thickness (CMT). Specifically, the proportion of patients with ≥15 ETDRS letters in DME ranged from 18.1% to 44.8%, whereas the mean changes in BCVA in RVO-related ME and nAMD patients were between +4.2 letters and +21.4 letters. The reduction in CMT in DME and RVO-related ME ranged from 183.1 µm to 294 µm. Pharmacological approaches also encompass dual-targeted strategies. A systematic review and meta-analysis comparing faricimab (a bispecific antibody targeting both VEGF and angiopoietin-2) with conventional anti-VEGF agents revealed that faricimab provided comparable BCVA improvements and better anatomical outcomes, such as reduced central foveal and choroidal thickness.24 Such dual-targeted approaches may yield greater benefits by simultaneously modulating angiogenic and inflammatory processes. Additionally, over the past 10–12 years, traditional medications such as the prostaglandin F2 α-agonist isopropyl unoprostone, the calcium channel blocker nilvadipine, valproic acid, and growth factors have been utilized to increase retinal sensitivity or slow the progression of inherited retinal diseases (IRDs).98

Emerging evidence advocates the repurposing of drugs with established safety profiles to protect the retina. A recent case-control study indicated that metformin, a widely used antidiabetic agent, was associated with reduced odds of developing AMD in non-diabetic patients.99 Metformin appears to have neuroprotective effects by modulating inflammatory pathways, reducing oxidative stress, and enhancing mitochondrial function. In addition, N-acetylcysteine, which is commonly used for pulmonary and psychiatric disorders, can directly scavenge free radicals, thereby reducing oxidative damage. This effect may enhance cone function and survival in RP. One study administered different doses of N-acetylcysteine (600 mg to 1,800 mg) twice daily to RP patients for 12 weeks and then three times daily for another 12 weeks, leading to a significant improvement in the mean BCVA.100

The therapeutic potential of specific nutrients for RDDs is increasingly being revealed. DHA is recognized for its critical role in retinal development and maintenance, and is thought to promote photoreceptor health because of its antioxidant properties.101 DHA levels are reduced in both mouse RP models and human RP patients.102 Lutein, a xanthophyll and the primary carotenoid that accumulates in the human macula to form macular pigment, has antioxidant properties that may benefit retinal health. Studies on animal models have investigated their protective effects against RP, yielding promising results in preventing photoreceptor degeneration. Treatment with lutein and zeaxanthin, another xanthophyll, results in larger a-wave and b-wave amplitudes in dark-adapted electroretinography (ERG), as well as larger b-wave amplitudes in light-adapted ERG.103 Moreover, lutein administration has been associated with a significant increase in outer nuclear layer thickness in mice.104 Curcumin, a bioactive compound derived from the plant turmeric (Curcuma longa), has garnered interest because of its potential multifaceted pharmacological effects, including anti-inflammatory, antioxidant, and neuroprotective properties. Curcumin suppresses the production and release of pro-inflammatory cytokines, chemokines, and enzymes involved in retinal inflammation. Preclinical studies have reported that through these mechanisms, curcumin contributes to the preservation of the retinal structure, increased thickness of the outer and inner nuclear layers, and improved ERG responses.105 Moreover, promising results have been obtained in assays involving dietary supplementation with antioxidant compounds such as vitamin A, zinc, manganese, saffron, safranal, and coenzyme Q.106,107

Gene therapy and molecular approaches

Gene therapy has ushered in a new era in the treatment and potential cure of diseases, offering hope to millions of people impacted by inherited disorders or harboring disease-causing mutations.108 The development and application of gene therapy for RDDs have consistently been at the cutting edge of translational medicine. Retinal gene therapy methods differ depending on the type of mutation and may involve gene replacement/augmentation, silencing/editing of the mutated gene, or supplying a modifier gene that influences upstream or downstream pathways from the defective gene to enhance cellular function.

Gene replacement directly supplies a functional copy of a damaged or nonfunctional gene to increase functional protein production. It is ideal for monogenic recessive inherited diseases. For example, CEP290 gene mutations are a leading cause (15–20%) of Leber congenital amaurosis.109 Voretigene neparvovec (VN), the first U.S. Food and Drug Administration (FDA)-approved gene replacement therapy marketed as Luxturna, is used to treat severe Leber congenital amaurosis type 2. In a phase III trial, 29 patients with RPE65-linked retinal dystrophy received subretinal injections of Luxturna. After one year, 65% of participants showed functional vision improvement in the multiluminance mobility test.110 Gene silencing uses small interfering RNA (siRNA) to break down sequence-specific mRNAs, eliminating the product of a faulty gene. Gene silencing with siRNAs or microRNAs targeting VEGF is being developed for AMD, glaucoma, and other ocular diseases.111–113 Several clinical trials utilizing targeted gene silencing techniques are currently underway.114–116

Gene editing involves correcting individual mutations or reducing the expression of mutated proteins in a targeted way. This technique fixes gene mutations or decreases the expression of faulty proteins to change the disease state. Several gene-editing techniques have been developed, including clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), transcription activator-like effector nucleases (TALENs), zinc finger nucleases, and meganucleases.117 Among these, CRISPR/Cas9 is the most well known and has shown potential in gene therapy. In 2016, Bakondi et al.118 first demonstrated in vivo functional ablation of an inherited dominant mutation via CRISPR/Cas9 by targeting a mutant Rho allele in a rat model of autosomal dominant RP. Recently, Chen and colleagues created a human iPS cell line (CSUASOi006-A) from an RP patient with a pre-mRNA processing factor 8 (c. C5792T) mutation.119 They used CRISPR/Cas9 to correct the c.5792C > T mutation in pre-mRNA processing factor 8 and generated an isogenic control cell line (CSUASOi006-A-2), providing a key cellular resource for RP research. Siles et al.120 precisely corrected seven hiPS cell lines from IRD patients with mutations in ABCA4, BEST1, PDE6A, PDE6C, RHO, or USH2A via CRISPR/Cas9 and TALENs. The corrected clones reversed the disease-associated phenotype in retinal cellular models,120 strengthening the study and application of gene-editing-based IRD treatments. Modifier gene therapy can affect pathways downstream or upstream of multiple defective genes, addressing clinical phenotypes without genetic diagnosis in a mutation-agnostic way. Li et al.121 found that therapy with the nuclear hormone receptor gene Nr2e3 reduced retinal degeneration. They reported an increase in photoreceptor cells, improved electroretinogram, and a molecular reset of key transcription factors and gene networks, enhancing retinal homeostasis in diseased tissue. Chang et al.122 discovered that retinoic acid-related orphan receptor α, which acts as a genetic modifier, can rescue retinal degeneration in mouse models of Stargardt disease and dry AMD.

Retinal gene therapy employs a variety of delivery vehicles, including adenovirus, adeno-associated virus (AAV), retroviral and lentiviral vectors, naked DNA/RNA, synthetic polymers, niosomes, and lipid-based carriers such as liposomes and lipid nanoparticles. The administration routes for these delivery vehicles include subtenon, subconjunctival, subretinal, and suprachoroidal ocular implants. Optimizing vector delivery methods and dosing strategies is key to the success of gene therapy. Luo et al.123 introduced a novel AAV capsid, AAVv128, which has increased transduction efficiency for photoreceptors and RPE cells and broader retinal tissue distribution in various animal models after intraocular injection. Notably, suprachoroidal delivery of the AAVv128-antiVEGF vector effectively suppressed Grade IV lesions in a laser-induced CNV NHP model of nAMD. Campochiaro et al.124 evaluated the safety and efficacy of RGX-314, an AAV8 vector expressing an anti-VEGF-A antibody fragment, which was administered via subretinal injection in nAMD patients. In this phase 1/2a trial, 42 participants received single subretinal injections of RGX-314 across multiple doses (3 × 109 to 2.5 × 1011 genome copies per eye) and were followed for 2 years. The results showed that RGX-314 was generally well tolerated, with no clinically significant immune responses. At doses ≥6 × 1010 genome copies, most patients maintain stable/improved vision and retinal thickness with few or no additional anti-VEGF injections needed.

Cell-based approaches

Cell-based therapies have come to the forefront as promising solutions to address the complex challenges of retinal degeneration. These therapies have the potential to restore lost retinal tissues or repair damaged ones, to restore vision or at least prevent further deterioration. Over the years, extensive preclinical and clinical trials have been carried out to explore the efficacy of cell therapy for RDDs. A variety of cell types, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and progenitor cell-derived cells, have been employed in these trials to treat different retinal conditions and enhance functional outcomes. Such research has been conducted across a range of animal models and has addressed diseases from RP to AMD, glaucoma, and general retinal degeneration.

