Next Article in Journal
Antimicrobial Resistance Gene Patterns in Traditional Montenegrin Njeguški Cheese Revealed by qPCR
Previous Article in Journal
Interplay Between Genetic Diversity and Tree Vitality in Fraxinus excelsior Populations Affected by Ash Dieback
Previous Article in Special Issue
Exploring Concomitant Ophthalmic Comorbidities in Portuguese Patients with Inherited Retinal Diseases: A Comprehensive Clinical Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Single-Cell Transcriptomics in Inherited Retinal Dystrophies: Current Findings and Emerging Perspectives

1
MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
2
Princess Alexandra Eye Pavilion, NHS Lothian, Chalmers Street, Edinburgh EH3 9HA, UK
*
Authors to whom correspondence should be addressed.
Genes 2025, 16(9), 1088; https://doi.org/10.3390/genes16091088
Submission received: 9 August 2025 / Revised: 9 September 2025 / Accepted: 10 September 2025 / Published: 16 September 2025
(This article belongs to the Special Issue Genetics in Retinal Diseases—2nd Edition)

Abstract

Inherited retinal dystrophies (IRDs) represent a diverse group of disorders caused by mutations in genes essential for retinal function and maintenance. Traditional bulk RNA sequencing techniques provide valuable information for deciphering disease pathogenesis but lack the resolution to capture variation among specific cell clusters during disease progression. In contrast, single-cell transcriptomics, including single-cell RNA sequencing (scRNA-seq), enables detailed examination of distinct retinal clusters in both healthy and diseased states, uncovering unique gene expression signatures and early molecular changes preceding photoreceptor cell death in IRDs. These insights not only deepen our understanding of the complex pathogenesis of IRDs but also highlight potential targets for novel therapeutic interventions. In this review, we examine the recent literature on the application of single-cell transcriptomics in IRDs to explore how these techniques enhance our understanding of disease mechanisms and contribute to the identification of new therapeutic targets.

1. Introduction

Inherited retinal dystrophies (IRDs) represent a clinically and genetically heterogeneous group of disorders characterised by progressive photoreceptor degeneration, ultimately leading to severe vision loss [1]. These conditions affect approximately 1 in 3000 [1,2] to 4000 individuals worldwide [3].
IRDs are characterised by remarkable genetic complexity, with mutations in at least 300 genes identified to date [4], expressed through multiple inheritance patterns including autosomal dominant, autosomal recessive, and X-linked forms [3]. Major IRD subtypes examined in this review include retinitis pigmentosa (RP), Leber congenital amaurosis (LCA), enhanced S-cone syndrome (ESCS), Stargardt disease (STGD), and achromatopsia (ACHM), each presenting distinct clinical phenotypes and underlying genetic causes. Significant phenotypic variability exists even amongst individuals carrying the same genetic mutations [5], creating complex genotype–phenotype relationships that complicate diagnosis and treatment strategies [6].
IRD pathogenesis involves complex interactions between multiple retinal cell types that both respond to and influence the degenerative process [7], with supporting cells such as Müller glia and microglia often exhibiting early transcriptional changes that may precede overt photoreceptor death [7,8]. Characterising cell-type-specific responses is crucial, as disease progression varies highly between distinct retinal cell populations, requiring targeted approaches that can distinguish different cellular responses to disease.
This review reports studies applying single-cell transcriptomics to IRD samples, examining how these approaches have advanced our understanding of disease mechanisms and dynamics with cellular resolution. We present findings by specific IRD subtypes and genetic mutations, highlighting key mechanistic insights and therapeutic targets identified through single-cell approaches. We then identify common themes across different IRD studies and therapeutic implications before identifying current challenges and future considerations for using single-cell technologies in IRD research.

2. From Bulk to Single-Cell Transcriptomics in IRDs

RNA sequencing (RNA-seq) has revolutionised the study of gene expression profiles in cells and tissues, enabling comprehensive, high-throughput, and quantitative analysis of the entire transcriptome [9]. The unbiased nature of RNA-seq facilitates the identification of novel genes and pathways without requiring prior hypotheses about specific targets [9,10], making it particularly valuable for identifying novel molecular mechanisms in retinal diseases. RNA-seq provides precise quantification across a wide dynamic range, capturing both abundant and rare transcripts, whilst detecting subtle changes in gene expression [9]. However, traditional bulk RNA-seq approaches have significant limitations when applied to heterogeneous tissues like the retina.
Bulk RNA-seq averages gene expression across all cells in a sample, masking cell-type-specific pathological changes that may be crucial for understanding disease mechanisms [11,12]. Early degenerative events affecting only small subsets of cells become diluted and undetectable in bulk analysis, potentially missing critical early disease signatures that could inform therapeutic interventions [13]. This averaging effect is particularly problematic in retinal tissue, which contains over 100 distinct cell types (with approximately 130 identified in mouse retina [14]), each of which may exhibit distinct responses to disease-causing genetic mutations [11].
Single-cell RNA sequencing (scRNA-seq) overcomes these limitations by capturing gene expression profiles with cellular resolution, providing detailed transcriptome profiling and revealing cellular heterogeneity within complex tissues [11,12]. Computational analyses to cluster cells with similar expression profiles enables the identification of distinct cell populations, facilitating unbiased cell type classification [10], including rare cell types [12] that may be missed in bulk analysis. This single-cell resolution allows investigation of how IRDs affect specific cell types and enables detection of early pathological changes in vulnerable cell populations [12]. Additionally, scRNA-seq can reveal novel cell states, such as stress-responsive or transitional cell populations that emerge during disease progression [11].
The development of single-cell transcriptomics has enabled the creation of comprehensive retinal cell atlases, providing essential reference datasets for understanding both healthy retinal biology and disease-associated changes [11]. Macosko et al. [15] delivered one of the first single-cell transcriptomic atlases of the mouse retina using Drop-seq, establishing a foundational framework for retinal cell type classification. Building upon these foundational studies, Menon et al. [10] created the first human retinal transcriptomic atlas using droplet-based and nanowell platforms, bridging the gap between model organism data and human retinal biology. These atlases have undergone continuous refinement and expansion, now serving as essential reference datasets for interpreting disease-associated transcriptional changes in IRD research.

3. Disease-Specific Insights from Single-Cell Transcriptomics

We searched the literature using PubMed to identify relevant studies examining single-cell transcriptomic approaches in IRDs, employing a combination of controlled vocabulary (Medical Subject Headings, MeSH) and free-text terms. The search strategy combined two main groups using the Boolean operator “AND”: single-cell transcriptomic approaches and inherited retinal dystrophies. Single-cell methodologies included “single-cell RNA sequencing”, “scRNA-seq”, “single-nucleus RNA sequencing”, platform-specific terms like “10x Genomics” and “Drop-seq”, and relevant MeSH terms including “Single-Cell Analysis” and “Transcriptome”. IRD terms contained broad categories such as “retinal degeneration” and “inherited retinal dystrophies”, specific conditions including “retinitis pigmentosa”, “Stargardt disease”, and “Leber congenital amaurosis”, plus corresponding MeSH terms such as “Retinal Degeneration”, “Retinal Dystrophies”, and “Eye Diseases, Hereditary”. We restricted the search terms to title and abstract fields ([tiab]) and limited the results to English-language publications with full-text availability, published between 2019 and 2025 to capture recent technological advances in single-cell transcriptomics.
The initial PubMed search yielded 412 results on 20 June 2025. Studies were included if they utilised single-cell transcriptomics methodologies to investigate IRDs. We excluded studies examining exclusively healthy retinal samples to maintain focus on pathological processes in IRDs. After screening, 27 papers met the inclusion criteria, representing 23 distinct datasets (some papers shared data from the same experiment). These 23 studies form the basis for this review and are summarised in Table 1. The following sections present gene-specific findings grouped by disease type: RP (with subsections on RHO, PDE6B, RPGR, USH, and other RP gene mutations), LCA, ESCS, STGD, and ACHM (Figure 1).

3.1. Retinitis Pigmentosa

Retinitis pigmentosa (RP) is the most common IRD [2,43], affecting more than 1.5 million people worldwide [43] with a prevalence of approximately 1 in 4000 individuals [3,5,43]. RP is the leading cause of inherited blindness in working-age adults and has substantial impacts on life quality of patients and healthcare systems [3,43].
The progression of RP varies between individuals but classically begins with night blindness, followed by progressively narrowing visual fields (tunnel vision) and can ultimately lead to the loss of central vision [3]. While some individuals experience noticeable vision loss during childhood, others remain free of symptoms until reaching mid-adulthood, making age of onset an unreliable indicator of disease severity [3,5,6]. In most typical RP cases, the decline in rod photoreceptors exceeds that of cone photoreceptors, though some instances show cone–rod degeneration patterns with reduced visual acuity and impaired colour vision [3].
RP is genetically heterogeneous, with the majority of cases being monogenic [3] and linked to mutations in more than 80 genes [5]. In most cases, RP remains restricted to the eye (non-syndromic RP) [5], whilst 20–30% of RP patients present with an associated non-ocular condition (syndromic RP) [5]. The most common syndromic forms include Usher syndrome (USH; characterised by hearing impairment) [3,5] and Bardet–Biedl syndrome (involving obesity, renal abnormalities, and cognitive impairment) [3,5].
Currently, no curative treatments are available that can stop RP progression or restore vision [43]. Given that RP-associated genes have different localisations and functions across retinal cell types [3,5], there is a critical need for molecular-level understanding to develop personalised therapeutic approaches targeting specific cellular pathways. The following sections present findings from single-cell studies organised by the specific RP-associated genes for which we found transcriptomic studies.

3.1.1. RHO Mutations

Mutations in the rhodopsin gene (RHO), first identified in 1990 by Dryja et al. [5,44,45], are associated with autosomal dominant RP (adRP) [5], accounting for approximately 25% of adRP cases [3,6,46]. The clinical course is highly variable, with onset typically before age 10 and legal blindness often developing between 50 and 80 years [5]. Additionally, mutations in RHO are also associated with congenital stationary night blindness (CSNB), a nonprogressive condition [5].
RHO encodes the G protein-coupled receptor rhodopsin, which is activated by photons and initiates the phototransduction cascade in the outer segment of rod photoreceptors [45]. Over 200 mutations in RHO have been identified, with the P23H mutation being the most common mutation in the human RHO gene [45] and R135W representing another missense variant affecting rhodopsin structure [47]. P23H results in pronounced misfolding and endoplasmic reticulum (ER) retention [45,48], whereas R135W causes partial misfolding, disrupting interactions with signalling proteins [47]. Both mutations trigger ER stress, elevate levels of reactive oxygen species, and ultimately cause photoreceptor cell death through rhodopsin accumulation in the ER [45,48,49].
Single-cell transcriptomics studies have examined these molecular defects across disease progression stages using complementary model systems: P23H mouse models during active degeneration (postnatal weeks 7 and 10) [16,17], adult zebrafish to understand cell-type-specific changes [18], and human iPSC-derived retinal organoids (rOrgs) at immature (D120) and mature (D270) stages [19].
During active degeneration, shrinking numbers of mature rod photoreceptors [16,18,19] showed functional decline through decreased expression of phototransduction genes and disrupted structural integrity through reduced expression of genes involved in the photoreceptor cilium, inner/outer segments, and overall photoreceptor development [16]. Elevated stress responses included upregulated misfolded protein responses, oxidative stress markers, and metabolic shifts toward enhanced glycolysis [18].
In response to oxidative stress, zebrafish cones, retinal pigment epithelium (RPE) cells, and bipolar cells showed upregulated antioxidant genes [18], whilst mouse Müller glia exhibited compensatory responses by upregulating photoreceptor maintenance genes and, during advanced degeneration, co-expression of rod/cone markers [16]. Patient organoids demonstrated enhanced cell–cell communication networks and increased astrocyte proportions with activated inflammatory signalling [19]. Therapeutic intervention with fibroblast growth factor 21 (FGF21) in RHO mice activated axon development and synaptic remodelling pathways in Müller glia, suggesting improved glial function through the serum response factor (SRF) pathway. SRF motifs were enriched in FGF21-responsive genes, with Srf specifically expressed in Müller glia and astrocytes and increased by FGF21 treatment [17].
Unlike mammalian models, the zebrafish P23H model uniquely demonstrated ongoing regenerative capacity alongside degeneration, with dramatically increased retinal progenitor cells and newly forming rods, indicative of continued regeneration [18]. Müller cells shifted transcriptionally towards a proliferative state, whilst microglial populations showed distinct roles in phagocytic clearance and apoptosis regulation to facilitate regeneration [18].

3.1.2. PDE6B Mutations

Rod phosphodiesterase 6 (PDE6) consists of two catalytic subunits (PDE6A and PDE6B) and two inhibitory gamma subunits (PDE6G) [50], with both catalytic subunits essential for full enzymatic activity [51]. PDE6 plays an important role in rod phototransduction by hydrolysing cGMP following light activation of rhodopsin and transducin, leading to closure of cGMP-gated ion channels and membrane hyperpolarisation [50]. PDE6B mutations lead to PDE dysfunction, resulting in failure to hydrolyse cGMP and subsequent cGMP accumulation in the retina [3].
PDE6B mutations cause autosomal recessive RP (arRP) with rod–cone dystrophy patterns, accounting for roughly 2–4% of arRP cases, and autosomal dominant CSNB [5,52,53,54]. Clinical onset typically occurs before age 10 [5,54], with significant peripheral visual field loss manifesting between 10 and 30 years [5].
Single-cell studies have extensively examined rd1 and rd10 mouse strains carrying nonsense and missense Pde6b mutations, respectively, with the rd1 strain showing earlier onset (P7–10) [55] and faster progression (complete photoreceptor loss by P17–21) [56,57] compared to rd10 mice (onset P16–18 [58,59] and complete loss P40–45 [59,60]).
Rod photoreceptors showed severe degeneration (55% to 16% population reduction), with consistently downregulated Pde6b and dysregulated phototransduction, apoptosis, and calcium signalling pathways [20,21,23,24,25].
Abnormal calcium (Ca2+) signalling represented a core pathological mechanism, with rods showing more disruption than cones [20,23]. These changes appeared early before cell death [25]. From early to peak degeneration, upregulated cyclic nucleotide-gated (CNG) channel genes [20,23] and voltage-gated calcium channel (VGCC) genes [23] drive heightened Ca2+ influx. Increased sodium–calcium exchanger (NCX) genes (e.g., Slc8a1) [20,23] and downregulated calcium release-activated channels (CRACs) [23] suggest compensatory responses to elevated Ca2+ levels. Late phases showed reduced CNG channel expression but an upregulated protein kinase G (PKG) pathway (Prkg1) [20], with T-type VGCC and NCX pathways validated as therapeutic targets in explant cultures [23]. Overall, modulating Ca2+ channels or inhibiting PKG may preserve photoreceptor viability.
Ca2+-dependent mitogen-activated protein kinase (MAPK) pathways (ERKs, JNKs, and p38 kinases) showed upregulation in rd1 rods, with MAPK/c-Jun and JNK signalling validated as therapeutic targets [20,21,24,61,62]. Downstream, early growth response 1 (EGR1) functions as a critical transcriptional mediator of cellular stress responses [63], with consistent but temporally distinct patterns: sustained upregulation in rd1 rods [21] versus transient early expression in rd10 rods [25]. Egr1 regulated early degenerative genes as both an activator and repressor depending on degeneration stage, whilst Cebpd became highly upregulated in late phases as a potential neuroprotective mechanism [25].
Integration of single-cell transcriptomics with epigenetic analysis identified chromatin-modifying enzymes histone deacetylase (HDAC) and poly (ADP-ribose) polymerase (PARP) as regulators of non-apoptotic cell death [20,22], with cell-type-specific expression patterns: high Hdac1 in late-stage rd1 rods and reduced Parp1, particularly in cones [20,22]. Conversely, Dong et al. [22] demonstrated that Pde6b mutations triggered HDAC overactivation, causing chromatin condensation and PARP overactivation, with suberoylanilide hydroxamic acid (SAHA) treatment delaying photoreceptor loss through dual-pathway inhibition [22].
During peak [25] and late degeneration [20] in both rd1 and rd10 rods, cones showed early transcriptomic responses preceding secondary death, with rd10 cones more similar to degenerating rods than wild-type cones. Cone responses included ER stress, unfolded protein response, mitochondrial dysfunction, and increased tubulin cytoskeleton transcripts [20,25].
Beyond the primary photoreceptor dysfunction and secondary cone degeneration, single-cell transcriptomics revealed extensive tissue-level responses involving glial and immune populations. Müller glia showed strong gliotic responses with upregulated Gfap, increased transcripts, and higher mitochondrial/ribosomal content [26], as well as greater proliferative capacity alongside rods [21]. Upregulated major histocompatibility complex (MHC) components and complement system genes suggest antigen-presenting and innate immunity functions [26]. Microglial activation was marked by elevated Cd68 expression and dramatic population expansion from 0.34% to 3.94% in diseased retinas [24].
Metabolic profiling revealed complex disruptions across retinal cell types in both rd1 and rd10 mice. Cones consistently showed downregulated glycolysis genes [20,25], whilst rod responses varied by model: downregulated glycolytic enzymes in early rd10 phases [25] versus upregulated glycolysis and oxidative phosphorylation (OXPHOS) genes in rd1 rods [20]. Both models eventually showed mitochondrial dysfunction with downregulated respiratory complex genes [20,25]. Müller cells demonstrated enhanced OXPHOS and metal homeostasis genes, suggesting compensatory metabolic support and antioxidant defence [20,26].
In summary, PDE6B-mediated RP involves elevated cGMP levels causing abnormal Ca2+ influx, which activates MAPK/JNK/EGR1 and HDAC/PARP pathways, triggering metabolic reprogramming and coordinated cell death responses. Primary rod death and secondary cone degeneration occur alongside compensatory Müller glial responses and broader immune activation.

3.1.3. RPGR Mutations

Mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene are responsible for X-linked retinitis pigmentosa (XLRP), accounting for approximately 70–90% of XLRP cases and 10–20% of all RP cases [3,64,65,66]. RPGR interacts with actin-binding proteins to regulate outer segment maintenance and protein trafficking in the connecting cilium, essential for facilitating rhodopsin transport to the outer segments [67,68]. As RPGR is located on the X chromosome, males are more severely affected, whilst female carriers can exhibit variable symptoms ranging from mild to severe depending on patterns of X-chromosome inactivation [69]. RPGR mutations are associated with early onset of central vision loss before age 5 and a mean age of legal blindness of 45 years [5,70]. Known syndromic associations include hearing loss and respiratory infections [5,71,72]. Both rods and cones can be affected and several IRD phenotypes are linked to the disease, including cone–rod dystrophy (CRD), cone dystrophy (CD), and macular dystrophy (MD) [5,73,74,75,76]. The RPGR gene encodes 10 different transcripts through alternative splicing, 5 of which are protein-coding. Two major isoforms are especially important: RPGREx1-19 and RPGRORF15. The constitutive RPGREx1-19 isoform spans 19 exons and is ubiquitously expressed, whilst the RPGRORF15 isoform contains a unique ORF15 exon with highly repetitive sequence coding for glutamate–glycine repeats [66]. Although RPGRORF15 has a restricted expression pattern that includes the retina, where it is predominantly expressed, recent evidence demonstrates its importance in other ciliated tissues, including airway epithelia where it regulates motile cilia function [77]. Despite its widespread expression, RPGREx1-19’s unique exons have not been associated with human disease. In contrast, RPGRORF15 is critical for both photoreceptor function and survival as well as motile cilia regulation, with up to 60% of RPGR mutations occurring within the ORF15 region [66,77].
Despite the high frequency of RPGR mutations, only two single-cell studies were identified. Li et al. [28] established patient-derived iPSC organoid models, analysing developmental stages D40-D200 and documenting cell composition differences between control and RPGR patient organoids. Newton et al. [27] provided deeper mechanistic insights by combining single-cell transcriptomics with multiple complementary validation methods using RPGR-mutant mouse models (RpgrEx3d8 and RpgrORFd5).
Pseudotime analysis mapped disease progression from healthy to degenerating rod photoreceptors, revealing progressive downregulation of phototransduction genes and upregulated PI3K/AKT signalling. Dysregulated autophagy showed increased autophagy-related genes and lysosome biogenesis, leading to mitochondrial dysfunction with upregulated mitochondrial genes as compensatory responses. Single-cell analysis revealed highly upregulated necroptosis genes, identifying necroptosis as the predominant cell death mechanism and therapeutic target.
In summary, the mouse study from Newton et al. [27] provides detailed mechanistic insights into RPGR pathogenesis, revealing autophagy disruption, mitochondrial stress, and necroptotic cell death, whilst the organoid study from Li et al. [28] establishes a valuable human disease model system and demonstrates developmental and transcriptomic changes in RPGR-mutant photoreceptors.

3.1.4. USH Mutations

Usher syndrome (USH) represents the most frequent syndromic form of RP, accounting for 20–40% of arRP cases [3]. It is associated with hearing impairment and is classified into three clinical subtypes based on severity, age of onset, and vestibular involvement, each of which is linked to a specific genetic cause [78,79]. Among these subtypes, USH1 represents the most severe form, with congenital hearing loss, loss of vestibular function, and early-onset RP typically manifesting before puberty [79,80,81]. USH2 is the most common subtype, representing over half of all USH cases, and is characterised by moderate-to-severe congenital hearing impairment, intact vestibular function, and post-pubertal onset of RP [79,80,81]. USH3 represents the least common USH subtype, characterised by progressive hearing loss appearing from late childhood to age 35, varying degrees of vestibular impairment, and the onset of RP usually by the second decade of life [81]. USH proteins play important roles in maintaining the structure and function of sensory cells by building protein networks (molecular scaffolds) that coordinate cellular transport and signalling. They help maintain normal hearing, balance, and vision [82].
In our literature screen, we found only one study by Leong et al. [29] that definitely meets all inclusion criteria (Table 1). The authors performed single-cell transcriptomics on retinal organoids derived from induced pluripotent stem cells (iPSCs) from Usher syndrome type 1B (USH1B) patients and controls, representing the first human RP model for USH1B in a pre-symptomatic disease state consistent with childhood onset. The disease is caused by a mutation in the myosin VIIA (MYO7A) gene, which encodes an ATP-dependent myosin motor protein that travels along actin filaments, transporting and anchoring protein complexes [83,84,85].
While MYO7A has traditionally been associated with RPE function, the scRNA-seq analysis by Leong et al. [29] detected MYO7A expression in Müller cells, bipolar cells, rods, and cones. USH1B rods showed GO enrichment for chemical and oxidative stress responses and upregulated pro-apoptotic and antioxidant markers, whilst cones showed no significant molecular pathology. These rod-specific stress responses aligned with clinical observations of primary rod degeneration in USH1B-RP. Müller cells showed differentially expressed genes enriched for apoptotic signalling regulation, suggesting potential vulnerability of this cell type.

3.1.5. Other RP Gene Mutations

Beyond the better-characterised gene mutations in RHO, PDE6B, RPGR, and USH, studies on other RP-associated gene mutations remain relatively sparse. Here, we focus on studies for five RP genes, CWC27, PRPF8, CRB1, PROM1, and ADIPOR1, for each of which we only identified one relevant paper (Table 1). Despite the limited number of studies, these genes represent a diverse spectrum of molecular functions and pathophysiological mechanisms. Together, they illustrate how defects in distinct processes (i.e., pre-mRNA splicing, retinal cell polarity, photoreceptor disc morphogenesis, and metabolic homeostasis) can each culminate in an RP-like phenotype.
Mutations in the spliceosomal gene CWC27 can cause non-syndromic and syndromic forms of arRP. Clinical phenotypes include retinal degeneration, skeletal defects, and neurological defects [86,87]. CWC27 encodes a spliceosome-associated protein crucial for pre-mRNA splicing [88].
Bertrand et al. [30] provided the first single-cell transcriptomics analysis of Cwc27-associated retinal degeneration using a mouse model at early disease stages. The analysis provided cellular-level insights into how CWC27 splicing defects primarily impact rod photoreceptor mitochondrial function and trigger Müller glial inflammatory responses, while other retinal cell populations showed minimal changes.
Another splicing factor mutation associated with retinal degeneration involves PRPF8. PRPF8 mutations cause adRP type 13 (RP13), accounting for approximately 11% of adRP cases and representing one of the most frequent causes of IRD [31,46], leading to early-onset RP typically before age 10 [5]. PRPF8 encodes the highly conserved scaffolding protein PRPF8, a precursor mRNA processing factor (PRPF) that is part of the spliceosome [46,89,90,91]. Despite its ubiquitous expression and essential role in pre-mRNA splicing, mutations in PRPF8 lead to retina-specific pathology [31,90].
Atkinson et al. [31] provided the first comprehensive single-cell analysis using RP13 patient-derived iPSCs, revealing higher proportions of degenerating rods in retinal organoids. Both photoreceptor types showed significant downregulation of cell-type markers and phototransduction genes, with pathway analysis identifying phototransduction and photoreceptor degeneration as the top disrupted processes. These findings complement broader tissue comparison studies (i.e., bulk RNA-seq across iPSCs, kidney organoids, RPE, and retinal organoids), providing cellular-level resolution of how PRPF8 splicing dysfunction translates into photoreceptor-specific pathology.
Beyond splicing-related mechanisms, cellular polarity can impact photoreceptor viability, as shown by CRB1. CRB1 encodes a large transmembrane protein that functions as a regulator of apical–basal cell polarity and cell adhesion between photoreceptors and Müller cells in the retina [92,93,94]. Mutations in the Crumbs homolog 1 (CRB1) gene can cause the early-onset IRDs arRP and LCA [5,94]. Loss of CRB1 function disrupts critical cellular junctions and leads to photoreceptor degeneration [93,94,95].
Boon et al. [32] employed scRNA-seq on CRB1 patient-derived iPSC retinal organoids, identifying endosomal dysfunction as the primary pathological mechanism in rods and Müller cells, alongside rod phototransduction defects. Müller cells showed pathway dysregulation, including the endosomal system, cell maintenance/motility/adhesion, protein binding, cell death, and iron transport/phosphorylation. AAV-mediated gene therapy partially restored endosomal gene expression patterns and increased photoreceptor numbers, leading to identification of endosomal recycling proteins as potential therapeutic targets.
Another transmembrane protein implicated in RP is PROM1 (prominin-1/CD133), which plays a crucial role in photoreceptor outer segment disc morphogenesis [96,97]. Mutations in PROM1 disrupt the membrane organisation and shedding of outer segment discs, impairing the continuous renewal of photoreceptor cells. This structural dysfunction compromises photoreceptor function, ultimately resulting in vision loss [96,97]. They are associated with arRP [98,99] but can also cause autosomal dominant MD [5,100].
Shigesada et al. [33] used scRNA-seq in pre-symptomatic Prom1-deficient mice, capturing immediate transcriptional responses before morphological changes. Visual function genes were downregulated in photoreceptors, whilst stress responses involved cell–cell communication through upregulated Edn2 in rod signalling to glial cells, and widespread stress markers across multiple cell types were observed. Rod-specific Igf1 downregulation led to the identification of two therapeutic strategies, metabolic support via IGF1 and anti-inflammation via endothelin blockade, both validated through AAV-IGF1 gene therapy and endothelin antagonist therapy (bosentan) improving retinal function.
The final RP gene for which we found a single-cell transcriptomics study is ADIPOR1, linking retinal degeneration to disrupted metabolic homeostasis [101]. ADIPOR1 encodes the adiponectin receptor protein 1 and is involved in cell signalling pathways that regulate energy homeostasis, inflammation, and cell survival [102]. Though widely expressed, ADIPOR1 shows the highest protein levels in neural tissues, particularly photoreceptors and RPE cells [103]. ADIPOR1 possesses intrinsic ceramidase activity [102,104] that breaks down toxic ceramides [105,106]. Mutations have been associated with both non-syndromic and syndromic forms of RP [107,108], as well as being identified as a genetic risk factor for advanced age-related macular degeneration [109].
Lewandowski et al. [34] conducted comprehensive single-cell analysis of Adipor1 knockout mice across multiple disease stages, revealing ADIPOR1 as the most abundant retinal ceramidase. Its absence caused ceramide accumulation and progressive photoreceptor death, identifying ceramide metabolism as a therapeutic target. This led to successful pharmacological intervention with FDA-approved drugs (desipramine/L-cycloserine) that reduced ceramide levels and provided neuroprotection.