One significant benefit of employing ESCs for degenerative diseases is their ability to indefinitely differentiate into virtually any cell type. Wu and colleagues effectively induced rat ESCs to differentiate into RPEs and photoreceptors, and retinal transplantation in RCS rats resulted in the restoration of visual function.125 In RCS rats, following the transplantation of human embryonic stem cell (hESC)-derived RPEs, an improvement in visual performance was observed compared to untreated controls.126 Nevertheless, although mouse ESCs integrate into mild retinal degeneration models and exhibit mature morphologies with photoreceptor markers, in severe retinal degeneration models, the transplanted cells survive but do not acquire mature morphological characteristics.127

Although ESC-based replacement therapy is highly important for retinal regeneration, its application is hindered by immune rejection, tumor formation, and ethical issues. Sharma et al.128 developed clinical-grade iPSCs from AMD patients without oncogene mutations and differentiated them into RPE patches on biodegradable scaffolds. This allows the cells to integrate into rodent and porcine models of AMD-like eye diseases.128 Furthermore, Salas et al.129 reported that a combination of hiPSC-derived RPE cells and retinal progenitor cells was more effective at preserving intrinsic photoreceptors and visual function in both early- and late-stage disease degeneration than transplanting either cell type alone. The long-term effectiveness of human iPSC-derived cells after transplantation has also been demonstrated. Human iPSC-derived retinal grafts have been shown to survive for up to five months in rats and for two years in monkey models.130,131 Nonetheless, in porcine models mimicking advanced AMD, subretinal transplantation of hiPSC-derived RPE cells was less effective in atrophic regions than in healthy areas.132

Another approach that supports autologous cell transplantation and reduces immune rejection involves the use of MSCs. Owing to their anti-inflammatory properties, growth factor-producing ability, and role in promoting tissue regeneration, MSCs are well-suited for retinal degenerative cell therapies.133 Notably, MSCs can differentiate into photoreceptor-like cells, amacrine cells, bipolar cells, and RPE cells.134–136 Recent studies have demonstrated that intravitreal injections of MSCs have protective effects on the retina and can enhance visual function.137,138 Additionally, MSCs can serve as biological patches to preserve the blood-retinal barrier, thereby promoting functional recovery following retinal ischemia/reperfusion.139 Moreover, MSC-derived exosomal miR-125b-5p has been shown to suppress retinal microvascular endothelial cell ferroptosis in diabetic retinopathy.140 Additionally, miR-125a-5p in small extracellular vesicles derived from MSCs alleviates Muller cell injury in diabetic retinopathy by modulating mitophagy through the PTP1B pathway.

Retinal prostheses and nanotechnology

Retinal prostheses aim to provide functional vision for those suffering from severe vision loss. Their effective operation depends on a posterior visual pathway that is reasonably well preserved, including the optic nerve, lateral geniculate nucleus, and visual cortex. For individuals in the advanced stages of retinal degenerative conditions such as RP and AMD, retinal implants serve as a viable option.141,142 Owing to their tunable optoelectronic characteristics, high surface-to-volume ratios, and favorable tissue compatibility, nanomaterials provide promising platforms for next-generation retinal prostheses.143 Recently, Yang et al.144 explored subretinally implanted gold nanoparticle-coated titania nanowire arrays as artificial photoreceptors. Tests in mice and monkeys with induced photoreceptor degeneration showed that arrays have advanced spatial and temporal resolution in ex vivo retinas. In blind mice, they improve visual acuity and help detect certain stimuli. In monkeys, long-term stability and positive impacts on the primary visual cortex were observed. Nie et al.145 demonstrated that intravitreal injection of anti-Thy1-conjugated near-infrared-resonant gold nanorods enables targeted photothermal activation of bipolar cells via a 20 µm-patterned near-infrared (NIR) laser scan, eliciting robust visual cortex responses in both wild-type and blind mouse models without systemic toxicity or retinal damage, thereby offering high-resolution, wide-coverage, and a minimally invasive strategy for customizable vision restoration. Wang et al.146 engineered a subretinal nanoprosthesis composed of tellurium nanowire networks (TeNWNs) capable of transducing both visible and near-infrared II light into electrical impulses. When implanted in blind mice, these TeNWNs reinstated the pupillary light reflex and supported visually guided learning under illumination at visible wavelengths and 1,550 nm. In nonhuman primate studies, TeNWNs evoked strong neural responses originating from the retina.

Optogenetic therapeutic approaches

Groundbreaking advances in optogenetics are driving the development of novel therapeutic approaches to restore vision in individuals suffering from RDDs such as RP. Optogenetics operates by leveraging light-sensitive proteins to make surviving retinal neurons responsive to light, with a primary focus on secondary and tertiary neurons in the retina.147 This allows these neurons to take over the function of the degenerated photoreceptors. Through the use of microbial opsins such as channelrhodopsins, researchers can turn inner retinal neurons into photosensitive cells. Consequently, these neurons can respond to light and restore some visual function even after photoreceptor loss has occurred. Ng et al.148 conducted a retrospective analysis of retinal structure in patients with late-stage IRD to evaluate its suitability for optogenetic gene therapy. In this study, 36 patients (54 eyes) with late-stage IRD were categorized into three groups based on clinical phenotype and history. Spectral-domain optical coherence tomography was employed to analyze structural parameters, including subfoveal thickness and individual inner layers. The results revealed that 46.3% of the degenerated retinas still retained some inner retinal layers or exhibited thickening of the inner nuclear layer. These findings suggest that cell-specific optogenetic therapy may be advantageous. In contrast, patients with unclear or disrupted inner layers might require non-cell-specific approaches that target all surviving neurons. Rohet et al.149 investigated the hybrid approach of combining optogenetic and electrical stimulation to decrease optical power and enhance the effectiveness of retinal stimulation. Compared with optogenetic stimulation alone, hybrid stimulation with a 10 µA square pulse markedly increased spiking activity and reduced latency across all light intensities in wild-type mice. Rodgers et al.150 showed that while introducing the optogenetic protein ReaChR into depolarizing (ON) bipolar cells or retinal ganglion cells (RGCs) in retinally degenerate mice restores visual responses with significant fidelity, targeting ON BCs results in more favorable outcomes, including more diverse and reproducible responses, better-preserved contrast sensitivity and temporal frequency tuning, and less disruption to the visual feature selectivity of individual RGCs than does targeting RGCs directly, thus highlighting that ON BC targeting yields a richer visual code closer to that of wild-type mice.

Complementary therapies

Electrical stimulation (ES) is a non-pharmacological method that delivers microcurrents to target tissues. These microcurrents induce biochemical effects on cells, potentially preserving or restoring vision.151,152 In a patient cohort monitored for up to one year, increased ES was linked to a trend toward preventing visual field loss and enhancing photopic ERG b-wave amplitudes, suggesting an impact on the cone photoreceptor system, which aligns with preclinical biomarker studies.153 Bittner et al.154 reported a significant improvement in retinal blood flow (RBF) in macular capillaries after six weeks of ES treatment. Half a year following transcorneal electrical stimulation (TES) therapy, there was an increase in the mean retinal arteriolar oxygen saturation compared to baseline, whereas the venular saturation decreased, indicating that TES treatment for RP results in increased oxygen consumption in the retina.155

Acupuncture, a vital element of complementary and alternative medicine, has been increasingly applied to treat a range of conditions, including pain, neuropathy, migraine, and insomnia.156 Bittner et al.154 reported that electroacupuncture significantly increased the mean flow velocity of the retrobulbar central retinal artery after two weeks and increased the RBF after one month of treatment compared with controls. Wang et al. revealed that electroacupuncture might reduce RGC injury by modulating the lncRNA-XR_002789763.1/miR-342-5p axis, activating the PINK1/Parkin pathway, and promoting Mfn2 ubiquitination.157

Physical activity is widely acknowledged for its positive impact on overall health and well-being. Preclinical studies have demonstrated that in RP mouse models, exercise exerts a beneficial effect on retinal degeneration by reducing vision loss and retinal damage. This involves an increase in the number of cone cells,158 a reduction in photoreceptor loss, and a decrease in retinal inflammation.159 In a preliminary study, physical activity was found to enhance self-reported visual function and quality of life in patients with RP.160 Additionally, Jiang and colleagues investigated the relationship between retinal microcirculatory responses and improvements in cognitive function in Parkinson’s disease patients following yoga training. Their results indicated that enhanced RBF and increased retinal capillary perfusion density in the superficial vascular plexus were associated with improved performance on the Trail-making A test. Furthermore, changes in capillary perfusion density within the retinal vascular network were linked to better scores on the Hopkins Verbal Delayed Recall test.

Among individuals with retinal dystrophies, the interplay between psychological aspects such as anxiety, fear, and decreased vision-related quality of life can aggravate the condition, establishing a negative feedback loop where psychological distress may impact the disease’s trajectory. RP and progressive visual disability can also lead to significant economic burdens on patients, affecting healthcare expenses and personal support costs and resulting in a loss of working hours, work quality, and income.161,162

Challenges

Despite notable progress in elucidating the molecular underpinnings of retinal diseases, the rapid advancement of gene therapies, cell-based interventions, and regenerative medicine for retinal degeneration has outpaced traditional regulatory frameworks. Advancing treatments for retinal degenerative disorders requires overcoming a multitude of challenges, including biological complexity, limitations in preclinical modeling, delivery barriers, ethical considerations, and the need to balance innovation with rigorous risk management. Below, we detail these obstacles and their implications for advancing therapies.

Biological complexity and patient heterogeneity

Retinal degenerative disorders exhibit profound genetic and phenotypic heterogeneity, posing a formidable challenge to any universal therapeutic approach. To date, mutations in more than 330 genes have been linked to IRDs, each giving rise to distinct clinical phenotypes and rates of progression.163,164 In addition to monogenic causes, epigenetic alterations further diversify disease mechanisms, increasing the complexity of pathogenesis.165 Furthermore, both intra- and interfamilial variability in severity and trajectory impedes the reliable prediction of treatment outcomes.166 Elhusseiny et al.167 investigated multiple affected members within a single pedigree harboring the same pathogenic PRPH2 mutation and reported marked interindividual differences in BCVA and the rate of clinical progression, despite a shared genetic background. Birch et al.168 demonstrated that the type of mutant allele (null vs. expression-modulating) was significantly correlated with phenotypic severity, even among individuals from the same family (p < 0.01).