3.2. Leber Congenital Amaurosis

Among the IRDs, Leber congenital amaurosis (LCA) represents one of the most severe forms [110], characterised by profound visual impairment that typically presents at or in early infancy [111]. It accounts for approximately 5% of all IRDs and 20% of childhood blindness worldwide [111].
The clinical presentation can vary but typically includes severe visual impairment, reduced or absent electroretinography (ERG) responses, involuntary eye movements (nystagmus), compromised or absent pupillary responses, and eye pressing or poking with fingers (oculodigital sign) [111]. The exact clinical phenotype varies depending on the specific gene affected [111].
Most of the LCA genes follow an autosomal recessive inheritance pattern [110], but some cause autosomal dominant forms (such as CRX) [110,111]. There are 19 distinct LCA subtypes (LCA1-LCA19) caused by genetic variants in different genes, with over 25 genes associated with the condition [112,113]. Among these, three genes have been studied using single-cell transcriptomics approaches: CEP290, RPE65, and CRX.
CEP290 (290 kDa centrosomal protein) represents one of the most frequently mutated genes in LCA [114], accounting for 15–30% of all LCA cases and designated as LCA10 [110,113,115,116]. It encodes a centrosomal protein that localises to the connecting cilium of photoreceptors, where it plays an important role in protein trafficking between inner and outer segments [110,117]. Mutations in CEP290 disrupt ciliary function and protein transport, leading to photoreceptor degeneration [117].
Fogerty et al. [35] used scRNA-seq in zebrafish cep290 mutants, revealing that unlike typical zebrafish regenerative responses [118], Müller glia remained in rest-associated states despite active immune responses during progressive degeneration. Although regeneration-promoting genes stat3 and yap1 were upregulated, persistently high notch3 expression blocked Müller glia reprogramming, as Notch signalling must be downregulated for successful regeneration [119,120]. The study identified a novel Müller glia state with simultaneous quiescence and reactivity markers, suggesting that sustained Notch signalling prevents regeneration despite inflammatory signals and highlighting notch3 as a potential therapeutic target for CEP290-LCA10.
RPE65-associated LCA (LCA2) accounts for 4–16% of LCA cases [110,115,121,122,123] and 2% of arRP [121,124]. RPE65 encodes the retinal pigment epithelium-specific 65 kDa protein, a retinoid isomerase that is critical in the visual (retinoid) cycle [125]. Loss-of-function mutations in RPE65 disrupt the visual cycle, leading to childhood-onset retinal degeneration [126,127]. RPE65-associated LCA2 became the first IRD to receive approved gene therapy [128], but long-term studies have revealed continued retinal degeneration despite initial visual improvements [129]. Recent advances in base editing technology have enabled direct correction of these mutations in mouse models [36,130], offering a potentially more durable therapeutic approach than conventional gene augmentation strategies.
Choi et al. [36] demonstrated successful in vivo base editing correction of RPE65 mutations in LCA2 mice (rd12), with treated cones showing gene expression profiles more similar to wild-type. Single-cell analysis revealed restoration of key cone phototransduction genes and upregulated neuroprotective genes whilst showing downregulated cell death markers. Long-term follow-up at 6 months demonstrated sustained cone function and survival, indicating durable therapeutic correction and highlighting base editing’s potential for treating LCA2 and other IRDs.
Mutations in the cone–rod homeobox gene (CRX) represent a notable exception to the predominantly recessive forms of LCA discussed above. CRX mutations account for 0.6–2.8% of LCA [115,122,123] but can also cause CRD and MD [37]. Although CRX mutations primarily lead to autosomal dominant LCA7 [115,131,132], both dominant and recessive inheritance patterns have been reported [111,133]. CRX encodes a photoreceptor-specific transcription factor that is essential for both rod and cone photoreceptor development and function [116,131]. Mutations lead to early-onset severe visual impairment with functional deficits seen in ERGs [134].
Kruczek et al. [37] used patient-derived retinal organoids with scRNA-seq to study CRX-LCA7 and evaluate AAV-mediated gene augmentation therapy. Single-cell analysis revealed clear separation between patient and control populations, with AAV-treated cells exhibiting an intermediate gene expression pattern. Key photoreceptor genes were partially rescued upon treatment, with therapeutic effects persisting for 6 months without toxicity, demonstrating gene augmentation efficacy at the single-cell level.
Apart from the studies presented here, the study from Boon et al. [32] also investigated the CRB1 mutation that causes both arRP and LCA and was therefore presented earlier in the RP section. This study similarly combined scRNA-seq with AAV-mediated gene therapy, demonstrating the broader applicability of single-cell approaches for evaluating therapeutic interventions across multiple LCA subtypes. The integration of single-cell data with therapeutic interventions has proven particularly valuable in LCA research, enabling molecular assessment of treatment efficacy and identification of biomarkers for therapeutic response.

3.3. Enhanced S-Cone Syndrome

Enhanced S-cone syndrome (ESCS) is a rare IRD, characterised by an overabundance of the short-wavelength (S) cones alongside rod dysfunction [135,136,137].
It follows an autosomal recessive inheritance pattern [138,139], with typical symptoms including night blindness (nyctalopia) at an average age of 4 years, light sensitivity (photophobia), and colour vision anomalies, with increased sensitivity to blue light [139]. Patients’ ERGs have a characteristic pattern with reduced rod and cone responses but a strong S-cone response [136,138].
Known causative genes include NRL (neural retina leucine zipper) or CRX [140], but the most frequently implicated gene is NR2E3 (nuclear receptor subfamily 2, group E, member 3), which accounts for around 94% of ESCS cases [135]. The encoded NR2E3 protein is a photoreceptor-specific transcription factor that plays a crucial role in retinal development [140] by suppressing cone differentiation and promoting rod photoreceptor specification [141,142]. NR2E3 dysfunction causes rod precursors to develop into S-cone-like photoreceptors and become the predominant cone subtype [143].
Two studies published in 2024 have applied single-cell transcriptomics to better understand NR2E3-related pathogenesis.
Mullin et al. [38] used scRNA-seq on NR2E3 patient organoids across developmental stages (D40-D260), identifying D80 as the photoreceptor commitment point. They discovered novel “divergent rods” emerging at D120, unique to NR2E3-null organoids, which abnormally co-expressed rod and cone genes but lacked critical RHO expression despite normal NRL. This demonstrated that NR2E3 loss affects rod maturation rather than commitment. AAV gene therapy administered after divergent rod formation (D130-D160) showed only a 22% response rate, highlighting the need for early intervention before divergent rods emerge.
Complementing the human organoid study, Aísa-Marín et al. [39] employed two different Nr2e3 mutant mouse models, similarly identifying “hybrid” photoreceptors co-expressing rod and cone markers, particularly increased S-opsin, with decreased differentiated cones (70% to 20%) and increased hybrid cones (20% to 40–50%). Like Mullin et al. [38], they found altered differentiation pathways rather than complete fate conversion, as well as retinal remodelling responses and non-apoptotic cell death mechanisms including necroptosis and parthanatos. One major difference between both studies was retained rhodopsin expression in mouse rods (unlike complete absence in human organoids), likely reflecting hypomorphic rather than complete null mutations with residual NR2E3 protein function.
Together, these scRNA-seq studies have redefined ESCS as a disorder of photoreceptor maturation rather than commitment, revealing hybrid cell states and critical therapeutic windows to guide future gene therapy strategies.

3.4. Stargardt Disease

Stargardt disease (STGD) represents one of the most common inherited juvenile-onset MDs, with 5.5 million individuals affected worldwide [144,145].
The condition is inherited in an autosomal recessive manner, typically beginning during childhood [144,145], and is characterised by progressive central vision loss due to lipofuscin accumulation in the RPE and subsequent photoreceptor degeneration [146,147]. Other characteristics include deficiency in colour vision, central macular atrophy, and yellow-white flecks in the RPE [145].
More than 95% of STGD cases are caused by mutations in ABCA4 (ATP-binding cassette subfamily A member 4; STGD1) [148], which encodes a protein localised to the disc membranes of photoreceptors. This protein removes toxic retinal derivatives from disc membranes, enabling their clearance from the cell. When ABCA4 is defective, toxic compounds remain and react with membrane lipids, leading to lipofuscin accumulation in the RPE and subsequent progressive retinal degeneration [149,150].
Two 2025 studies used scRNA-seq in ABCA4 patient iPSC organoids to model STGD1, both finding photoreceptor structural alterations. Zhao et al. [41] conducted a longitudinal analysis across five developmental time points (D40-D260), showing cellular evolution from progenitors to rod/cone predominance and reduced cone populations with morphological abnormalities. Watson et al. [40] focused on late-stage organoids (D200), discovering spatial mislocalisation of photoreceptor cells when performing IHC, appearing in both basal and apical positions. Single-cell data showed phototransduction disruption, impaired maturation, and upregulated stress pathways but minimal apoptosis, suggesting that cellular stress rather than death drives mislocalisation. Both studies established valuable platforms for drug development and future STGD research.

3.5. Achromatopsia

Achromatopsia (ACHM) is a rare autosomal recessive IRD, in which cone dysfunction leads to severely reduced or absent colour vision, photophobia, nystagmus, and severely reduced visual acuity [151,152]. In the majority of ACHM cases, cones are present in the retina but lack the ability to respond to light [153]. It typically presents at birth or early infancy and affects approximately 1 in 30,000–50,000 individuals worldwide [151,152,154].
Most ACHM cases (up to 70–90%) are due to mutations in CNGA3 or CNGB3, whilst fewer than 10% of cases are caused by mutations in GNAT2, PDE6C, PDE6H, and ATF6 [151,155,156,157]. Although PDE6C represents one of the less common genetic causes of ACHM (1.4–2.4% [154,158]), its study provides crucial insights into cone phototransduction mechanisms. The gene encodes the catalytic alpha subunit of cone-specific phosphodiesterase, an enzyme responsible for hydrolysing cGMP to terminate the phototransduction signal and restore the cone to its dark-adapted state [158]. Mutations in PDE6C disrupt this critical step in the phototransduction cascade, resulting in loss or dysfunction of cones and the typical ACHM phenotype [158]. Given that PDE6C operates within the same functional pathway as CNGA3, CNGB3, GNAT2, and PDE6H [152], therapeutic interventions targeting this mechanism may offer benefits across multiple genetic forms of ACHM.
Our literature search revealed only one study that examined diseased ACHM samples using single-cell transcriptomics. Miller et al. [42] investigated HDAC inhibition effects on cone degeneration in ACHM, discovering that diseased cones had decreased H3K27me3 (trimethylation of histone H3 protein at lysine 27 residue) marks. They tested histone H3 lysine 27 (H3K27) demethylase inhibition in Pde6c-mutant mice, with scRNA-seq revealing beneficial transcriptional changes, including suppressed oxidative phosphorylation, mitochondrial dysfunction, and ER stress pathways. Despite improved transcriptional changes, single injection showed no histological improvement, whilst continuous treatment in retinal explants increased cone survival and improved opsin localisation, indicating that sustained H3K27 demethylase inhibition is required for cone protection.

4. Common Themes and Therapeutic Implications

We organised the studies by disease type and genetic mutation for presentation (Figure 1), recognising that certain mutations may be associated with overlapping disease phenotypes. Our literature search identified 15 studies investigating mutations associated with RP, comprising 4 studies of RHO mutations, 3 of PDE6B mutations, 2 of RPGR mutations, and 6 examining other RP-related mutations. Additionally, three studies investigated LCA-associated mutations, whilst two studies each examined ESCS- and STGD-associated mutations, and one study focused on an ACHM-associated mutation. Most studies were performed in the USA (9 out of 23), followed by China (5 studies) and the UK (4 studies), whilst the Netherlands, Australia, Japan, Spain, and Switzerland each contributed 1 study. In terms of model systems, 12 studies used mouse models, 2 used zebrafish, and 9 studied human iPSC-derived retinal organoids. The majority employed 10x Genomics technology for their studies (19 out of 23), consistent with its robust performance profile amongst high-throughput single-cell platforms [159].
Single-cell transcriptomics has become a valuable approach for investigating IRD pathogenesis. By utilising its cellular-level resolution, several studies have elucidated how individual retinal cell populations respond at various stages of the degenerative process. Across diverse IRDs, scRNA-seq consistently revealed cell-type-specific disease mechanisms and heterogeneity (Figure 2), demonstrating how multiple retinal cell types respond differently at various stages of degeneration.
Single-cell approaches confirmed photoreceptor degeneration with deficits in phototransduction genes across models. Misfolded protein accumulation, heightened ER stress, and metabolic dysfunction emerged in degenerating photoreceptors across multiple models, leading to cell-intrinsic stress responses. These cellular stress responses triggered inflammatory cascades demonstrated across multiple studies, often involving microglia activation and Müller glia reactivity. Studies consistently revealed excessive immune-related signalling and inflammation driven by glial cells. Multiple studies demonstrated the dual capacity of Müller glia to shift between neuroprotective and proinflammatory roles. Understanding glial activity is therefore crucial for therapeutic approaches.
Multiple studies combined scRNA-seq with treatments and showed rescue of photoreceptor gene expression, improved survival, and reduced markers of stress or inflammation. Several research groups highlight the importance of early interventions for better outcomes. Single-cell analyses have identified novel therapeutic targets, including specific pathways and cell-type-specific vulnerabilities that could guide precision medicine approaches.

5. Challenges and Future Considerations

The field has benefited from rapid advances in single-cell platforms, yet the extensive genetic heterogeneity underlying IRDs, with hundreds of causative mutations, combined with variable disease onset and progression, creates substantial analytical complexity.
Retinal tissue processing presents unique challenges due to the elaborate morphology of photoreceptors and other retinal cells, making them particularly vulnerable during dissociation protocols. Given the extensive use of mouse models for single-cell transcriptomics in IRD research (12 out of 23 studies), small tissue samples such as mouse retinas present further complications due to low RNA yields, which can introduce sampling bias and reduce the reliability of gene expression measurements [160,161]. A technical optimisation is to increase the number of polymerase chain reaction (PCR) cycles during complementary DNA (cDNA) amplification, although this might increase PCR artifacts [161]. To address the challenges of low-input samples, several alternative scRNA-seq platforms have been developed that are especially well-suited for low-yield samples [162], such as the deterministic, mRNA-capture bead and cell co-encapsulation dropleting system, DisCo [163], and the open-source hydrogel droplet platform, HyDrop [164]. Additionally, increasing the number of biological replicates enables validation of cell clusters based on their reproducibility across samples, thereby preventing over-interpretation of clusters that may arise from technical variation [165]. To address sample preservation challenges, methodological innovations such as methanol fixation protocols have been developed to facilitate sample transport and storage whilst maintaining RNA integrity [166,167]. Single-nucleus RNA-seq represents an alternative approach that can be applied to frozen tissues [168]. Established protocols [166,167] are essential for minimising cell death and preserving cellular integrity. Batch effects can obscure genuine biological differences, and whilst computational tools can partially address technical variation [12], multiple biological replicates are essential for a robust approach, though this is often constrained by the high cost of single-cell experiments. Statistical interpretation requires careful consideration, as the large number of cells per condition can artificially inflate statistical power, potentially detecting differences that may not represent reproducible biological variation, particularly in studies with limited biological replicates [169,170,171]. Cell abundance interpretations require particular caution, as scRNA-seq-derived cell numbers may not accurately reflect the original tissue composition due to processing biases during tissue dissociation, technical variability, and quality control filtering steps [168] and should be validated through complementary methods such as flow cytometry or imaging.
Analysing scRNA-seq data presents challenges due to sparse gene expression, technical noise, and the complexity of heterogeneous cell populations [12]. Fortunately, sophisticated computational frameworks address these complexities, including the widely used Seurat pipeline [172,173] and numerous specialised packages within the Bioconductor project [174,175]. This open-source ecosystem allows researchers to inspect and modify analytical code, promoting transparency and reproducibility in single-cell research.

6. Conclusions

Although the application of single-cell transcriptomics remains relatively limited in IRD research, the field is rapidly evolving and showing considerable promise. The IRD studies presented in this review revealed the cellular complexity of the retinal tissue and demonstrated how single-cell approaches enabled the detection of early molecular changes preceding cell death. Across multiple IRD studies, researchers have identified common pathomechanisms, including disrupted phototransduction cascades, altered metabolic pathways, and compromised cellular stress responses across retinal cell types, with Müller glia consistently playing a crucial role during retinal degeneration.
These findings demonstrate how single-cell transcriptomics is advancing both mechanistic understanding and therapeutic development for IRDs. Future studies should consider combining single-cell and bulk RNA-seq to leverage the complementary strengths of both techniques, as bulk RNA-seq generally provides a higher sequencing depth per sample, while single-cell approaches reveal cellular heterogeneity [176,177]. Innovative analytical approaches, such as the deconvolution method applied by Lyu et al. [178] to bulk RNA-seq data using scRNA-seq references, demonstrate how these techniques can be combined to estimate cell-type proportions within bulk profiles.
Emerging advances in spatial multi-omics approaches promise to preserve tissue architecture whilst providing single-cell resolution, enabling researchers to understand not only gene expression in individual cells, but also how spatial organisation and cell–cell interactions contribute to disease progression [179,180,181]. Computational methods are now being developed to integrate scRNA-seq data with spatial transcriptomics data, offering powerful approaches to understand dynamic changes in complex tissue architecture during disease progression [182,183]. These integrated analytical frameworks are particularly valuable for IRDs, where understanding the spatial and temporal context of cellular dysfunction, such as the progression of photoreceptor degeneration from specific retinal regions or the spatial patterns of glial activation over time, provides crucial insights into disease mechanisms and optimal intervention points [184,185]. Moreover, spatial transcriptomics shows particular promise for evaluating therapeutic interventions, as it can capture regional variations in treatment efficacy and identify areas of the retina that respond differentially to gene therapy or pharmacological treatments over time [184,186]. These insights can inform optimisation of delivery and dosing strategies, ultimately making therapies more precisely targeted and potentially more effective. As scRNA-seq becomes increasingly accessible, its integration with established genetic and clinical knowledge will enhance our understanding of disease processes and enable more precise therapeutic interventions. The field would benefit from broader adoption of single-cell approaches in IRD research, supported by collaborative efforts to improve data sharing that enable large-scale integrative analyses.

Author Contributions

L.N. conceptualised this review, conducted the literature search, performed data extraction, and wrote the original manuscript. R.M., P.M. and C.A.V. provided supervision, reviewed and edited the manuscript, and contributed to the final approval of the version for publication. All authors have read and agreed to the published version of the manuscript.

Funding

L.N. is funded by the Medical Research Council (MR/N013166/1). R.M. is funded by the Wellcome Trust (219607/Z/19/Z). P.M. is funded by the UKRI Medical Research Council (MR/Y015002/1 and MC/PC/21044) and the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement n°866355).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the writing of the manuscript.

Abbreviations

The following abbreviations are used in this manuscript:
AAVAdeno-associated virus
ACHMAchromatopsia
adRPAutosomal dominant retinitis pigmentosa
Ca2+Calcium ion
CDCone dystrophy
CNG channelsCyclic nucleotide-gated channels
CRDCone–rod dystrophy
CTRLControl
CSNBCongenital stationary night blindness
DDay
EGR1Early growth response 1
EREndoplasmic reticulum
ERGElectroretinography
ERKExtracellular signal-regulated kinase
ESCSEnhanced S-cone syndrome
FGF21Fibroblast growth factor 21
GSE IDGene expression omnibus series identifier
H3K27Histone H3 lysine 27
HDACHistone deacetylase
HETHeterozygous
IFImmunofluorescence
IHCImmunohistochemistry
iPSCsInduced pluripotent stem cells
IRDInherited retinal dystrophy
JNKc-Jun N-terminal kinase
LCALeber congenital amaurosis
MAPKMitogen-activated protein kinase
MDMacular dystrophy
MeSHMedical Subject Headings
MUTMutant
NCXSodium–calcium exchanger
PPostnatal day
PARPPoly (ADP-ribose) polymerase
PCRPolymerase chain reaction
PKGProtein kinase G
PWPostnatal week
rdRetinal degeneration
RNA-seqRNA sequencing
rOrgRetinal organoid
RPRetinitis pigmentosa
RPERetinal pigment epithelium
S-coneShort-wavelength cone
scRNA-seqSingle-cell RNA sequencing
STGDStargardt disease
USHUsher syndrome
VVersion
VGCCVoltage-gated calcium channel
vs.Versus
WTWild-type
XLRPX-linked retinitis pigmentosa