Limitations of preclinical models

Bridging the divide between animal models and human retinal disease remains a critical translational challenge. Although rodent and porcine models have illuminated key aspects of degeneration, their differing retinal anatomy, immune milieu, and regenerative capacity limit their ability to mirror human pathology.169 Lu et al.170 demonstrated that following subretinal injection of clinical-grade human neural progenitor cells into Yucatan miniature pigs, daily intraperitoneal dexamethasone for two weeks combined with long-term oral cyclosporine A administration was required to maintain graft survival. This contrasts with murine models, where hNPCs with low MHC expression exhibit prolonged survival without extensive immunosuppression, highlighting a more robust intraocular immune rejection response in pigs toward allogeneic or xenogeneic cells. In vitro systems, particularly retinal organoids, offer controlled studies of human tissue but still fall short of replicating the in vivo microenvironment and long-term disease dynamics.171–173 Multiple studies have reported that the inner retinal layers of retinal organoids undergo progressive degeneration during long-term culture (approximately 4–6 months), characterized by a decrease in retinal ganglion cells and thinning of the inner plexiform layer.174–176 These shortcomings contribute to the high attrition rate of preclinically promising therapies in human trials, highlighting the imperative for more representative models and comprehensive translational frameworks.

Delivery and immune response challenges

The delivery of therapies to the retina is complicated by the distinct anatomical and immunological defenses of the eye. Subretinal injections and other invasive delivery methods are necessary to access retinal tissue, but they are accompanied by procedural risks and variability in therapeutic distribution.177–179 The complications associated with subretinal injection include endophthalmitis, elevated intraocular pressure, cataract formation, and vitreous or choroidal hemorrhage.180 However, owing to the limited number of treated patients and the scarcity of long-term follow-up studies, the precise incidence rates of these adverse events remain undetermined. Guest et al.181 reported that the use of different syringe types or injection techniques (e.g., prefilled syringes versus Luer-Lok syringes) can result in up to a 40% discrepancy in injection volume, thereby compromising the uniform distribution of the drug across the vitreous and retinal surfaces. Moreover, immune reactions to viral carriers or transplanted cells can erode treatment gains and provoke inflammation. Clinical variability in AAV-based gene therapies, for example, is partly driven by the host’s clearance of the vector and local immune activation.179,182 Addressing these challenges requires both a smarter vector design to evade immune detection and tailored immunomodulatory strategies to safeguard safety and sustain efficacy.

Balancing innovation with risk management

Innovation in retinal degeneration treatment must be accompanied by a comprehensive risk assessment. Strategies such as gene augmentation therapy and extracellular vesicle-based interventions show exciting potential but bring concerns about immune reactions, unintended targeting, and sustained treatment effects.183,184 In response, regulators are increasingly insisting on in-depth preclinical evidence and early-phase trial data that prove safety over prolonged follow-up periods as well as therapeutic benefit. The development of targeted gene therapies against proteins such as FAM161A highlights the need for precision in vector dose selection and gene regulation to avoid cell toxicity and immune complications.185 The key is to strike the right balance between accelerating approval pathways and enforcing stringent safety requirements. In addition, regulators must adopt a flexible approach to trial requirements, tailoring endpoints to the specific disease mechanisms under investigation while still ensuring that generated data remain rigorous and reproducible.186,187

Ethical challenges in patient safety and informed consent

Patient safety is the foremost ethical priority, as novel retinal therapies have advanced from preclinical studies to human trials. Recent data from stem cell transplantation studies have revealed serious adverse events in inadequately regulated centers, underscoring the necessity of rigorous oversight and transparent inclusion criteria.188,189 Informed consent must likewise evolve to reflect the experimental nature of these treatments, detailing potential off-target effects and the long-term consequences of genetically modified or stem cell-derived products.184,190 Haapaniemi et al.191 reported that prolonged Cas9 expression can elicit p53-dependent growth arrest, which may selectively increase p53-deficient or mutant populations and increase the potential for neoplastic transformation. Ethical considerations also demand attention to equity and access. Advanced gene and cell therapies carry high price tags, making policy interventions and innovative funding strategies essential to ensure broad availability rather than limiting benefits to a privileged few.192,193

Limitations

Despite our efforts to provide a comprehensive overview, this review has several limitations that should be acknowledged. First, as a narrative review, the selection of literature may be subject to bias, and it is possible that not all relevant studies were included. Second, the field of retinal degeneration is highly heterogeneous, encompassing both monogenic inherited retinal dystrophies and multifactorial diseases such as AMD. Given this complexity, certain mechanisms and therapeutic approaches have been emphasized more than others. Third, owing to space constraints, some rapidly evolving areas, including advanced biomaterials, combinatorial nanomedicine strategies, and digital health applications, can only be briefly discussed. Finally, while this review highlights emerging therapeutic strategies, many of these approaches are still in preclinical or early clinical trial stages, and their long-term safety and efficacy remain uncertain.

Conclusions

RDDs pose a significant threat to global vision health, arising from a multifaceted network of genetic and epigenetic alterations, oxidative stress, mitochondrial dysfunction, inflammation, immune dysregulation, protein misfolding, ER stress, impaired autophagy, vascular abnormalities, and ECM disruption. To date, pharmacological treatments, gene- and molecular-based approaches, retinal prostheses, cell-based approaches, optogenetics, TES, and other strategies have all shown promise in clinical trials. Progress in elucidating these molecular mechanisms and advancing therapeutic strategies continues to deepen our understanding of RDDs and holds promise for improving patient outcomes and quality of life. Nevertheless, to realize this objective, several barriers must be surmounted, such as disease heterogeneity, the limitations of preclinical models, and the problem of immune rejection.

Declarations

Acknowledgement

None.

Funding

This work was supported by the Natural Science Foundation of Zhejiang Province (LQN25H120010, LTGD23H120002); the National Natural Science Foundation of China (82501339, 82101176); the Health Technology Plan Project in Zhejiang Province (2023KY151); and the Science and Technology Project of Wenzhou (Y20220774).

Conflict of interest

The authors have no conflicts of interest to declare.

Authors’ contributions

Article search (YYZ, JYL, JWL), data analysis and interpretation (YYZ), funding acquisition (YYZ, CC, XTL), manuscript writing (YYZ), critical revision of the manuscript (JYL, JWL, CC, XTL), project administration (CC, XTL), technical and material support (CC, XTL). All authors have made substantial contributions to this study and have approved the final manuscript.