References

  1. Sahel, J.A.; Marazova, K.; Audo, I. Clinical characteristics and current therapies for inherited retinal degenerations. Cold Spring Harb. Perspect. Med. 2014, 5, a017111. [Google Scholar] [CrossRef]
  2. Wright, A.F.; Chakarova, C.F.; Abd El-Aziz, M.M.; Bhattacharya, S.S. Photoreceptor degeneration: Genetic and mechanistic dissection of a complex trait. Nat. Rev. Genet. 2010, 11, 273–284. [Google Scholar] [CrossRef]
  3. Hartong, D.T.; Berson, E.L.; Dryja, T.P. Retinitis pigmentosa. Lancet 2006, 368, 1795–1809. [Google Scholar] [CrossRef]
  4. RetNet, the Retinal Information Network. Available online: https://RetNet.org/ (accessed on 11 July 2025).
  5. Verbakel, S.K.; van Huet, R.A.C.; Boon, C.J.F.; den Hollander, A.I.; Collin, R.W.J.; Klaver, C.C.W.; Hoyng, C.B.; Roepman, R.; Klevering, B.J. Non-syndromic retinitis pigmentosa. Prog. Retin. Eye Res. 2018, 66, 157–186. [Google Scholar] [CrossRef] [PubMed]
  6. Yu, X.; Shi, W.; Cheng, L.; Wang, Y.; Chen, D.; Hu, X.; Xu, J.; Xu, L.; Wu, Y.; Qu, J.; et al. Identification of a rhodopsin gene mutation in a large family with autosomal dominant retinitis pigmentosa. Sci. Rep. 2016, 6, 19759. [Google Scholar] [CrossRef] [PubMed]
  7. Martínez-Gil, N.; Maneu, V.; Kutsyr, O.; Fernández-Sánchez, L.; Sánchez-Sáez, X.; Sánchez-Castillo, C.; Campello, L.; Lax, P.; Pinilla, I.; Cuenca, N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front. Neuroanat. 2022, 16, 984052. [Google Scholar] [CrossRef] [PubMed]
  8. Zhao, L.; Zabel, M.K.; Wang, X.; Ma, W.; Shah, P.; Fariss, R.N.; Qian, H.; Parkhurst, C.N.; Gan, W.B.; Wong, W.T. Microglial phagocytosis of living photoreceptors contributes to inherited retinal degeneration. EMBO Mol. Med. 2015, 7, 1179–1197. [Google Scholar] [CrossRef]
  9. Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef]
  10. Menon, M.; Mohammadi, S.; Davila-Velderrain, J.; Goods, B.A.; Cadwell, T.D.; Xing, Y.; Stemmer-Rachamimov, A.; Shalek, A.K.; Love, J.C.; Kellis, M.; et al. Single-cell transcriptomic atlas of the human retina identifies cell types associated with age-related macular degeneration. Nat. Commun. 2019, 10, 4902. [Google Scholar]
  11. Voigt, A.P.; Mullin, N.K.; Stone, E.M.; Tucker, B.A.; Scheetz, T.E.; Mullins, R.F. Single-cell RNA sequencing in vision research: Insights into human retinal health and disease. Prog. Retin. Eye Res. 2021, 83, 100934. [Google Scholar] [CrossRef]
  12. Tzec-Interián, J.A.; González-Padilla, D.; Góngora-Castillo, E.B. Bioinformatics perspectives on transcriptomics: A comprehensive review of bulk and single-cell RNA sequencing analyses. Quant. Biol. 2025, 13, e78. [Google Scholar] [CrossRef]
  13. Nguyen, A.; Khoo, W.H.; Moran, I.; Croucher, P.I.; Phan, T.G. Single Cell RNA Sequencing of Rare Immune Cell Populations. Front. Immunol. 2018, 9, 1553. [Google Scholar] [CrossRef] [PubMed]
  14. Shekhar, K.; Whitney, I.E.; Butrus, S.; Peng, Y.-R.; Sanes, J.R. Diversification of multipotential postmitotic mouse retinal ganglion cell precursors into discrete types. eLife 2022, 11, e73809. [Google Scholar] [CrossRef] [PubMed]
  15. Macosko, E.Z.; Basu, A.; Satija, R.; Nemesh, J.; Shekhar, K.; Goldman, M.; Tirosh, I.; Bialas, A.R.; Kamitaki, N.; Martersteck, E.M.; et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell 2015, 161, 1202–1214. [Google Scholar] [CrossRef]
  16. Tomita, Y.; Qiu, C.; Bull, E.; Allen, W.; Kotoda, Y.; Talukdar, S.; Smith, L.E.H.; Fu, Z. Müller glial responses compensate for degenerating photoreceptors in retinitis pigmentosa. Exp. Mol. Med. 2021, 53, 1748–1758. [Google Scholar] [CrossRef]
  17. Fu, Z.; Qiu, C.; Cagnone, G.; Tomita, Y.; Huang, S.; Cakir, B.; Kotoda, Y.; Allen, W.; Bull, E.; Akula, J.D.; et al. Retinal glial remodeling by FGF21 preserves retinal function during photoreceptor degeneration. iScience 2021, 24, 102376. [Google Scholar] [CrossRef]
  18. Santhanam, A.; Shihabeddin, E.; Wei, H.; Wu, J.; O’Brien, J. Molecular basis of retinal remodeling in a zebrafish model of retinitis pigmentosa. Cell. Mol. Life Sci. 2023, 80, 362. [Google Scholar] [CrossRef]
  19. Lin, X.; Liu, Z.L.; Zhang, X.; Wang, W.; Huang, Z.Q.; Sun, S.N.; Jin, Z.B. Modeling autosomal dominant retinitis pigmentosa by using patient-specific retinal organoids with a class-3 RHO mutation. Exp. Eye Res. 2024, 241, 109856. [Google Scholar] [CrossRef]
  20. Chen, Y.; Dong, Y.; Yan, J.; Wang, L.; Yu, S.; Jiao, K.; Paquet-Durand, F. Single-Cell Transcriptomic Profiling in Inherited Retinal Degeneration Reveals Distinct Metabolic Pathways in Rod and Cone Photoreceptors. Int. J. Mol. Sci. 2022, 23, 12170. [Google Scholar] [CrossRef]
  21. Dong, Y.; Xu, W.; Li, Y.; Wei, C.; Hu, Y.; Hu, Z.; Paquet-Durand, F.; Jiao, K. Inhibition of the MAPK/c-Jun-EGR1 Pathway Decreases Photoreceptor Cell Death in the Rd1 Mouse Model Inherit. Retin. Degeneration. Int. J. Mol. Sci. 2022, 23, 14600. [Google Scholar] [CrossRef]
  22. Dong, Y.; Yan, J.; Xu, W.; Paquet-Durand, F.; Hu, Z.; Jiao, K. HDAC inhibition delays photoreceptor loss in Pde6b Mutant Mice Retin. Pigment. Insights ScRNA-Seq CUT&Tag. PeerJ 2023, 11, e15659. [Google Scholar] [PubMed]
  23. Yan, J.; Wang, L.; Yang, Q.L.; Yang, Q.X.; He, X.; Dong, Y.; Hu, Z.; Seeliger, M.W.; Jiao, K.; Paquet-Durand, F. T-type voltage-gated channels, Na+/Ca2+-exchanger, and calpain-2 promote photoreceptor cell death in inherited retinal degeneration. Cell Commun. Signal. 2024, 22, 92. [Google Scholar] [CrossRef] [PubMed]
  24. Liao, C.; Chen, S.; Chen, X.; Yi, W.; Fan, Y.; Chen, Y.; Ye, T.; Chen, Y. Inhibition of JNK ameliorates rod photoreceptor degeneration in a mouse model of retinitis pigmentosa. FEBS Lett. 2024, 598, 2683–2701. [Google Scholar] [CrossRef] [PubMed]
  25. Karademir, D.; Todorova, V.; Ebner, L.J.A.; Samardzija, M.; Grimm, C. Single-cell RNA sequencing of the retina in a model of retinitis pigmentosa reveals early responses to degeneration in rods and cones. BMC Biol. 2022, 20, 86. [Google Scholar] [CrossRef]
  26. Sigurdsson, D.; Grimm, C. Single-Cell Transcriptomic Profiling of Müller Glia in the rd10 Retina. Adv. Exp. Med. Biol. 2023, 1415, 377–381. [Google Scholar]
  27. 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, 4024. [Google Scholar] [CrossRef]
  28. Li, T.; Ma, Y.; Cheng, Y.; Zhao, Y.; Qiu, Z.; Liu, H.; Zhang, D.; Wu, J.; Li, J.; Zhang, S.; et al. Single-Cell Transcriptomic Dataset of RPGR-associated Retinitis Pigmentosa Patient-Derived Retinal Organoids. Sci. Data 2024, 11, 1285. [Google Scholar] [CrossRef]
  29. Leong, Y.C.; Di Foggia, V.; Pramod, H.; Bitner-Glindzicz, M.; Patel, A.; Sowden, J.C. Molecular pathology of Usher 1B patient-derived retinal organoids at single cell resolution. Stem Cell Rep. 2022, 17, 2421–2437. [Google Scholar] [CrossRef]
  30. Bertrand, R.E.; Wang, J.; Li, Y.; Cheng, X.; Wang, K.; Stoilov, P.; Chen, R. Cwc27, associated with retinal degeneration, functions as a splicing factor in Vivo. Hum. Mol. Genet. 2022, 31, 1278–1292. [Google Scholar] [CrossRef]
  31. Atkinson, R.; Georgiou, M.; Yang, C.; Szymanska, K.; Lahat, A.; Vasconcelos, E.J.R.; Ji, Y.; Moya Molina, M.; Collin, J.; Queen, R.; et al. PRPF8-mediated dysregulation of hBrr2 helicase disrupts human spliceosome kinetics and 5-splice-site selection causing tissue-specific defects. Nat. Commun. 2024, 15, 3138. [Google Scholar] [CrossRef]
  32. Boon, N.; Lu, X.; Andriessen, C.A.; Moustakas, I.; Buck, T.M.; Freund, C.; Arendzen, C.H.; Böhringer, S.; Mei, H.; Wijnholds, J. AAV-mediated gene augmentation therapy of CRB1 patient-derived retinal organoids restores the histological and transcriptional retinal phenotype. Stem Cell Rep. 2023, 18, 1123–1137. [Google Scholar] [CrossRef] [PubMed]
  33. Shigesada, N.; Shikada, N.; Shirai, M.; Toriyama, M.; Higashijima, F.; Kimura, K.; Kondo, T.; Bessho, Y.; Shinozuka, T.; Sasai, N. Combination of blockade of endothelin signalling and compensation of IGF1 expression protects the retina from degeneration. Cell. Mol. Life Sci. 2024, 81, 51. [Google Scholar] [CrossRef]
  34. Lewandowski, D.; Foik, A.T.; Smidak, R.; Choi, E.H.; Zhang, J.; Hoang, T.; Tworak, A.; Suh, S.; Leinonen, H.; Dong, Z.; et al. Inhibition of ceramide accumulation in AdipoR1-/- mice increases photoreceptor survival and improves vision. JCI Insight 2022, 7, e156301. [Google Scholar] [CrossRef] [PubMed]
  35. Fogerty, J.; Song, P.; Boyd, P.; Grabinski, S.E.; Hoang, T.; Reich, A.; Cianciolo, L.T.; Blackshaw, S.; Mumm, J.S.; Hyde, D.R.; et al. Notch Inhibition Promotes Regeneration and Immunosuppression Supports Cone Survival in a Zebrafish Model of Inherited Retinal Dystrophy. J. Neurosci. 2022, 42, 5144–5158. [Google Scholar] [CrossRef] [PubMed]
  36. Choi, E.H.; Suh, S.; Foik, A.T.; Leinonen, H.; Newby, G.A.; Gao, X.D.; Banskota, S.; Hoang, T.; Du, S.W.; Dong, Z.; et al. In Vivo Base Editing Rescues Cone Photoreceptors A Mouse Model of Early-Onset Inherited Retinal Degeneration. Nat. Commun. 2022, 13, 1830. [Google Scholar] [CrossRef]
  37. Kruczek, K.; Qu, Z.; Gentry, J.; Fadl, B.R.; Gieser, L.; Hiriyanna, S.; Batz, Z.; Samant, M.; Samanta, A.; Chu, C.J.; et al. Gene Therapy of Dominant CRX-Leber Congenital Amaurosis using Patient Stem Cell-Derived Retinal Organoids. Stem Cell Rep. 2021, 16, 252–263. [Google Scholar] [CrossRef]
  38. Mullin, N.K.; Bohrer, L.R.; Voigt, A.P.; Lozano, L.P.; Wright, A.T.; Bonilha, V.L.; Mullins, R.F.; Stone, E.M.; Tucker, B.A. NR2E3 loss disrupts photoreceptor cell maturation and fate in human organoid models of retinal development. J. Clin. Investig. 2024, 134, e173892. [Google Scholar] [CrossRef]
  39. Aísa-Marín, I.; Rovira, Q.; Díaz, N.; Calvo-López, L.; Vaquerizas, J.M.; Marfany, G. Specific photoreceptor cell fate pathways are differentially altered in NR2E3-associated diseases. Neurobiol. Dis. 2024, 194, 106463. [Google Scholar] [CrossRef]
  40. Watson, A.; Queen, R.; Ferrández-Peral, L.; Dorgau, B.; Collin, J.; Nelson, A.; Hussain, R.; Coxhead, J.; McCorkindale, M.; Atkinson, R.; et al. Unravelling genotype-phenotype correlations in Stargardt disease using patient-derived retinal organoids. Cell Death Dis. 2025, 16, 108. [Google Scholar] [CrossRef]
  41. Zhao, Y.; Cheng, Y.; Li, T.; Wu, J.; Li, C.; Zhang, S.; Wu, J. Longitudinal scRNA-seq of retinal organoids derived from Stargardt disease patient with ABCA4 mutation. Sci. Data 2025, 12, 878. [Google Scholar] [CrossRef]
  42. Miller, A.L.; Fuller-Carter, P.I.; Masarini, K.; Samardzija, M.; Carter, K.W.; Rashwan, R.; Lim, X.R.; Brunet, A.A.; Chopra, A.; Ram, R.; et al. Increased H3K27 trimethylation contributes to cone survival in a mouse model of cone dystrophy. Cell. Mol. Life Sci. 2022, 79, 409. [Google Scholar] [CrossRef]
  43. Cross, N.; van Steen, C.; Zegaoui, Y.; Satherley, A.; Angelillo, L. Retinitis Pigmentosa: Burden of Disease and Current Unmet Needs. Clin. Ophthalmol. 2022, 16, 1993–2010. [Google Scholar] [CrossRef]
  44. Dryja, T.P.; McGee, T.L.; Hahn, L.B.; Cowley, G.S.; Olsson, J.E.; Reichel, E.; Sandberg, M.A.; Berson, E.L. Mutations within the rhodopsin gene in patients with autosomal dominant retinitis pigmentosa. N. Engl. J. Med. 1990, 323, 1302–1307. [Google Scholar] [CrossRef]
  45. Zhen, F.; Zou, T.; Wang, T.; Zhou, Y.; Dong, S.; Zhang, H. Rhodopsin-associated retinal dystrophy: Disease mechanisms and therapeutic strategies. Front. Neurosci. 2023, 17, 1132179. [Google Scholar] [CrossRef]
  46. Yang, C.; Georgiou, M.; Atkinson, R.; Collin, J.; Al-Aama, J.; Nagaraja-Grellscheid, S.; Johnson, C.; Ali, R.; Armstrong, L.; Mozaffari-Jovin, S.; et al. Pre-mRNA Processing Factors and Retinitis Pigmentosa: RNA Splicing and Beyond. Front. Cell Dev. Biol. 2021, 9, 700276. [Google Scholar] [CrossRef] [PubMed]