References

  1. Fleckenstein M, Keenan TDL, Guymer RH, Chakravarthy U, Schmitz-Valckenberg S, Klaver CC, et al. Age-related macular degeneration. Nat Rev Dis Primers 2021;7(1):31 View Article PubMed/NCBI
  2. Hamel C. Retinitis pigmentosa. Orphanet J Rare Dis 2006;1:40 View Article PubMed/NCBI
  3. Tanna P, Strauss RW, Fujinami K, Michaelides M. Stargardt disease: clinical features, molecular genetics, animal models and therapeutic options. Br J Ophthalmol 2017;101(1):25-30 View Article PubMed/NCBI
  4. Bianco L, Navarro J, Michiels C, Sangermano R, Condroyer C, Antonio A, et al. Identification of IDH3G, encoding the gamma subunit of mitochondrial isocitrate dehydrogenase, as a novel candidate gene for X-linked retinitis pigmentosa. Genet Med 2025;27(6):101418 View Article PubMed/NCBI
  5. Monson E, Cideciyan AV, Roman AJ, Sumaroka A, Swider M, Wu V, et al. Inherited Retinal Degeneration Caused by Dehydrodolichyl Diphosphate Synthase Mutation-Effect of an ALG6 Modifier Variant. Int J Mol Sci 2024;25(2):1004 View Article PubMed/NCBI
  6. Ullah E, Lin S, Lu J, Bender C, Webster AR, Malka S, et al. Biallelic Loss-of-Function Variants in UBAP1L and Nonsyndromic Retinal Dystrophies. JAMA Ophthalmol 2024;142(11):1081-1086 View Article PubMed/NCBI
  7. Gorman BR, Voloudakis G, Igo RP, Kinzy T, Halladay CW, Bigdeli TB, et al. Genome-wide association analyses identify distinct genetic architectures for age-related macular degeneration across ancestries. Nat Genet 2024;56(12):2659-2671 View Article PubMed/NCBI
  8. Seddon JM, Silver RE, Kwong M, Rosner B. Risk Prediction for Progression of Macular Degeneration: 10 Common and Rare Genetic Variants, Demographic, Environmental, and Macular Covariates. Invest Ophthalmol Vis Sci 2015;56(4):2192-2202 View Article PubMed/NCBI
  9. Stradiotto E, Allegrini D, Fossati G, Raimondi R, Sorrentino T, Tripepi D, et al. Genetic Aspects of Age-Related Macular Degeneration and Their Therapeutic Potential. Int J Mol Sci 2022;23(21):13280 View Article PubMed/NCBI
  10. Rajanala K, Upadhyay A. Epigenetic Switches in Retinal Homeostasis and Target for Drug Development. Int J Mol Sci 2024;25(5):2840 View Article PubMed/NCBI
  11. Maurya M, Bora K, Blomfield AK, Pavlovich MC, Huang S, Liu CH, et al. Oxidative stress in retinal pigment epithelium degeneration: from pathogenesis to therapeutic targets in dry age-related macular degeneration. Neural Regen Res 2023;18(10):2173-2181 View Article PubMed/NCBI
  12. Onishi M, Yamano K, Sato M, Matsuda N, Okamoto K. Molecular mechanisms and physiological functions of mitophagy. EMBO J 2021;40(3):e104705 View Article PubMed/NCBI
  13. Wang J, Li M, Geng Z, Khattak S, Ji X, Wu D, et al. Role of Oxidative Stress in Retinal Disease and the Early Intervention Strategies: A Review. Oxid Med Cell Longev 2022;2022:7836828 View Article PubMed/NCBI
  14. Kaur G, Singh NK. The Role of Inflammation in Retinal Neurodegeneration and Degenerative Diseases. Int J Mol Sci 2021;23(1):386 View Article PubMed/NCBI
  15. Kaarniranta K, Blasiak J, Liton P, Boulton M, Klionsky DJ, Sinha D. Autophagy in age-related macular degeneration. Autophagy 2023;19(2):388-400 View Article PubMed/NCBI
  16. Kang MJ, Chung J, Ryoo HD. CDK5 and MEKK1 mediate pro-apoptotic signalling following endoplasmic reticulum stress in an autosomal dominant retinitis pigmentosa model. Nat Cell Biol 2012;14(4):409-415 View Article PubMed/NCBI
  17. Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, et al. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024;98:101231 View Article PubMed/NCBI
  18. Yamoah A, Tripathi P, Guo H, Scheve L, Walter P, Johnen S, et al. Early Alterations of RNA Binding Protein (RBP) Homeostasis and ER Stress-Mediated Autophagy Contributes to Progressive Retinal Degeneration in the rd10 Mouse Model of Retinitis Pigmentosa (RP). Cells 2023;12(7):1094 View Article PubMed/NCBI
  19. Yuan L, Mao LH, Huang YY, Outeiro TF, Li W, Vieira TCRG, et al. Stress granules: emerging players in neurodegenerative diseases. Transl Neurodegener 2025;14(1):22 View Article PubMed/NCBI
  20. DiCesare SM, Ortega AJ, Collier GE, Daniel S, Thompson KN, McCoy MK, et al. GSK3 inhibition reduces ECM production and prevents age-related macular degeneration-like pathology. JCI Insight 2024;9(15):e178050 View Article PubMed/NCBI
  21. Duch Hurtado M, Vidal Oliver L, Marín Lambies C, Salom Alonso D. Microvascular quantitative metrics in retinitis pigmentosa using optical coherence tomography angiography. Arch Soc Esp Oftalmol (Engl Ed) 2023;98(5):270-275 View Article PubMed/NCBI
  22. Matsumoto H, Hoshino J, Nakamura K, Akiyama H. One-year results of treat-and-extend regimen with intravitreal faricimab for treatment-naïve neovascular age-related macular degeneration. Jpn J Ophthalmol 2024;68(2):83-90 View Article PubMed/NCBI
  23. Thangamathesvaran L, Kong J, Bressler SB, Singh M, Wenick AS, Scott AW, et al. Severe Intraocular Inflammation Following Intravitreal Faricimab. JAMA Ophthalmol 2024;142(4):365-370 View Article PubMed/NCBI
  24. Yen WT, Wu CS, Yang CH, Chen YH, Lee CH, Hsu CR. Efficacy and safety of intravitreal faricimab for neovascular age-related macular degeneration: a systematic review and meta-analysis. Sci Rep 2024;14(1):2485 View Article PubMed/NCBI
  25. Drag S, Dotiwala F, Upadhyay AK. Gene Therapy for Retinal Degenerative Diseases: Progress, Challenges, and Future Directions. Invest Ophthalmol Vis Sci 2023;64(7):39 View Article PubMed/NCBI
  26. Ampie L, McGavern DB. Immunological defense of CNS barriers against infections. Immunity 2022;55(5):781-799 View Article PubMed/NCBI
  27. Hirami Y, Mandai M, Sugita S, Maeda A, Maeda T, Yamamoto M, et al. Safety and stable survival of stem-cell-derived retinal organoid for 2 years in patients with retinitis pigmentosa. Cell Stem Cell 2023;30(12):1585-1596.e6 View Article PubMed/NCBI
  28. Liu H, Lu S, Chen M, Gao N, Yang Y, Hu H, et al. Towards Stem/Progenitor Cell-Based Therapies for Retinal Degeneration. Stem Cell Rev Rep 2024;20(6):1459-1479 View Article PubMed/NCBI
  29. Maeda T, Mandai M, Sugita S, Kime C, Takahashi M. Strategies of pluripotent stem cell-based therapy for retinal degeneration: update and challenges. Trends Mol Med 2022;28(5):388-404 View Article PubMed/NCBI
  30. Radu M, Brănișteanu DC, Pirvulescu RA, Dumitrescu OM, Ionescu MA, Zemba M. Exploring Stem-Cell-Based Therapies for Retinal Regeneration. Life (Basel) 2024;14(6):668 View Article PubMed/NCBI
  31. Wu KY, Dhaliwal JK, Sasitharan A, Kalevar A. Cell Therapy for Retinal Degenerative Diseases: Progress and Prospects. Pharmaceutics 2024;16(10):1299 View Article PubMed/NCBI
  32. Ali MU, Rahman MSU, Cao J, Yuan PX. Genetic characterization and disease mechanism of retinitis pigmentosa; current scenario. 3 Biotech 2017;7(4):251 View Article PubMed/NCBI
  33. Fritsche LG, Igl W, Bailey JN, Grassmann F, Sengupta S, Bragg-Gresham JL, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet 2016;48(2):134-143 View Article PubMed/NCBI
  34. Wahlin KJ, Enke RA, Fuller JA, Kalesnykas G, Zack DJ, Merbs SL. Epigenetics and cell death: DNA hypermethylation in programmed retinal cell death. PLoS One 2013;8(11):e79140 View Article PubMed/NCBI
  35. Advani J, Mehta PA, Hamel AR, Mehrotra S, Kiel C, Strunz T, et al. QTL mapping of human retina DNA methylation identifies 87 gene-epigenome interactions in age-related macular degeneration. Nat Commun 2024;15(1):1972 View Article PubMed/NCBI
  36. Peng Y, Bui CH, Zhang XJ, Chen JS, Tham CC, Chu WK, et al. The role of EZH2 in ocular diseases: a narrative review. Epigenomics 2023;15(9):557-570 View Article PubMed/NCBI
  37. Zhang X, Zhang Q, Jiang Y, Zhang S, Hong Q, Guo X, et al. Expression and significance of miR - 20b in retinal photoreceptor cells exposed to PCB(1254). Aging (Albany NY) 2019;11(20):8969-8981 View Article PubMed/NCBI
  38. Weigelt CM, Hahn O, Arlt K, Gruhn M, Jahn AJ, Eßer J, et al. Loss of miR-210 leads to progressive retinal degeneration in Drosophila melanogaster. Life Sci Alliance 2019;2(1):e201800149 View Article PubMed/NCBI
  39. Lyu J, Chen Y, Yang W, Guo T, Xu X, Xi Y, et al. The conserved microRNA miR-210 regulates lipid metabolism and photoreceptor maintenance in the Drosophila retina. Cell Death Differ 2021;28(2):764-779 View Article PubMed/NCBI
  40. Shahriari F, Satarian L, Moradi S, Zarchi AS, Günther S, Kamal A, et al. MicroRNA profiling reveals important functions of miR-125b and let-7a during human retinal pigment epithelial cell differentiation. Exp Eye Res 2020;190:107883 View Article PubMed/NCBI
  41. Ohana R, Weiman-Kelman B, Raviv S, Tamm ER, Pasmanik-Chor M, Rinon A, et al. MicroRNAs are essential for differentiation of the retinal pigmented epithelium and maturation of adjacent photoreceptors. Development 2015;142(14):2487-2498 View Article PubMed/NCBI
  42. Moore SM, Christoforidis JB. Advances in Ophthalmic Epigenetics and Implications for Epigenetic Therapies: A Review. Genes (Basel) 2023;14(2):417 View Article PubMed/NCBI
  43. Rao RC, Hennig AK, Malik MT, Chen DF, Chen S. Epigenetic regulation of retinal development and disease. J Ocul Biol Dis Infor 2011;4(3):121-136 View Article PubMed/NCBI
  44. Barnstable CJ. Epigenetics and Degenerative Retinal Diseases: Prospects for New Therapeutic Approaches. Asia Pac J Ophthalmol (Phila) 2022;11(4):328-334 View Article PubMed/NCBI
  45. Cecilia OM, José Alberto CG, José NP, Ernesto Germán CM, Ana Karen LC, Luis Miguel RP, et al. Oxidative Stress as the Main Target in Diabetic Retinopathy Pathophysiology. J Diabetes Res 2019;2019:8562408 View Article PubMed/NCBI
  46. Nolfi-Donegan D, Braganza A, Shiva S. Mitochondrial electron transport chain: Oxidative phosphorylation, oxidant production, and methods of measurement. Redox Biol 2020;37:101674 View Article PubMed/NCBI
  47. Zhao RZ, Jiang S, Zhang L, Yu ZB. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int J Mol Med 2019;44(1):3-15 View Article PubMed/NCBI
  48. Kanan Y, Hackett SF, Taneja K, Khan M, Campochiaro PA. Oxidative stress-induced alterations in retinal glucose metabolism in Retinitis Pigmentosa. Free Radic Biol Med 2022;181:143-153 View Article PubMed/NCBI
  49. Sreekumar PG, Ferrington DA, Kannan R. Glutathione Metabolism and the Novel Role of Mitochondrial GSH in Retinal Degeneration. Antioxidants (Basel) 2021;10(5):661 View Article PubMed/NCBI
  50. Wang L, Yu X, Zhang D, Wen Y, Zhang L, Xia Y, et al. Long-term blue light exposure impairs mitochondrial dynamics in the retina in light-induced retinal degeneration in vivo and in vitro. J Photochem Photobiol B 2023;240:112654 View Article PubMed/NCBI
  51. Menger KE, Logan A, Luhmann UFO, Smith AJ, Wright AF, Ali RR, et al. In vivo measurement of mitochondrial ROS production in mouse models of photoreceptor degeneration. Redox Biochem Chem 2023;5–6:100007 View Article PubMed/NCBI
  52. Zou GP, Wang T, Xiao JX, Wang XY, Jiang LP, Tou FF, et al. Lactate protects against oxidative stress-induced retinal degeneration by activating autophagy. Free Radic Biol Med 2023;194:209-219 View Article PubMed/NCBI
  53. Bhattacharya S, Yin J, Huo W, Chaum E. Modeling of mitochondrial bioenergetics and autophagy impairment in MELAS-mutant iPSC-derived retinal pigment epithelial cells. Stem Cell Res Ther 2022;13(1):260 View Article PubMed/NCBI
  54. Averill-Bates D. Reactive oxygen species and cell signaling. Review. Biochim Biophys Acta Mol Cell Res 2024;1871(2):119573 View Article PubMed/NCBI
  55. Son Y, Cheong YK, Kim NH, Chung HT, Kang DG, Pae HO. Mitogen-Activated Protein Kinases and Reactive Oxygen Species: How Can ROS Activate MAPK Pathways?. J Signal Transduct 2011;2011:792639 View Article PubMed/NCBI
  56. Glover HL, Schreiner A, Dewson G, Tait SWG. Mitochondria and cell death. Nat Cell Biol 2024;26(9):1434-1446 View Article PubMed/NCBI
  57. Grant CM. Metabolic reconfiguration is a regulated response to oxidative stress. J Biol 2008;7(1):1 View Article PubMed/NCBI
  58. Kaur G, Singh NK. Inflammation and retinal degenerative diseases. Neural Regen Res 2023;18(3):513-518 View Article PubMed/NCBI
  59. Yu C, Lad EM, Mathew R, Shiraki N, Littleton S, Chen Y, et al. Microglia at sites of atrophy restrict the progression of retinal degeneration via galectin-3 and Trem2. J Exp Med 2024;221(3):e20231011 View Article PubMed/NCBI
  60. Mercau ME, Akalu YT, Mazzoni F, Gyimesi G, Alberto EJ, Kong Y, et al. Inflammation of the retinal pigment epithelium drives early-onset photoreceptor degeneration in Mertk-associated retinitis pigmentosa. Sci Adv 2023;9(3):eade9459 View Article PubMed/NCBI
  61. He M, Wu T, Zhang L, Ye W, Ma J, Zhao C, et al. Correlation between neutrophil-to-lymphocyte ratio and clinical manifestations and complications of retinitis pigmentosa. Acta Ophthalmol 2022;100(1):e278-e287 View Article PubMed/NCBI
  62. Fan Q, Song X, Li M, Xu Q, Yan C, Li H, et al. Neutrophils promote laser-induced choroidal neovascularization by increasing pro-inflammatory cytokines secretion and cell cycle arrest in retinal pigment epithelium. Int Immunopharmacol 2025;145:113735 View Article PubMed/NCBI
  63. Liu J, Copland DA, Clare AJ, Gorski M, Richards BT, Scott L, et al. Replenishing IRAK-M expression in retinal pigment epithelium attenuates outer retinal degeneration. Sci Transl Med 2024;16(750):eadi4125 View Article PubMed/NCBI
  64. Zhu X, Liu W, Tang X, Chen Y, Ge X, Ke Q, et al. The BET PROTAC inhibitor dBET6 protects against retinal degeneration and inhibits the cGAS-STING in response to light damage. J Neuroinflammation 2023;20(1):119 View Article PubMed/NCBI
  65. Zou M, Ke Q, Nie Q, Qi R, Zhu X, Liu W, et al. Inhibition of cGAS-STING by JQ1 alleviates oxidative stress-induced retina inflammation and degeneration. Cell Death Differ 2022;29(9):1816-1833 View Article PubMed/NCBI
  66. McLaughlin T, Medina A, Perkins J, Yera M, Wang JJ, Zhang SX. Cellular stress signaling and the unfolded protein response in retinal degeneration: mechanisms and therapeutic implications. Mol Neurodegener 2022;17(1):25 View Article PubMed/NCBI
  67. Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol 2020;21(8):421-438 View Article PubMed/NCBI
  68. Massoudi D, Gorman S, Kuo YM, Iwawaki T, Oakes SA, Papa FR, et al. Deletion of the Unfolded Protein Response Transducer IRE1α Is Detrimental to Aging Photoreceptors and to ER Stress-Mediated Retinal Degeneration. Invest Ophthalmol Vis Sci 2023;64(4):30 View Article PubMed/NCBI
  69. Gomez-Sintes R, Xin Q, Jimenez-Loygorri JI, McCabe M, Diaz A, Garner TP, et al. Targeting retinoic acid receptor alpha-corepressor interaction activates chaperone-mediated autophagy and protects against retinal degeneration. Nat Commun 2022;13(1):4220 View Article PubMed/NCBI
  70. Wei J, Chen X, Xiong Y, Gao Y. Advances in Ubiquitination and Proteostasis in Retinal Degeneration. Front Biosci (Landmark Ed) 2024;29(7):260 View Article PubMed/NCBI
  71. Lei H, Xu H, Wu Y. Role of UCHL3 in health and disease. Biochem Biophys Res Commun 2024;734:150626 View Article PubMed/NCBI
  72. Newton F, Halachev M, Nguyen L, McKie L, Mill P, Megaw R. Autophagy disruption and mitochondrial stress precede photoreceptor necroptosis in multiple mouse models of inherited retinal disorders. Nat Commun 2025;16(1):4024 View Article PubMed/NCBI
  73. Zielke S, Kardo S, Zein L, Mari M, Covarrubias-Pinto A, Kinzler MN, et al. ATF4 links ER stress with reticulophagy in glioblastoma cells. Autophagy 2021;17(9):2432-2448 View Article PubMed/NCBI
  74. Deutsch S, Lommatzsch A, Weinitz S, Farmand G, Kellner U. Optical coherence tomography angiography (OCT-A) in retinitis pigmentosa and macular dystrophy patients: a retrospective study. Graefes Arch Clin Exp Ophthalmol 2022;260(6):1923-1931 View Article PubMed/NCBI
  75. Overbey K, Romano F, Ding X, Bennett CF, Stettler I, Garg I, et al. Choriocapillaris impairment in dry AMD: insights from swept-source OCT angiography and associations with structural biomarkers. Br J Ophthalmol 2025;109(9):1020-1027 View Article PubMed/NCBI
  76. Abdolrahimzadeh S, Zweifel SA, Di Pippo M, Bajka A, Scuderi G, Lotery AJ. Central macular choriocapillaris impairment as a manifestation of microvascular disease in eyes with subretinal drusenoid deposits. Eye (Lond) 2024;38(1):173-178 View Article PubMed/NCBI
  77. Obasanmi G, Uppal M, Cui JZ, Xi J, Ju MJ, Song J, et al. Granzyme B degrades extracellular matrix and promotes inflammation and choroidal neovascularization. Angiogenesis 2024;27(3):351-373 View Article PubMed/NCBI
  78. Navneet S, Brandon C, Simpson K, Rohrer B. Exploring the Therapeutic Potential of Elastase Inhibition in Age-Related Macular Degeneration in Mouse and Human. Cells 2023;12(9):1308 View Article PubMed/NCBI