  47. Wu, Y.; Guo, Y.; Yi, J.; Xu, H.; Yuan, L.; Yang, Z.; Deng, H. Heterozygous RHO P.R135W Missense Mutat. A Large Han-Chin. Fam. Retin. Pigment. Differ. Refract. Errors. Biosci. Rep. 2019, 39, BSR20182198. [Google Scholar] [CrossRef] [PubMed]
  48. Mendes, H.F.; van der Spuy, J.; Chapple, J.P.; Cheetham, M.E. Mechanisms of cell death in rhodopsin retinitis pigmentosa: Implications for therapy. Trends Mol. Med. 2005, 11, 177–185. [Google Scholar] [CrossRef] [PubMed]
  49. Yu, Y.; Xia, X.; Li, H.; Zhang, Y.; Zhou, X.; Jiang, H. A new rhodopsin R135W mutation induces endoplasmic reticulum stress and apoptosis in retinal pigment epithelial cells. J. Cell. Physiol. 2019, 234, 14100–14108. [Google Scholar] [CrossRef]
  50. Khramtsov, N.V.; Feshchenko, E.A.; Suslova, V.A.; Shmukler, B.E.; Terpugov, B.E.; Rakitina, T.V.; Atabekova, N.V.; Lipkin, V.M. The human rod photoreceptor cGMP phosphodiesterase beta-subunit. Structural studies of its cDNA and gene. FEBS Lett. 1993, 327, 275–278. [Google Scholar] [CrossRef]
  51. Huang, S.H.; Pittler, S.J.; Huang, X.; Oliveira, L.; Berson, E.L.; Dryja, T.P. Autosomal recessive retinitis pigmentosa caused by mutations in the alpha subunit of rod cGMP phosphodiesterase. Nat. Genet. 1995, 11, 468–471. [Google Scholar] [CrossRef]
  52. Li, Y.; Li, R.; Dai, H.; Li, G. Novel variants in PDE6A and PDE6B genes and its phenotypes in patients with retinitis pigmentosa in Chinese families. BMC Ophthalmol. 2022, 22, 27. [Google Scholar] [CrossRef]
  53. Mittal, R.; Bencie, N.; Parrish, J.M.; Liu, G.; Mittal, J.; Yan, D.; Liu, X.Z. An Update on Phosphodiesterase Mutations Underlying Genetic Etiology of Hearing Loss and Retinitis Pigmentosa. Front. Genet. 2018, 9, 9. [Google Scholar] [CrossRef]
  54. Kuehlewein, L.; Zobor, D.; Stingl, K.; Kempf, M.; Nasser, F.; Bernd, A.; Biskup, S.; Cremers, F.P.M.; Khan, M.I.; Mazzola, P.; et al. Clinical Phenotype of PDE6B-Assoc. Retin. Pigmentosa. Int. J. Mol. Sci. 2021, 22, 2374. [Google Scholar] [CrossRef]
  55. Caley, D.W.; Johnson, C.; Liebelt, R.A. The postnatal development of the retina in the normal and rodless CBA mouse: A light and electron microscopic study. Am. J. Anat. 1972, 133, 179–212. [Google Scholar] [CrossRef] [PubMed]
  56. LaVail, M.M.; Sidman, R.L. C57BL-6J mice with inherited retinal degeneration. Arch. Ophthalmol. 1974, 91, 394–400. [Google Scholar] [CrossRef] [PubMed]
  57. Carter-Dawson, L.D.; LaVail, M.M.; Sidman, R.L. Differential effect of the rd mutation on rods and cones in the mouse retina. Investig. Ophthalmol. Vis. Sci. 1978, 17, 489–498. [Google Scholar]
  58. Samardzija, M.; Wariwoda, H.; Imsand, C.; Huber, P.; Heynen, S.R.; Gubler, A.; Grimm, C. Activation of survival pathways in the degenerating retina of rd10 mice. Exp. Eye Res. 2012, 99, 17–26. [Google Scholar] [CrossRef]
  59. Gargini, C.; Terzibasi, E.; Mazzoni, F.; Strettoi, E. Retinal organization in the retinal degeneration 10 (rd10) mutant mouse: A morphological and ERG study. J. Comp. Neurol. 2007, 500, 222–238. [Google Scholar] [CrossRef]
  60. Barhoum, R.; Martínez-Navarrete, G.; Corrochano, S.; Germain, F.; Fernandez-Sanchez, L.; de la Rosa, E.J.; de la Villa, P.; Cuenca, N. Functional and structural modifications during retinal degeneration in the rd10 mouse. Neuroscience 2008, 155, 698–713. [Google Scholar] [CrossRef]
  61. Scimeca, J.C.; Servant, M.J.; Dyer, J.O.; Meloche, S. Essential role of calcium in the regulation of MAP kinase phosphatase-1 expression. Oncogene 1997, 15, 717–725. [Google Scholar] [CrossRef]
  62. Papa, S.; Choy, P.M.; Bubici, C. The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene 2019, 38, 2223–2240. [Google Scholar] [CrossRef]
  63. Hoffmann, E.; Ashouri, J.; Wolter, S.; Doerrie, A.; Dittrich-Breiholz, O.; Schneider, H.; Wagner, E.F.; Troppmair, J.; Mackman, N.; Kracht, M. Transcriptional regulation of EGR-1 by the interleukin-1-JNK-MKK7-c-Jun pathway. J. Biol. Chem. 2008, 283, 12120–12128. [Google Scholar] [CrossRef] [PubMed]
  64. Shu, X.; Black, G.C.; Rice, J.M.; Hart-Holden, N.; Jones, A.; O’Grady, A.; Ramsden, S.; Wright, A.F. RPGR mutation analysis and disease: An update. Hum. Mutat. 2007, 28, 322–328. [Google Scholar] [CrossRef] [PubMed]
  65. Megaw, R.D.; Soares, D.C.; Wright, A.F. RPGR: Its role in photoreceptor physiology, human disease, and future therapies. Exp. Eye Res. 2015, 138, 32–41. [Google Scholar] [CrossRef] [PubMed]
  66. Cehajic Kapetanovic, J.; McClements, M.E.; Martinez-Fernandez de la Camara, C.; MacLaren, R.E. Molecular Strategies for RPGR Gene Therapy. Genes 2019, 10, 674. [Google Scholar] [CrossRef]
  67. Megaw, R.; Abu-Arafeh, H.; Jungnickel, M.; Mellough, C.; Gurniak, C.; Witke, W.; Zhang, W.; Khanna, H.; Mill, P.; Dhillon, B.; et al. Gelsolin dysfunction causes photoreceptor loss in induced pluripotent cell and animal retinitis pigmentosa models. Nat. Commun. 2017, 8, 271. [Google Scholar] [CrossRef]
  68. Megaw, R.; Moye, A.; Zhang, Z.; Newton, F.; McPhie, F.; Murphy, L.C.; McKie, L.; He, F.; Jungnickel, M.K.; von Kriegsheim, A.; et al. Ciliary tip actin dynamics regulate photoreceptor outer segment integrity. Nat. Commun. 2024, 15, 4316. [Google Scholar] [CrossRef]
  69. Birch, D.G.; Cheetham, J.K.; Daiger, S.P.; Hoyng, C.; Kay, C.; MacDonald, I.M.; Pennesi, M.E.; Sullivan, L.S. Overcoming the Challenges to Clinical Development of X-Linked Retinitis Pigmentosa Therapies: Proceedings of an Expert Panel. Transl. Vis. Sci. Technol. 2023, 12, 5. [Google Scholar] [CrossRef]
  70. Sandberg, M.A.; Rosner, B.; Weigel-DiFranco, C.; Dryja, T.P.; Berson, E.L. Disease course of patients with X-linked retinitis pigmentosa due to RPGR gene mutations. Investig. Ophthalmol. Vis. Sci. 2007, 48, 1298–1304. [Google Scholar] [CrossRef]
  71. Zito, I.; Downes, S.M.; Patel, R.J.; Cheetham, M.E.; Ebenezer, N.D.; Jenkins, S.A.; Bhattacharya, S.S.; Webster, A.R.; Holder, G.E.; Bird, A.C.; et al. RPGR mutation associated with retinitis pigmentosa, impaired hearing, and sinorespiratory infections. J. Med. Genet. 2003, 40, 609–615. [Google Scholar] [CrossRef]
  72. Iannaccone, A.; Breuer, D.K.; Wang, X.F.; Kuo, S.F.; Normando, E.M.; Filippova, E.; Baldi, A.; Hiriyanna, S.; MacDonald, C.B.; Baldi, F.; et al. Clinical and immunohistochemical evidence for an X linked retinitis pigmentosa syndrome with recurrent infections and hearing loss in association with an RPGR mutation. J. Med. Genet. 2003, 40, e118. [Google Scholar] [CrossRef] [PubMed]
  73. Demirci, F.Y.; Rigatti, B.W.; Wen, G.; Radak, A.L.; Mah, T.S.; Baic, C.L.; Traboulsi, E.I.; Alitalo, T.; Ramser, J.; Gorin, M.B. X-linked cone-rod dystrophy (locus COD1): Identification of mutations in RPGR exon ORF15. Am. J. Hum. Genet. 2002, 70, 1049–1053. [Google Scholar] [CrossRef] [PubMed]
  74. Ebenezer, N.D.; Michaelides, M.; Jenkins, S.A.; Audo, I.; Webster, A.R.; Cheetham, M.E.; Stockman, A.; Maher, E.R.; Ainsworth, J.R.; Yates, J.R.; et al. Identification of novel RPGR ORF15 mutations in X-linked progressive cone-rod dystrophy (XLCORD) families. Investig. Ophthalmol. Vis. Sci. 2005, 46, 1891–1898. [Google Scholar] [CrossRef] [PubMed]
  75. Robson, A.G.; Michaelides, M.; Luong, V.A.; Holder, G.E.; Bird, A.C.; Webster, A.R.; Moore, A.T.; Fitzke, F.W. Functional correlates of fundus autofluorescence abnormalities in patients with RPGR or RIMS1 mutations causing cone or cone rod dystrophy. Br. J. Ophthalmol. 2008, 92, 95–102. [Google Scholar] [CrossRef]
  76. Birtel, J.; Eisenberger, T.; Gliem, M.; Müller, P.L.; Herrmann, P.; Betz, C.; Zahnleiter, D.; Neuhaus, C.; Lenzner, S.; Holz, F.G.; et al. Clinical and genetic characteristics of 251 consecutive patients with macular and cone/cone-rod dystrophy. Sci. Rep. 2018, 8, 4824. [Google Scholar] [CrossRef]
  77. Wu, Y.; Tavares, E.; Liang, B.; Wee, W.; Mennella, V.; Feng, H.; Cao, J.; Zheng, J.; He, M.; Stephenson, K.; et al. RPGR regulates motile cilia by interfering with actin dynamics. bioRxiv 2025. [Google Scholar] [CrossRef]
  78. D’Esposito, F.; Gagliano, G.; Gagliano, C.; Maniaci, A.; Avitabile, A.; Giglio, R.; Reibaldi, M.; Cordeiro, M.F.; Zeppieri, M. Usher Syndrome: New Insights into Classification, Genotype-Phenotype Correlation, and Management. Genes 2025, 16, 332. [Google Scholar] [CrossRef]
  79. Kremer, H.; van Wijk, E.; Märker, T.; Wolfrum, U.; Roepman, R. Usher syndrome: Molecular links of pathogenesis, proteins and pathways. Hum. Mol. Genet. 2006, 15, R262–R270. [Google Scholar] [CrossRef]
  80. Smith, R.J.; Berlin, C.I.; Hejtmancik, J.F.; Keats, B.J.; Kimberling, W.J.; Lewis, R.A.; Möller, C.G.; Pelias, M.Z.; Tranebjaerg, L. Clinical diagnosis of the Usher syndromes. Usher Syndrome Consortium. Am. J. Med. Genet. 1994, 50, 32–38. [Google Scholar] [CrossRef]
  81. Delmaghani, S.; El-Amraoui, A. The genetic and phenotypic landscapes of Usher syndrome: From disease mechanisms to a new classification. Hum. Genet. 2022, 141, 709–735. [Google Scholar] [CrossRef]
  82. Mathur, P.; Yang, J. Usher syndrome: Hearing loss, retinal degeneration and associated abnormalities. Biochim. Biophys. Acta 2015, 1852, 406–420. [Google Scholar] [CrossRef]
  83. Weil, D.; Blanchard, S.; Kaplan, J.; Guilford, P.; Gibson, F.; Walsh, J.; Mburu, P.; Varela, A.; Levilliers, J.; Weston, M.D. Defective myosin VIIA gene responsible for Usher syndrome type 1B. Nature 1995, 374, 60–61. [Google Scholar] [CrossRef]
  84. Hasson, T.; Gillespie, P.G.; Garcia, J.A.; MacDonald, R.B.; Zhao, Y.; Yee, A.G.; Mooseker, M.S.; Corey, D.P. Unconventional myosins in inner-ear sensory epithelia. J. Cell Biol. 1997, 137, 1287–1307. [Google Scholar] [CrossRef]
  85. El-Amraoui, A.; Petit, C. Usher I syndrome: Unravelling the mechanisms that underlie the cohesion of the growing hair bundle in inner ear sensory cells. J. Cell Sci. 2005, 118, 4593–4603. [Google Scholar] [CrossRef]
  86. Xu, M.; Xie, Y.; Abouzeid, H.; Gordon, C.T.; Fiorentino, A.; Sun, Z.; Lehman, A.; Osman, I.S.; Dharmat, R.; Riveiro-Alvarez, R.; et al. Mutations in the Spliceosome Component CWC27 Cause Retinal Degeneration with or without Additional Developmental Anomalies. Am. J. Hum. Genet. 2017, 100, 592–604. [Google Scholar] [CrossRef] [PubMed]
  87. Brea-Fernández, A.J.; Cabanas, P.; Dacruz-Álvarez, D.; Caamaño, P.; Limeres, J.; Loidi, L. Expanding the clinical and molecular spectrum of the CWC27-related spliceosomopathy. J. Hum. Genet. 2019, 64, 1133–1136. [Google Scholar] [CrossRef] [PubMed]
  88. Busetto, V.; Barbosa, I.; Basquin, J.; Marquenet, É.; Hocq, R.; Hennion, M.; Paternina, J.A.; Namane, A.; Conti, E.; Bensaude, O.; et al. Structural and functional insights into CWC27/CWC22 heterodimer linking the exon junction complex to spliceosomes. Nucleic Acids Res. 2020, 48, 5670–5683. [Google Scholar] [CrossRef] [PubMed]
  89. Grainger, R.J.; Beggs, J.D. Prp8 protein: At the heart of the spliceosome. RNA 2005, 11, 533–557. [Google Scholar] [CrossRef]
  90. Mordes, D.; Luo, X.; Kar, A.; Kuo, D.; Xu, L.; Fushimi, K.; Yu, G.; Sternberg, P., Jr.; Wu, J.Y. Pre-mRNA splicing and retinitis pigmentosa. Mol. Vis. 2006, 12, 1259–1271. [Google Scholar]
  91. Mozaffari-Jovin, S.; Wandersleben, T.; Santos, K.F.; Will, C.L.; Lührmann, R.; Wahl, M.C. Inhibition of RNA helicase Brr2 by the C-terminal tail of the spliceosomal protein Prp8. Science 2013, 341, 80–84. [Google Scholar] [CrossRef]
  92. den Hollander, A.I.; ten Brink, J.B.; de Kok, Y.J.; van Soest, S.; van den Born, L.I.; van Driel, M.A.; van de Pol, D.J.; Payne, A.M.; Bhattacharya, S.S.; Kellner, U.; et al. Mutations in a human homologue of Drosophila crumbs cause retinitis pigmentosa (RP12). Nat. Genet. 1999, 23, 217–221. [Google Scholar] [CrossRef] [PubMed]