  79. Chen Y, Coorey NJ, Zhang M, Zeng S, Madigan MC, Zhang X, et al. Metabolism Dysregulation in Retinal Diseases and Related Therapies. Antioxidants (Basel) 2022;11(5):942 View Article PubMed/NCBI
  80. Wu BX, Fan J, Boyer NP, Jenkins RW, Koutalos Y, Hannun YA, et al. Lack of Acid Sphingomyelinase Induces Age-Related Retinal Degeneration. PLoS One 2015;10(7):e0133032 View Article PubMed/NCBI
  81. Gabrielle PH. Lipid metabolism and retinal diseases. Acta Ophthalmol 2022;100(Suppl 269):3-43 View Article PubMed/NCBI
  82. Lewandowski D, Sander CL, Tworak A, Gao F, Xu Q, Skowronska-Krawczyk D. Dynamic lipid turnover in photoreceptors and retinal pigment epithelium throughout life. Prog Retin Eye Res 2022;89:101037 View Article PubMed/NCBI
  83. van Leeuwen EM, Emri E, Merle BMJ, Colijn JM, Kersten E, Cougnard-Gregoire A, et al. A new perspective on lipid research in age-related macular degeneration. Prog Retin Eye Res 2018;67:56-86 View Article PubMed/NCBI
  84. Kaur G, Tan LX, Rathnasamy G, La Cunza N, Germer CJ, Toops KA, et al. Aberrant early endosome biogenesis mediates complement activation in the retinal pigment epithelium in models of macular degeneration. Proc Natl Acad Sci U S A 2018;115(36):9014-9019 View Article PubMed/NCBI
  85. Storti F, Klee K, Todorova V, Steiner R, Othman A, van der Velde-Visser S, et al. Impaired ABCA1/ABCG1-mediated lipid efflux in the mouse retinal pigment epithelium (RPE) leads to retinal degeneration. Elife 2019;8:e45100 View Article PubMed/NCBI
  86. Toops KA, Tan LX, Jiang Z, Radu RA, Lakkaraju A. Cholesterol-mediated activation of acid sphingomyelinase disrupts autophagy in the retinal pigment epithelium. Mol Biol Cell 2015;26(1):1-14 View Article PubMed/NCBI
  87. Farnoodian M, Bose D, Barone F, Nelson LM, Boyle M, Jun B, et al. Retina and RPE lipid profile changes linked with ABCA4 associated Stargardt’s maculopathy. Pharmacol Ther 2023;249:108482 View Article PubMed/NCBI
  88. García-García GP, Martínez-Rubio M, Moya-Moya MA, Pérez-Santonja JJ, Escribano J. Current perspectives in Bietti crystalline dystrophy. Clin Ophthalmol 2019;13:1379-1399 View Article PubMed/NCBI
  89. Zhao T, Guo X, Sun Y. Iron Accumulation and Lipid Peroxidation in the Aging Retina: Implication of Ferroptosis in Age-Related Macular Degeneration. Aging Dis 2021;12(2):529-551 View Article PubMed/NCBI
  90. Henning Y, Blind US, Larafa S, Matschke J, Fandrey J. Hypoxia aggravates ferroptosis in RPE cells by promoting the Fenton reaction. Cell Death Dis 2022;13(7):662 View Article PubMed/NCBI
  91. Huang S, Sun Y, Yu X, Ren X, Wang L, Sun Y, et al. Ferroptosis in ocular diseases: mechanisms, crosstalk with other cell death pathways, and therapeutic prospects. Front Med (Lausanne) 2025;12:1608975 View Article PubMed/NCBI
  92. Law BA, Liao X, Moore KS, Southard A, Roddy P, Ji R, et al. Lipotoxic very-long-chain ceramides cause mitochondrial dysfunction, oxidative stress, and cell death in cardiomyocytes. FASEB J 2018;32(3):1403-1416 View Article PubMed/NCBI
  93. Tahia F, Ma D, Stephenson DJ, Basu SK, Del Mar NA, Lenchik N, et al. Inhibiting De Novo Biosynthesis of Ceramide by L-Cycloserine Can Prevent Light-Induced Retinal Degeneration in Albino BALB/c Mice. Int J Mol Sci 2024;25(24):13389 View Article PubMed/NCBI
  94. Niu Y, Ji J, Yao K, Fu Q. Regenerative treatment of ophthalmic diseases with stem cells: Principles, progress, and challenges. Adv Ophthalmol Pract Res 2024;4(2):52-64 View Article PubMed/NCBI
  95. Van Gelder RN, Chiang MF, Dyer MA, Greenwell TN, Levin LA, Wong RO, et al. Regenerative and restorative medicine for eye disease. Nat Med 2022;28(6):1149-1156 View Article PubMed/NCBI
  96. Yang Y, Bailey C, Loewenstein A, Massin P. INTRAVITREAL CORTICOSTEROIDS IN DIABETIC MACULAR EDEMA: PHARMACOKINETIC CONSIDERATIONS. Retina 2015;35(12):2440-2449 View Article PubMed/NCBI
  97. Aldokhail LS, Alhadlaq AM, Alaradi LM, Alaradi LM, AlShaikh FY. Outcomes of Anti-VEGF Therapy in Eyes with Diabetic Macular Edema, Vein Occlusion-Related Macular Edema, and Neovascular Age-Related Macular Degeneration: A Systematic Review. Clin Ophthalmol 2024;18:3837-3851 View Article PubMed/NCBI
  98. Colombo L, Baldesi J, Martella S, Quisisana C, Antico A, Mapelli L, et al. Managing Retinitis Pigmentosa: A Literature Review of Current Non-Surgical Approaches. J Clin Med 2025;14(2):330 View Article PubMed/NCBI
  99. Aggarwal S, Moir J, Hyman MJ, Kaufmann GT, Flores A, Hariprasad SM, et al. Metformin Use and Age-Related Macular Degeneration in Patients Without Diabetes. JAMA Ophthalmol 2024;142(1):53-57 View Article PubMed/NCBI
  100. Russell S, Bennett J, Wellman JA, Chung DC, Yu ZF, Tillman A, 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 2017;390(10097):849-860 View Article PubMed/NCBI
  101. Bazan NG. Cell survival matters: docosahexaenoic acid signaling, neuroprotection and photoreceptors. Trends Neurosci 2006;29(5):263-271 View Article PubMed/NCBI
  102. Ruiz-Pastor MJ, Kutsyr O, Lax P, Cuenca N. Decrease in DHA and other fatty acids correlates with photoreceptor degeneration in retinitis pigmentosa. Exp Eye Res 2021;209:108667 View Article PubMed/NCBI
  103. Yu M, Yan W, Beight C. Lutein and Zeaxanthin Isomers Reduce Photoreceptor Degeneration in the Pde6b (rd10) Mouse Model of Retinitis Pigmentosa. Biomed Res Int 2018;2018:4374087 View Article PubMed/NCBI
  104. Zhang HJ, Liu XB, Chen XM, Kong QH, Liu YS, So KF, et al. Lutein delays photoreceptor degeneration in a mouse model of retinitis pigmentosa. Neural Regen Res 2022;17(7):1596-1603 View Article PubMed/NCBI
  105. Franzone F, Nebbioso M, Pergolizzi T, Attanasio G, Musacchio A, Greco A, et al. Anti-inflammatory role of curcumin in retinal disorders (Review). Exp Ther Med 2021;22(1):790 View Article PubMed/NCBI
  106. Cuenca N, Fernández-Sánchez L, Campello L, Maneu V, De la Villa P, Lax P, et al. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog Retin Eye Res 2014;43:17-75 View Article PubMed/NCBI
  107. Newton F, Megaw R. Mechanisms of Photoreceptor Death in Retinitis Pigmentosa. Genes (Basel) 2020;11(10):1120 View Article PubMed/NCBI
  108. Hanany M, Rivolta C, Sharon D. Worldwide carrier frequency and genetic prevalence of autosomal recessive inherited retinal diseases. Proc Natl Acad Sci U S A 2020;117(5):2710-2716 View Article PubMed/NCBI
  109. Kumaran N, Moore AT, Weleber RG, Michaelides M. Leber congenital amaurosis/early-onset severe retinal dystrophy: clinical features, molecular genetics and therapeutic interventions. Br J Ophthalmol 2017;101(9):1147-1154 View Article PubMed/NCBI
  110. Dhurandhar D, Sahoo NK, Mariappan I, Narayanan R. Gene therapy in retinal diseases: A review. Indian J Ophthalmol 2021;69(9):2257-2265 View Article PubMed/NCBI
  111. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001;411(6836):494-498 View Article PubMed/NCBI
  112. Jiang J, Zhang X, Tang Y, Li S, Chen J. Progress on ocular siRNA gene-silencing therapy and drug delivery systems. Fundam Clin Pharmacol 2021;35(1):4-24 View Article PubMed/NCBI
  113. Saw PE, Song EW. siRNA therapeutics: a clinical reality. Sci China Life Sci 2020;63(4):485-500 View Article PubMed/NCBI
  114. Hoy SM. Patisiran: First Global Approval. Drugs 2018;78(15):1625-1631 View Article PubMed/NCBI
  115. Padda IS, Mahtani AU, Patel P, Parmar M. Small Interfering RNA (siRNA) Therapy. StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025
  116. Salminen A, Kauppinen A, Hyttinen JM, Toropainen E, Kaarniranta K. Endoplasmic reticulum stress in age-related macular degeneration: trigger for neovascularization. Mol Med 2010;16(11-12):535-542 View Article PubMed/NCBI
  117. Gaj T, Sirk SJ, Shui SL, Liu J. Genome-Editing Technologies: Principles and Applications. Cold Spring Harb Perspect Biol 2016;8(12):a023754 View Article PubMed/NCBI
  118. Bakondi B, Lv W, Lu B, Jones MK, Tsai Y, Kim KJ, et al. In vivo CRISPR/Cas9 gene editing corrects retinal dystrophy in the S334ter-3 rat model of autosomal dominant retinitis pigmentosa. Mol Ther 2016;24(3):556-563 View Article PubMed/NCBI
  119. Chen H, Liang Y, Liang Y, Chen Y, Li X, Zhang R, et al. Generation of a gene-corrected isogenic human iPS cell line (CSUASOi006-A-2) from a retinitis pigmentosa patient using CRISPR/Cas9 technology. Stem Cell Res 2025;86:103727 View Article PubMed/NCBI
  120. Siles L, Pomares E. Rescue of the disease-associated phenotype in CRISPR-corrected hiPSCs as a therapeutic approach for inherited retinal dystrophies. Mol Ther Nucleic Acids 2025;36(1):102482 View Article PubMed/NCBI
  121. Li S, Datta S, Brabbit E, Love Z, Woytowicz V, Flattery K, et al. Nr2e3 is a genetic modifier that rescues retinal degeneration and promotes homeostasis in multiple models of retinitis pigmentosa. Gene Ther 2021;28(5):223-241 View Article PubMed/NCBI
  122. Akula M, McNamee SM, Love Z, Nasraty N, Chan NPM, Whalen M, et al. Retinoic acid related orphan receptor α is a genetic modifier that rescues retinal degeneration in a mouse model of Stargardt disease and Dry AMD. Gene Ther 2024;31(7-8):413-421 View Article PubMed/NCBI
  123. Luo S, Jiang H, Li Q, Qin Y, Yang S, Li J, et al. An adeno-associated virus variant enabling efficient ocular-directed gene delivery across species. Nat Commun 2024;15(1):3780 View Article PubMed/NCBI
  124. Campochiaro PA, Avery R, Brown DM, Heier JS, Ho AC, Huddleston SM, et al. Gene therapy for neovascular age-related macular degeneration by subretinal delivery of RGX-314: a phase 1/2a dose-escalation study. Lancet 2024;403(10436):1563-1573 View Article PubMed/NCBI
  125. Wu H, Li J, Mao X, Li G, Xie L, You Z. Transplantation of rat embryonic stem cell-derived retinal cells restores visual function in the Royal College of Surgeons rats. Doc Ophthalmol 2018;137(2):71-78 View Article PubMed/NCBI
  126. Lund RD, Wang S, Klimanskaya I, Holmes T, Ramos-Kelsey R, Lu B, et al. Human embryonic stem cell-derived cells rescue visual function in dystrophic RCS rats. Cloning Stem Cells 2006;8(3):189-199 View Article PubMed/NCBI
  127. Santos-Ferreira T, Völkner M, Borsch O, Haas J, Cimalla P, Vasudevan P, et al. Stem Cell-Derived Photoreceptor Transplants Differentially Integrate Into Mouse Models of Cone-Rod Dystrophy. Invest Ophthalmol Vis Sci 2016;57(7):3509-3520 View Article PubMed/NCBI
  128. Sharma R, Khristov V, Rising A, Jha BS, Dejene R, Hotaling N, et al. Clinical-grade stem cell-derived retinal pigment epithelium patch rescues retinal degeneration in rodents and pigs. Sci Transl Med 2019;11(475):eaat5580 View Article PubMed/NCBI
  129. Salas A, Duarri A, Fontrodona L, Ramírez DM, Badia A, Isla-Magrané H, et al. Cell therapy with hiPSC-derived RPE cells and RPCs prevents visual function loss in a rat model of retinal degeneration. Mol Ther Methods Clin Dev 2021;20:688-702 View Article PubMed/NCBI
  130. Shirai H, Mandai M, Matsushita K, Kuwahara A, Yonemura S, Nakano T, et al. Transplantation of human embryonic stem cell-derived retinal tissue in two primate models of retinal degeneration. Proc Natl Acad Sci U S A 2016;113(1):E81-E90 View Article PubMed/NCBI
  131. Tu HY, Watanabe T, Shirai H, Yamasaki S, Kinoshita M, Matsushita K, et al. Medium- to long-term survival and functional examination of human iPSC-derived retinas in rat and primate models of retinal degeneration. EBioMedicine 2019;39:562-574 View Article PubMed/NCBI
  132. Duarri A, Rodríguez-Bocanegra E, Martínez-Navarrete G, Biarnés M, García M, Ferraro LL, et al. Transplantation of Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium in a Swine Model of Geographic Atrophy. Int J Mol Sci 2021;22(19):10497 View Article PubMed/NCBI
  133. Holan V, Hermankova B, Kossl J. Perspectives of Stem Cell-Based Therapy for Age-Related Retinal Degenerative Diseases. Cell Transplant 2017;26(9):1538-1541 View Article PubMed/NCBI
  134. Idelson M, Alper R, Obolensky A, Ben-Shushan E, Hemo I, Yachimovich-Cohen N, et al. Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells. Cell Stem Cell 2009;5(4):396-408 View Article PubMed/NCBI
  135. Kicic A, Shen WY, Wilson AS, Constable IJ, Robertson T, Rakoczy PE. Differentiation of marrow stromal cells into photoreceptors in the rat eye. J Neurosci 2003;23(21):7742-7749 View Article PubMed/NCBI
  136. Liang Q, Li Q, Ren B, Yin ZQ. Intravenous infusion of small umbilical cord mesenchymal stem cells could enhance safety and delay retinal degeneration in RCS rats. BMC Ophthalmol 2022;22(1):67 View Article PubMed/NCBI
  137. Brown C, Agosta P, McKee C, Walker K, Mazzella M, Alamri A, et al. Human primitive mesenchymal stem cell-derived retinal progenitor cells improved neuroprotection, neurogenesis, and vision in rd12 mouse model of retinitis pigmentosa. Stem Cell Res Ther 2022;13(1):148 View Article PubMed/NCBI
  138. Zhang J, Li P, Zhao G, He S, Xu D, Jiang W, et al. Mesenchymal stem cell-derived extracellular vesicles protect retina in a mouse model of retinitis pigmentosa by anti-inflammation through miR-146a-Nr4a3 axis. Stem Cell Res Ther 2022;13(1):394 View Article PubMed/NCBI
  139. Wei X, Mu H, Zhang Q, Zhang Z, Ru Y, Lai K, et al. MSCs act as biopatches for blood-retinal barrier preservation to enhance functional recovery after retinal I/R. Mol Ther Nucleic Acids 2025;36(1):102445 View Article PubMed/NCBI
  140. Tong J, Chen Y, Ling X, Huang Z, Yao G, Xie Z. MSC-derived exosomal miR-125b-5p suppressed retinal microvascular endothelial cell ferroptosis in diabetic retinopathy. Stem Cells 2025;43(6):sxaf023 View Article PubMed/NCBI
  141. Ayton LN, Barnes N, Dagnelie G, Fujikado T, Goetz G, Hornig R, et al. An update on retinal prostheses. Clin Neurophysiol 2020;131(6):1383-1398 View Article PubMed/NCBI
  142. Ramirez KA, Drew-Bear LE, Vega-Garces M, Betancourt-Belandria H, Arevalo JF. An update on visual prosthesis. Int J Retina Vitreous 2023;9(1):73 View Article PubMed/NCBI
  143. Tang Z, Ye F, Ni N, Fan X, Lu L, Gu P. Frontier applications of retinal nanomedicine: progress, challenges and perspectives. J Nanobiotechnology 2025;23(1):143 View Article PubMed/NCBI
  144. Yang R, Zhao P, Wang L, Feng C, Peng C, Wang Z, et al. Assessment of visual function in blind mice and monkeys with subretinally implanted nanowire arrays as artificial photoreceptors. Nat Biomed Eng 2024;8(8):1018-1039 View Article PubMed/NCBI
  145. Nie J, Eom K, AlGhosain HM, Neifert A, Cherian A, Gerbaka GM, et al. Intravitreally Injected Plasmonic Nanorods Activate Bipolar Cells with Patterned Near-Infrared Laser Projection. ACS Nano 2025;19(12):11823-11840 View Article PubMed/NCBI
  146. Wang S, Jiang C, Yu Y, Zhang Z, Quhe R, Yang R, et al. Tellurium nanowire retinal nanoprosthesis improves vision in models of blindness. Science 2025;388(6751):eadu2987 View Article PubMed/NCBI
  147. Tochitsky I, Kramer RH. Optopharmacological tools for restoring visual function in degenerative retinal diseases. Curr Opin Neurobiol 2015;34:74-78 View Article PubMed/NCBI
  148. Ng BWJ, Tan TE, Kostin V, MacLaren RE, Cehajic-Kapetanovic J. Characterizing Inner Retinal Changes in End-Stage Inherited Retinal Diseases That Might be Suitable for Optogenetic Therapies. Transl Vis Sci Technol 2025;14(6):2 View Article PubMed/NCBI
  149. Roh H, Kang J, Lee HM, Im M. Enhanced Optogenetic Stimulation of Retinal Ganglion Cells With Assistive Electric Stimulation for Low Optical Power Artificial Vision. IEEE Trans Neural Syst Rehabil Eng 2025;33:1958-1968 View Article PubMed/NCBI
  150. Rodgers J, Hughes S, Ebrahimi AS, Allen AE, Storchi R, Lindner M, et al. Enhanced restoration of visual code after targeting ON bipolar cells compared with retinal ganglion cells with optogenetic therapy. Mol Ther 2025;33(3):1264-1281 View Article PubMed/NCBI
  151. Liu J, Ma AKH, So KF, Lee VWH, Chiu K. Mechanisms of electrical stimulation in eye diseases: A narrative review. Adv Ophthalmol Pract Res 2022;2(2):100060 View Article PubMed/NCBI
  152. Weiland JD, Walston ST, Humayun MS. Electrical Stimulation of the Retina to Produce Artificial Vision. Annu Rev Vis Sci 2016;2:273-294 View Article PubMed/NCBI
  153. Schatz A, Pach J, Gosheva M, Naycheva L, Willmann G, Wilhelm B, et al. Transcorneal Electrical Stimulation for Patients With Retinitis Pigmentosa: A Prospective, Randomized, Sham-Controlled Follow-up Study Over 1 Year. Invest Ophthalmol Vis Sci 2017;58(1):257-269 View Article PubMed/NCBI
  154. Bittner AK, Seger K, Salveson R, Kayser S, Morrison N, Vargas P, et al. Randomized controlled trial of electro-stimulation therapies to modulate retinal blood flow and visual function in retinitis pigmentosa. Acta Ophthalmol 2018;96(3):e366-e376 View Article PubMed/NCBI
  155. Della Volpe-Waizel M, Zuche HC, Müller U, Rickmann A, Scholl HPN, Todorova MG. Metabolic monitoring of transcorneal electrical stimulation in retinitis pigmentosa. Graefes Arch Clin Exp Ophthalmol 2020;258(1):79-87 View Article PubMed/NCBI