  93. van de Pavert, S.A.; Kantardzhieva, A.; Malysheva, A.; Meuleman, J.; Versteeg, I.; Levelt, C.; Klooster, J.; Geiger, S.; Seeliger, M.W.; Rashbass, P.; et al. Crumbs homologue 1 is required for maintenance of photoreceptor cell polarization and adhesion during light exposure. J. Cell Sci. 2004, 117, 4169–4177. [Google Scholar] [CrossRef]
  94. Bulgakova, N.A.; Knust, E. The Crumbs complex: From epithelial-cell polarity to retinal degeneration. J. Cell Sci. 2009, 122, 2587–2596. [Google Scholar] [CrossRef]
  95. Mehalow, A.K.; Kameya, S.; Smith, R.S.; Hawes, N.L.; Denegre, J.M.; Young, J.A.; Bechtold, L.; Haider, N.B.; Tepass, U.; Heckenlively, J.R.; et al. CRB1 is essential for external limiting membrane integrity and photoreceptor morphogenesis in the mammalian retina. Hum. Mol. Genet. 2003, 12, 2179–2189. [Google Scholar] [CrossRef]
  96. Zacchigna, S.; Oh, H.; Wilsch-Bräuninger, M.; Missol-Kolka, E.; Jászai, J.; Jansen, S.; Tanimoto, N.; Tonagel, F.; Seeliger, M.; Huttner, W.B.; et al. Loss of the cholesterol-binding protein prominin-1/CD133 causes disk dysmorphogenesis and photoreceptor degeneration. J. Neurosci. 2009, 29, 2297–2308. [Google Scholar] [CrossRef]
  97. Yang, Z.; Chen, Y.; Lillo, C.; Chien, J.; Yu, Z.; Michaelides, M.; Klein, M.; Howes, K.A.; Li, Y.; Kaminoh, Y.; et al. Mutant prominin 1 found in patients with macular degeneration disrupts photoreceptor disk morphogenesis in mice. J. Clin. Investig. 2009, 119, 1396. [Google Scholar] [CrossRef]
  98. Maw, M.A.; Corbeil, D.; Koch, J.; Hellwig, A.; Wilson-Wheeler, J.C.; Bridges, R.J.; Kumaramanickavel, G.; John, S.; Nancarrow, D.; Röper, K.; et al. A frameshift mutation in prominin (mouse)-like 1 causes human retinal degeneration. Hum. Mol. Genet. 2000, 9, 27–34. [Google Scholar] [CrossRef]
  99. Zhang, Q.; Zulfiqar, F.; Xiao, X.; Riazuddin, S.A.; Ahmad, Z.; Caruso, R.; MacDonald, I.; Sieving, P.; Riazuddin, S.; Hejtmancik, J.F. Severe retinitis pigmentosa mapped to 4p15 and associated with a novel mutation in the PROM1 gene. Hum. Genet. 2007, 122, 293–299. [Google Scholar] [CrossRef]
  100. Michaelides, M.; Gaillard, M.C.; Escher, P.; Tiab, L.; Bedell, M.; Borruat, F.X.; Barthelmes, D.; Carmona, R.; Zhang, K.; White, E.; et al. The PROM1 mutation p.R373C causes an autosomal dominant bull’s eye maculopathy associated with rod, rod-cone, and macular dystrophy. Investig. Ophthalmol. Vis. Sci. 2010, 51, 4771–4780. [Google Scholar] [CrossRef]
  101. Rice, D.S.; Calandria, J.M.; Gordon, W.C.; Jun, B.; Zhou, Y.; Gelfman, C.M.; Li, S.; Jin, M.; Knott, E.J.; Chang, B.; et al. Adiponectin receptor 1 conserves docosahexaenoic acid and promotes photoreceptor cell survival. Nat. Commun. 2015, 6, 6228. [Google Scholar] [CrossRef]
  102. Holland, W.L.; Miller, R.A.; Wang, Z.V.; Sun, K.; Barth, B.M.; Bui, H.H.; Davis, K.E.; Bikman, B.T.; Halberg, N.; Rutkowski, J.M.; et al. Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat. Med. 2011, 17, 55–63. [Google Scholar] [CrossRef]
  103. Sluch, V.M.; Banks, A.; Li, H.; Crowley, M.A.; Davis, V.; Xiang, C.; Yang, J.; Demirs, J.T.; Vrouvlianis, J.; Leehy, B.; et al. ADIPOR1 is essential for vision and its RPE expression is lost in the Mfrprd6 mouse. Sci. Rep. 2018, 8, 14339. [Google Scholar] [CrossRef]
  104. Vasiliauskaité-Brooks, I.; Sounier, R.; Rochaix, P.; Bellot, G.; Fortier, M.; Hoh, F.; De Colibus, L.; Bechara, C.; Saied, E.M.; Arenz, C.; et al. Structural insights into adiponectin receptors suggest ceramidase activity. Nature 2017, 544, 120–123. [Google Scholar] [CrossRef]
  105. Strettoi, E.; Gargini, C.; Novelli, E.; Sala, G.; Piano, I.; Gasco, P.; Ghidoni, R. Inhibition of ceramide biosynthesis preserves photoreceptor structure and function in a mouse model of retinitis pigmentosa. Proc. Natl. Acad. Sci. USA 2010, 107, 18706–18711. [Google Scholar] [CrossRef]
  106. Chen, H.; Tran, J.T.; Brush, R.S.; Saadi, A.; Rahman, A.K.; Yu, M.; Yasumura, D.; Matthes, M.T.; Ahern, K.; Yang, H.; et al. Ceramide signaling in retinal degeneration. Adv. Exp. Med. Biol. 2012, 723, 553–558. [Google Scholar]
  107. Zhang, J.; Wang, C.; Shen, Y.; Chen, N.; Wang, L.; Liang, L.; Guo, T.; Yin, X.; Ma, Z.; Zhang, B.; et al. A mutation in ADIPOR1 causes nonsyndromic autosomal dominant retinitis pigmentosa. Hum. Genet. 2016, 135, 1375–1387. [Google Scholar] [CrossRef]
  108. Xu, M.; Eblimit, A.; Wang, J.; Li, J.; Wang, F.; Zhao, L.; Wang, X.; Xiao, N.; Li, Y.; Wong, L.J.; et al. ADIPOR1 Is Mutated in Syndromic Retinitis Pigmentosa. Hum. Mutat. 2016, 37, 246–249. [Google Scholar] [CrossRef]
  109. Kaarniranta, K.; Paananen, J.; Nevalainen, T.; Sorri, I.; Seitsonen, S.; Immonen, I.; Salminen, A.; Pulkkinen, L.; Uusitupa, M. Adiponectin receptor 1 gene (ADIPOR1) variant is associated with advanced age-related macular degeneration in Finnish population. Neurosci. Lett. 2012, 513, 233–237. [Google Scholar] [CrossRef]
  110. Huang, C.H.; Yang, C.M.; Yang, C.H.; Hou, Y.C.; Chen, T.C. Leber’s Congenital Amaurosis: Current Concepts of Genotype-Phenotype Correlations. Genes 2021, 12, 1261. [Google Scholar] [CrossRef]
  111. Koenekoop, R.K. An overview of Leber congenital amaurosis: A model to understand human retinal development. Surv. Ophthalmol. 2004, 49, 379–398. [Google Scholar] [CrossRef]
  112. Sather, R., 3rd; Ihinger, J.; Simmons, M.; Lobo, G.P.; Montezuma, S.R. The Clinical Findings, Pathogenic Variants, and Gene Therapy Qualifications Found in a Leber Congenital Amaurosis Phenotypic Spectrum Patient Cohort. Int. J. Mol. Sci. 2024, 25, 1253. [Google Scholar] [CrossRef]
  113. Leroy, B.P.; Birch, D.G.; Duncan, J.L.; Lam, B.L.; Koenekoop, R.K.; Porto, F.B.O.; Russell, S.R.; Girach, A. Leber congenital amaurosis due to CEP290 mutations-severe vision impairment with a high unmet medical need: A review. Retina 2021, 41, 898–907. [Google Scholar] [CrossRef]
  114. den Hollander, A.I.; Koenekoop, R.K.; Yzer, S.; Lopez, I.; Arends, M.L.; Voesenek, K.E.J.; Zonneveld, M.N.; Strom, T.M.; Meitinger, T.; Brunner, H.G.; et al. Mutations in the CEP290 (NPHP6) Gene Are a Frequent Cause of Leber Congenital Amaurosis. Am. J. Hum. Genet. 2006, 79, 556–561. [Google Scholar] [CrossRef]
  115. Kumaran, N.; Moore, A.T.; Weleber, R.G.; Michaelides, M. Leber congenital amaurosis/early-onset severe retinal dystrophy: Clinical features, molecular genetics and therapeutic interventions. Br. J. Ophthalmol. 2017, 101, 1147–1154. [Google Scholar] [CrossRef]
  116. den Hollander, A.I.; Roepman, R.; Koenekoop, R.K.; Cremers, F.P. Leber congenital amaurosis: Genes, proteins and disease mechanisms. Prog. Retin. Eye Res. 2008, 27, 391–419. [Google Scholar] [CrossRef]
  117. Craige, B.; Tsao, C.C.; Diener, D.R.; Hou, Y.; Lechtreck, K.F.; Rosenbaum, J.L.; Witman, G.B. CEP290 tethers flagellar transition zone microtubules to the membrane and regulates flagellar protein content. J. Cell Biol. 2010, 190, 927–940. [Google Scholar] [CrossRef]
  118. Bernardos, R.L.; Barthel, L.K.; Meyers, J.R.; Raymond, P.A. Late-stage neuronal progenitors in the retina are radial Müller glia that function as retinal stem cells. J. Neurosci. 2007, 27, 7028–7040. [Google Scholar] [CrossRef]
  119. Conner, C.; Ackerman, K.M.; Lahne, M.; Hobgood, J.S.; Hyde, D.R. Repressing notch signaling and expressing TNFα are sufficient to mimic retinal regeneration by inducing Müller glial proliferation to generate committed progenitor cells. J. Neurosci. 2014, 34, 14403–14419. [Google Scholar] [CrossRef]
  120. Gorsuch, R.A.; Hyde, D.R. Regulation of Müller glial dependent neuronal regeneration in the damaged adult zebrafish retina. Exp. Eye Res. 2014, 123, 131–140. [Google Scholar] [CrossRef]
  121. Morimura, H.; Fishman, G.A.; Grover, S.A.; Fulton, A.B.; Berson, E.L.; Dryja, T.P. Mutations in the RPE65 gene in patients with autosomal recessive retinitis pigmentosa or leber congenital amaurosis. Proc. Natl. Acad. Sci. USA 1998, 95, 3088–3093. [Google Scholar] [CrossRef]
  122. Lotery, A.J.; Namperumalsamy, P.; Jacobson, S.G.; Weleber, R.G.; Fishman, G.A.; Musarella, M.A.; Hoyt, C.S.; Héon, E.; Levin, A.; Jan, J.; et al. Mutation analysis of 3 genes in patients with Leber congenital amaurosis. Arch. Ophthalmol. 2000, 118, 538–543. [Google Scholar] [CrossRef]
  123. Hanein, S.; Perrault, I.; Gerber, S.; Tanguy, G.; Barbet, F.; Ducroq, D.; Calvas, P.; Dollfus, H.; Hamel, C.; Lopponen, T.; et al. Leber congenital amaurosis: Comprehensive survey of the genetic heterogeneity, refinement of the clinical definition, and genotype-phenotype correlations as a strategy for molecular diagnosis. Hum. Mutat. 2004, 23, 306–317. [Google Scholar] [CrossRef]
  124. Thompson, D.A.; Gyürüs, P.; Fleischer, L.L.; Bingham, E.L.; McHenry, C.L.; Apfelstedt-Sylla, E.; Zrenner, E.; Lorenz, B.; Richards, J.E.; Jacobson, S.G.; et al. Genetics and phenotypes of RPE65 mutations in inherited retinal degeneration. Investig. Ophthalmol. Vis. Sci. 2000, 41, 4293–4299. [Google Scholar]
  125. Moiseyev, G.; Chen, Y.; Takahashi, Y.; Wu, B.X.; Ma, J.X. RPE65 is the isomerohydrolase in the retinoid visual cycle. Proc. Natl. Acad. Sci. USA 2005, 102, 12413–12418. [Google Scholar] [CrossRef]
  126. Gu, S.M.; Thompson, D.A.; Srikumari, C.R.; Lorenz, B.; Finckh, U.; Nicoletti, A.; Murthy, K.R.; Rathmann, M.; Kumaramanickavel, G.; Denton, M.J.; et al. Mutations in RPE65 cause autosomal recessive childhood-onset severe retinal dystrophy. Nat. Genet. 1997, 17, 194–197. [Google Scholar] [CrossRef]
  127. Redmond, T.M.; Yu, S.; Lee, E.; Bok, D.; Hamasaki, D.; Chen, N.; Goletz, P.; Ma, J.X.; Crouch, R.K.; Pfeifer, K. Rpe65 is necessary for production of 11-cis-vitamin A in the retinal visual cycle. Nat. Genet. 1998, 20, 344–351. [Google Scholar] [CrossRef]
  128. Maguire, A.M.; Bennett, J.; Aleman, E.M.; Leroy, B.P.; Aleman, T.S. Clinical Perspective: Treating RPE65-Associated Retinal Dystrophy. Mol. Ther. 2021, 29, 442–463. [Google Scholar] [CrossRef]
  129. Cideciyan, A.V.; Jacobson, S.G.; Beltran, W.A.; Sumaroka, A.; Swider, M.; Iwabe, S.; Roman, A.J.; Olivares, M.B.; Schwartz, S.B.; Komáromy, A.M.; et al. Human retinal gene therapy for Leber congenital amaurosis shows advancing retinal degeneration despite enduring visual improvement. Proc. Natl. Acad. Sci. USA 2013, 110, E517–E525. [Google Scholar] [CrossRef]
  130. Suh, S.; Choi, E.H.; Leinonen, H.; Foik, A.T.; Newby, G.A.; Yeh, W.-H.; Dong, Z.; Kiser, P.D.; Lyon, D.C.; Liu, D.R.; et al. Restoration of visual function in adult mice with an inherited retinal disease via adenine base editing. Nat. Biomed. Eng. 2021, 5, 169–178. [Google Scholar] [CrossRef]
  131. Freund, C.L.; Gregory-Evans, C.Y.; Furukawa, T.; Papaioannou, M.; Looser, J.; Ploder, L.; Bellingham, J.; Ng, D.; Herbrick, J.A.; Duncan, A.; et al. Cone-rod dystrophy due to mutations in a novel photoreceptor-specific homeobox gene (CRX) essential for maintenance of the photoreceptor. Cell 1997, 91, 543–553. [Google Scholar] [CrossRef]
  132. Tran, N.M.; Zhang, A.; Zhang, X.; Huecker, J.B.; Hennig, A.K.; Chen, S. Mechanistically distinct mouse models for CRX-associated retinopathy. PLoS Genet. 2014, 10, e1004111. [Google Scholar]
  133. Silva, E.; Yang, J.M.; Li, Y.; Dharmaraj, S.; Sundin, O.H.; Maumenee, I.H. A CRX null mutation is associated with both Leber congenital amaurosis and a normal ocular phenotype. Investig. Ophthalmol. Vis. Sci. 2000, 41, 2076–2079. [Google Scholar]
  134. Sohocki, M.M.; Sullivan, L.S.; Mintz-Hittner, H.A.; Birch, D.; Heckenlively, J.R.; Freund, C.L.; McInnes, R.R.; Daiger, S.P. A range of clinical phenotypes associated with mutations in CRX, a photoreceptor transcription-factor gene. Am. J. Hum. Genet. 1998, 63, 1307–1315. [Google Scholar] [CrossRef] [PubMed]
  135. Haider, N.B.; Jacobson, S.G.; Cideciyan, A.V.; Swiderski, R.; Streb, L.M.; Searby, C.; Beck, G.; Hockey, R.; Hanna, D.B.; Gorman, S.; et al. Mutation of a nuclear receptor gene, NR2E3, causes enhanced S cone syndrome, a disorder of retinal cell fate. Nat. Genet. 2000, 24, 127–131. [Google Scholar] [CrossRef] [PubMed]