  156. Yang J, Wahner-Roedler DL, Zhou X, Johnson LA, Do A, Pachman DR, et al. Acupuncture for palliative cancer pain management: systematic review. BMJ Support Palliat Care 2021;11(3):264-270 View Article PubMed/NCBI
  157. Wang X, Lin Q, Tian L, Li X, Fukuyama T, Ten W, et al. Electroacupuncture alleviates damage to myopic RGCs probably through lncRNA-XR_002789763.1-mediated mitophagy. Chin Med 2025;20(1):16 View Article PubMed/NCBI
  158. Chu-Tan JA, Kirkby M, Natoli R. Running to save sight: The effects of exercise on retinal health and function. Clin Exp Ophthalmol 2022;50(1):74-90 View Article PubMed/NCBI
  159. Zhang X, Girardot PE, Sellers JT, Li Y, Wang J, Chrenek MA, et al. Wheel running exercise protects against retinal degeneration in the I307N rhodopsin mouse model of inducible autosomal dominant retinitis pigmentosa. Mol Vis 2019;25:462-476 PubMed/NCBI
  160. Levinson JD, Joseph E, Ward LA, Nocera JR, Pardue MT, Bruce BB, et al. Physical Activity and Quality of Life in Retinitis Pigmentosa. J Ophthalmol 2017;2017:6950642 View Article PubMed/NCBI
  161. Le P, Nguyen M, Vu T, Dao DP, Olson D, Zhang AY. Anxiety and Depression in Patients With Retinitis Pigmentosa. J Vitreoretin Dis 2021;5(2):114-120 View Article PubMed/NCBI
  162. Watanabe K, Aouadj C, Hiratsuka Y, Yamamoto S, Murakami A. Quality of Life and Economic Impacts of Retinitis Pigmentosa on Japanese Patients: A Non-interventional Cross-sectional Study. Adv Ther 2023;40(5):2375-2393 View Article PubMed/NCBI
  163. Ma DJ, Lee HS, Kim K, Choi S, Jang I, Cho SH, et al. Whole-exome sequencing in 168 Korean patients with inherited retinal degeneration. BMC Med Genomics 2021;14(1):74 View Article PubMed/NCBI
  164. Weisschuh N, Obermaier CD, Battke F, Bernd A, Kuehlewein L, Nasser F, et al. Genetic architecture of inherited retinal degeneration in Germany: A large cohort study from a single diagnostic center over a 9-year period. Hum Mutat 2020;41(9):1514-1527 View Article PubMed/NCBI
  165. Miller AL, James RE, Harvey AR, Trifunović D, Carvalho LS. The role of epigenetic changes in the pathology and treatment of inherited retinal diseases. Front Cell Dev Biol 2023;11:1224078 View Article PubMed/NCBI
  166. Karuntu JS, Almushattat H, Nguyen XT, Plomp AS, Wanders RJA, Hoyng CB, et al. Syndromic retinitis pigmentosa. Prog Retin Eye Res 2025;107:101324 View Article PubMed/NCBI
  167. Elhusseiny AM, Zhang S, Sharabura AB, Dehnel JR, Uwaydat SH. Intrafamilial Phenotypic Variability in PRPH2-Related Retinopathy. Cureus 2024;16(10):e72580 View Article PubMed/NCBI
  168. Birch DG, Cheetham JK, Daiger SP, Hoyng C, Kay C, MacDonald IM, et al. Overcoming the Challenges to Clinical Development of X-Linked Retinitis Pigmentosa Therapies: Proceedings of an Expert Panel. Transl Vis Sci Technol 2023;12(6):5 View Article PubMed/NCBI
  169. Llonch S, Carido M, Ader M. Organoid technology for retinal repair. Dev Biol 2018;433(2):132-143 View Article PubMed/NCBI
  170. Lu B, Avalos P, Svendsen S, Zhang C, Nocito L, Jones MK, et al. GMP-grade human neural progenitors delivered subretinally protect vision in rat model of retinal degeneration and survive in minipigs. J Transl Med 2023;21(1):650 View Article PubMed/NCBI
  171. Bell CM, Zack DJ, Berlinicke CA. Human Organoids for the Study of Retinal Development and Disease. Annu Rev Vis Sci 2020;6:91-114 View Article PubMed/NCBI
  172. Liu Y, Gao L, Chen W, Yan Y, Ye Z, Li Z. “Armed in-vitro retina”-generating microglial retinal organoids, where are we now?. Front Cell Dev Biol 2025;13:1574283 View Article PubMed/NCBI
  173. Zhu Y, Cao B, Tolone A, Yan J, Christensen G, Arango-Gonzalez B, et al. In vitro Model Systems for Studies Into Retinal Neuroprotection. Front Neurosci 2022;16:938089 View Article PubMed/NCBI
  174. Brooks MJ, Chen HY, Kelley RA, Mondal AK, Nagashima K, De Val N, et al. Improved Retinal Organoid Differentiation by Modulating Signaling Pathways Revealed by Comparative Transcriptome Analyses with Development In Vivo. Stem Cell Reports 2019;13(5):891-905 View Article PubMed/NCBI
  175. Capowski EE, Samimi K, Mayerl SJ, Phillips MJ, Pinilla I, Howden SE, et al. Reproducibility and staging of 3D human retinal organoids across multiple pluripotent stem cell lines. Development 2019;146(1):dev171686 View Article PubMed/NCBI
  176. Zhang Z, Xu Z, Yuan F, Jin K, Xiang M. Retinal Organoid Technology: Where Are We Now?. Int J Mol Sci 2021;22(19):10244 View Article PubMed/NCBI
  177. Kim HM, Woo SJ. Ocular Drug Delivery to the Retina: Current Innovations and Future Perspectives. Pharmaceutics 2021;13(1):108 View Article PubMed/NCBI
  178. Silva-Cunha A. Advances in Ocular Drug Delivery Systems. Pharmaceutics 2021;13(9):1383 View Article PubMed/NCBI
  179. Sobh M, Lagali PS, Ghiasi M, Montroy J, Dollin M, Hurley B, et al. Safety and Efficacy of Adeno-Associated Viral Gene Therapy in Patients With Retinal Degeneration: A Systematic Review and Meta-Analysis. Transl Vis Sci Technol 2023;12(11):24 View Article PubMed/NCBI
  180. Tripepi D, Jalil A, Ally N, Buzzi M, Moussa G, Rothschild PR, et al. The Role of Subretinal Injection in Ophthalmic Surgery: Therapeutic Agent Delivery and Other Indications. Int J Mol Sci 2023;24(13):10535 View Article PubMed/NCBI
  181. Guest JM, Malbin B, Abrams G, Parendo A, Das S, Okeagu C, et al. Accuracy of intravitreal injection volume for aflibercept pre-filled syringe and BD Luer-Lok one-milliliter syringe. Int J Retina Vitreous 2022;8(1):27 View Article PubMed/NCBI
  182. Collins M, Thrasher A. Gene therapy: progress and predictions. Proc Biol Sci 2015;282(1821):20143003 View Article PubMed/NCBI
  183. Gao H, Zeng Y, Huang X, A L, Liang Q, Xie J, et al. Extracellular vesicles from organoid-derived human retinal progenitor cells prevent lipid overload-induced retinal pigment epithelium injury by regulating fatty acid metabolism. J Extracell Vesicles 2024;13(1):e12401 View Article PubMed/NCBI
  184. Maeda T, Takahashi M. iPSC-RPE in Retinal Degeneration: Recent Advancements and Future Perspectives. Cold Spring Harb Perspect Med 2023;13(8):a041308 View Article PubMed/NCBI
  185. Arsenijevic Y, Chang N, Mercey O, El Fersioui Y, Koskiniemi-Kuendig H, Joubert C, et al. Fine-tuning FAM161A gene augmentation therapy to restore retinal function. EMBO Mol Med 2024;16(4):805-822 View Article PubMed/NCBI
  186. Maguire MG, Birch DG, Duncan JL, Ayala AR, Ayton LN, Cheetham JK, et al. Endpoints and Design for Clinical Trials in USH2A-Related Retinal Degeneration: Results and Recommendations From the RUSH2A Natural History Study. Transl Vis Sci Technol 2024;13(10):15 View Article PubMed/NCBI
  187. Otte B, Andrews C, Lacy G, Branham K, Musch DC, Jayasundera KT. Clinical trial design for neuroprotection in RHO autosomal dominant retinitis pigmentosa; outcome measure considerations. Ophthalmic Genet 2021;42(2):170-177 View Article PubMed/NCBI
  188. Kuriyan AE, Albini TA, Townsend JH, Rodriguez M, Pandya HK, Leonard RE, et al. Vision Loss after Intravitreal Injection of Autologous “Stem Cells” for AMD. N Engl J Med 2017;376(11):1047-1053 View Article PubMed/NCBI
  189. Rong AJ, Lam BL, Ansari ZA, Albini TA. Vision Loss Secondary to Autologous Adipose Stem Cell Injections: A Rising Problem. JAMA Ophthalmol 2018;136(1):97-99 View Article PubMed/NCBI
  190. Singh MS, Park SS, Albini TA, Canto-Soler MV, Klassen H, MacLaren RE, et al. Retinal stem cell transplantation: Balancing safety and potential. Prog Retin Eye Res 2020;75:100779 View Article PubMed/NCBI
  191. Haapaniemi E, Botla S, Persson J, Schmierer B, Taipale J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat Med 2018;24(7):927-930 View Article PubMed/NCBI
  192. Lopata E, Terrone C, Gopalan A. Opportunities and challenges surrounding financial models for high-investment medications: A survey of access decision-makers and employers. J Manag Care Spec Pharm 2023;29(7):782-790 View Article PubMed/NCBI
  193. Phares S, Trusheim M, Emond SK, Pearson SD. Managing the challenges of paying for gene therapy: strategies for market action and policy reform in the United States. J Comp Eff Res 2024;13(12):e240118 View Article PubMed/NCBI
  • Nature Cell and Science
  • 2958-695X

Decoding Retinal Degeneration Diseases: From Molecular Pathways to Cutting-edge Therapies

Youyuan Zhuang, Jiuyi Lu, Jiawen Li, Chong Chen, Xinting Liu
  • Reset Zoom
  • Download TIFF