  136. Jacobson, S.G.; Marmor, M.F.; Kemp, C.M.; Knighton, R.W. SWS (blue) cone hypersensitivity in a newly identified retinal degeneration. Investig. Ophthalmol. Vis. Sci. 1990, 31, 827–838. [Google Scholar]
  137. Marmor, M.F.; Jacobson, S.G.; Foerster, M.H.; Kellner, U.; Weleber, R.G. Diagnostic Clinical Findings of a New Syndrome with Night Blindness, Maculopathy, and Enhanced S Cone Sensitivity. Am. J. Ophthalmol. 1990, 110, 124–134. [Google Scholar]
  138. Lam, B.L.; Goldberg, J.L.; Hartley, K.L.; Stone, E.M.; Liu, M. Atypical Mild Enhanced S-Cone Syndrome with Novel Compound Heterozygosity of the NR2E3 Gene. Am. J. Ophthalmol. 2007, 144, 157–159. [Google Scholar] [CrossRef]
  139. de Carvalho, E.R.; Robson, A.G.; Arno, G.; Boon, C.J.F.; Webster, A.A.; Michaelides, M. Enhanced S-Cone Syndrome: Spectrum of Clinical, Imaging, Electrophysiologic, and Genetic Findings in a Retrospective Case Series of 56 Patients. Ophthalmol. Retina 2021, 5, 195–214. [Google Scholar] [CrossRef]
  140. Schorderet, D.F.; Escher, P. NR2E3 mutations in enhanced S-cone sensitivity syndrome (ESCS), Goldmann-Favre syndrome (GFS), clumped pigmentary retinal degeneration (CPRD), and retinitis pigmentosa (RP). Hum. Mutat. 2009, 30, 1475–1485. [Google Scholar] [CrossRef]
  141. Oh, E.C.T.; Cheng, H.; Hao, H.; Jia, L.; Khan, N.W.; Swaroop, A. Rod differentiation factor NRL activates the expression of nuclear receptor NR2E3 to suppress the development of cone photoreceptors. Brain Res. 2008, 1236, 16–29. [Google Scholar] [CrossRef]
  142. Cheng, H.; Khanna, H.; Oh, E.C.T.; Hicks, D.; Mitton, K.P.; Swaroop, A. Photoreceptor-specific nuclear receptor NR2E3 functions as a transcriptional activator in rod photoreceptors. Hum. Mol. Genet. 2004, 13, 1563–1575. [Google Scholar] [CrossRef]
  143. Cheng, H.; Khan, N.W.; Roger, J.E.; Swaroop, A. Excess cones in the retinal degeneration rd7 mouse, caused by the loss of function of orphan nuclear receptor Nr2e3, originate from early-born photoreceptor precursors. Hum. Mol. Genet. 2011, 20, 4102–4115. [Google Scholar] [CrossRef] [PubMed]
  144. Zaydon, Y.A.; Tsang, S.H. The ABCs of Stargardt disease: The latest advances in precision medicine. Cell Biosci. 2024, 14, 98. [Google Scholar] [CrossRef] [PubMed]
  145. Tanna, P.; Strauss, R.W.; Fujinami, K.; Michaelides, M. Stargardt disease: Clinical features, molecular genetics, animal models and therapeutic options. Br. J. Ophthalmol. 2017, 101, 25–30. [Google Scholar] [CrossRef] [PubMed]
  146. Sparrow, J.R.; Gregory-Roberts, E.; Yamamoto, K.; Blonska, A.; Ghosh, S.K.; Ueda, K.; Zhou, J. The bisretinoids of retinal pigment epithelium. Prog. Retin. Eye Res. 2012, 31, 121–135. [Google Scholar] [CrossRef]
  147. Mata, N.L.; Tzekov, R.T.; Liu, X.; Weng, J.; Birch, D.G.; Travis, G.H. Delayed Dark-Adaptation and Lipofuscin Accumulation in abcr+/− Mice: Implications for Involvement of ABCR in Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1685–1690. [Google Scholar]
  148. Haji Abdollahi, S.; Hirose, T. Stargardt-Fundus Flavimaculatus: Recent Advancements and Treatment. Semin. Ophthalmol. 2013, 28, 372–376. [Google Scholar] [CrossRef]
  149. Quazi, F.; Lenevich, S.; Molday, R.S. ABCA4 is an N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine importer. Nat. Commun. 2012, 3, 925. [Google Scholar] [CrossRef]
  150. Molday, R.S.; Garces, F.A.; Scortecci, J.F.; Molday, L.L. Structure and function of ABCA4 and its role in the visual cycle and Stargardt macular degeneration. Prog. Retin. Eye Res. 2022, 89, 101036. [Google Scholar] [CrossRef]
  151. Zobor, D.; Zobor, G.; Kohl, S. Achromatopsia: On the Doorstep of a Possible Therapy. Ophthalmic Res. 2015, 54, 103–108. [Google Scholar] [CrossRef]
  152. Thiadens, A.A.H.J.; den Hollander, A.I.; Roosing, S.; Nabuurs, S.B.; Zekveld-Vroon, R.C.; Collin, R.W.J.; De Baere, E.; Koenekoop, R.K.; van Schooneveld, M.J.; Strom, T.M.; et al. Homozygosity Mapping Reveals PDE6C Mutations in Patients with Early-Onset Cone Photoreceptor Disorders. Am. J. Hum. Genet. 2009, 85, 240–247. [Google Scholar] [CrossRef] [PubMed]
  153. Brotherton, C.; Megaw, R. Molecular Mechanisms Governing Sight Loss in Inherited Cone Disorders. Genes 2024, 15, 727. [Google Scholar] [CrossRef] [PubMed]
  154. Weisschuh, N.; Stingl, K.; Audo, I.; Biskup, S.; Bocquet, B.; Branham, K.; Burstedt, M.S.; De Baere, E.; De Vries, M.J.; Golovleva, I.; et al. Mutations in the gene PDE6C encoding the catalytic subunit of the cone photoreceptor phosphodiesterase in patients with achromatopsia. Hum. Mutat. 2018, 39, 1366–1371. [Google Scholar] [CrossRef] [PubMed]
  155. Michalakis, S.; Gerhardt, M.; Rudolph, G.; Priglinger, S.; Priglinger, C. Achromatopsia: Genetics and Gene Therapy. Mol. Diagn. Ther. 2022, 26, 51–59. [Google Scholar] [CrossRef]
  156. Baxter, M.F.; Borchert, G.A. Gene Therapy for Achromatopsia. Int. J. Mol. Sci. 2024, 25, 9739. [Google Scholar] [CrossRef]
  157. Georgiou, M.; Robson, A.G.; Singh, N.; Pontikos, N.; Kane, T.; Hirji, N.; Ripamonti, C.; Rotsos, T.; Dubra, A.; Kalitzeos, A.; et al. Deep Phenotyping of PDE6C-Associated Achromatopsia. Investig. Ophthalmol. Vis. Sci. 2019, 60, 5112–5123. [Google Scholar] [CrossRef]
  158. Grau, T.; Artemyev, N.O.; Rosenberg, T.; Dollfus, H.; Haugen, O.H.; Cumhur Sener, E.; Jurklies, B.; Andreasson, S.; Kernstock, C.; Larsen, M.; et al. Decreased catalytic activity and altered activation properties of PDE6C mutants associated with autosomal recessive achromatopsia. Hum. Mol. Genet. 2011, 20, 719–730. [Google Scholar] [CrossRef]
  159. Ding, J.; Adiconis, X.; Simmons, S.K.; Kowalczyk, M.S.; Hession, C.C.; Marjanovic, N.D.; Hughes, T.K.; Wadsworth, M.H.; Burks, T.; Nguyen, L.T.; et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat. Biotechnol. 2020, 38, 737–746. [Google Scholar] [CrossRef]
  160. Lähnemann, D.; Köster, J.; Szczurek, E.; McCarthy, D.J.; Hicks, S.C.; Robinson, M.D.; Vallejos, C.A.; Campbell, K.R.; Beerenwinkel, N.; Mahfouz, A.; et al. Eleven grand challenges in single-cell data science. Genome Biol. 2020, 21, 31. [Google Scholar] [CrossRef]
  161. 10x Genomics. Are There Considerations for Small Cells or Cells with Low RNA? Available online: https://kb.10xgenomics.com/hc/en-us/articles/360037656471-Are-there-considerations-for-small-cells-or-cells-with-low-RNA (accessed on 29 August 2025).
  162. Sant, P.; Rippe, K.; Mallm, J.-P. Approaches for single-cell RNA sequencing across tissues and cell types. Transcription 2023, 14, 127–145. [Google Scholar] [CrossRef]
  163. Bues, J.; Biočanin, M.; Pezoldt, J.; Dainese, R.; Chrisnandy, A.; Rezakhani, S.; Saelens, W.; Gardeux, V.; Gupta, R.; Sarkis, R.; et al. Deterministic scRNA-seq captures variation in intestinal crypt and organoid composition. Nat. Methods 2022, 19, 323–330. [Google Scholar] [CrossRef]
  164. De Rop, F.V.; Ismail, J.N.; Bravo González-Blas, C.; Hulselmans, G.J.; Flerin, C.C.; Janssens, J.; Theunis, K.; Christiaens, V.M.; Wouters, J.; Marcassa, G.; et al. Hydrop enables droplet-based single-cell ATAC-seq and single-cell RNA-seq using dissolvable hydrogel beads. eLife 2022, 11, e73971. [Google Scholar] [CrossRef] [PubMed]
  165. Crow, M.; Gillis, J. Single cell RNA-sequencing: Replicability of cell types. Curr. Opin. Neurobiol. 2019, 56, 69–77. [Google Scholar] [CrossRef] [PubMed]
  166. Chen, J.; Cheung, F.; Shi, R.; Zhou, H.; Lu, W.; Candia, J.; Kotliarov, Y.; Stagliano, K.R.; Tsang, J.S.; CHI Consortium. PBMC fixation and processing for Chromium single-cell RNA sequencing. J. Transl. Med. 2018, 16, 198. [Google Scholar] [CrossRef] [PubMed]
  167. Fadl, B.R.; Brodie, S.A.; Malasky, M.; Boland, J.F.; Kelly, M.C.; Kelley, M.W.; Boger, E.; Fariss, R.; Swaroop, A.; Campello, L. An optimized protocol for retina single-cell RNA sequencing. Mol. Vis. 2020, 26, 705–717. [Google Scholar]
  168. Denisenko, E.; Guo, B.B.; Jones, M.; Hou, R.; de Kock, L.; Lassmann, T.; Poppe, D.; Clément, O.; Simmons, R.K.; Lister, R.; et al. Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 2020, 21, 130. [Google Scholar] [CrossRef]
  169. Zimmerman, K.D.; Espeland, M.A.; Langefeld, C.D. A practical solution to pseudoreplication bias in single-cell studies. Nat. Commun. 2021, 12, 738. [Google Scholar] [CrossRef]
  170. Squair, J.W.; Gautier, M.; Kathe, C.; Anderson, M.A.; James, N.D.; Hutson, T.H.; Hudelle, R.; Qaiser, T.; Matson, K.J.E.; Barraud, Q.; et al. Confronting false discoveries in single-cell differential expression. Nat. Commun. 2021, 12, 5692. [Google Scholar] [CrossRef]
  171. Junttila, S.; Smolander, J.; Elo, L.L. Benchmarking methods for detecting differential states between conditions from multi-subject single-cell RNA-seq data. Brief. Bioinform. 2022, 23, bbac286. [Google Scholar] [CrossRef]
  172. Satija, R.; Farrell, J.A.; Gennert, D.; Schier, A.F.; Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 2015, 33, 495–502. [Google Scholar] [CrossRef]
  173. Hao, Y.; Stuart, T.; Kowalski, M.H.; Choudhary, S.; Hoffman, P.; Hartman, A.; Srivastava, A.; Molla, G.; Madad, S.; Fernandez-Granda, C.; et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat. Biotechnol. 2024, 42, 293–304. [Google Scholar] [CrossRef] [PubMed]
  174. Gentleman, R.C.; Carey, V.J.; Bates, D.M.; Bolstad, B.; Dettling, M.; Dudoit, S.; Ellis, B.; Gautier, L.; Ge, Y.; Gentry, J.; et al. Bioconductor: Open software development for computational biology and bioinformatics. Genome Biol. 2004, 5, R80. [Google Scholar] [CrossRef] [PubMed]
  175. Amezquita, R.A.; Lun, A.T.L.; Becht, E.; Carey, V.J.; Carpp, L.N.; Geistlinger, L.; Marini, F.; Rue-Albrecht, K.; Risso, D.; Soneson, C.; et al. Orchestrating single-cell analysis with Bioconductor. Nat. Methods 2020, 17, 137–145. [Google Scholar] [CrossRef] [PubMed]
  176. Kolodziejczyk, A.A.; Kim, J.K.; Svensson, V.; Marioni, J.C.; Teichmann, S.A. The Technology and Biology of Single-Cell RNA Sequencing. Mol. Cell 2015, 58, 610–620. [Google Scholar] [CrossRef]
  177. Haque, A.; Engel, J.; Teichmann, S.A.; Lönnberg, T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 2017, 9, 75. [Google Scholar] [CrossRef]
  178. Lyu, Y.; Zauhar, R.; Dana, N.; Strang, C.E.; Hu, J.; Wang, K.; Liu, S.; Pan, N.; Gamlin, P.; Kimble, J.A.; et al. Implication of specific retinal cell-type involvement and gene expression changes in AMD progression using integrative analysis of single-cell and bulk RNA-seq profiling. Sci. Rep. 2021, 11, 15612. [Google Scholar] [CrossRef]
  179. Rao, A.; Barkley, D.; França, G.S.; Yanai, I. Exploring tissue architecture using spatial transcriptomics. Nature 2021, 596, 211–220. [Google Scholar] [CrossRef]
  180. Choi, J.; Li, J.; Ferdous, S.; Liang, Q.; Moffitt, J.R.; Chen, R. Spatial organization of the mouse retina at single cell resolution by MERFISH. Nat. Commun. 2023, 14, 4929. [Google Scholar] [CrossRef]
  181. Schumann, U.; Liu, L.; Aggio-Bruce, R.; Cioanca, A.V.; Shariev, A.; Madigan, M.C.; Valter, K.; Wen, J.; Natoli, R. Spatial transcriptomics reveals regionally altered gene expression that drives retinal degeneration. Commun. Biol. 2025, 8, 629. [Google Scholar] [CrossRef]
  182. Longo, S.K.; Guo, M.G.; Ji, A.L.; Khavari, P.A. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat. Rev. Genet. 2021, 22, 627–644. [Google Scholar] [CrossRef]
  183. Yan, C.; Zhu, Y.; Chen, M.; Yang, K.; Cui, F.; Zou, Q.; Zhang, Z. Integration tools for scRNA-seq data and spatial transcriptomics sequencing data. Brief. Funct. Genom. 2024, 23, 295–302. [Google Scholar] [CrossRef]
  184. Zhou, Y.; Sheng, Y.; Pan, M.; Tu, J.; Zhao, X.; Ge, Q.; Lu, Z. Spatial Transcriptomic Analysis Reveals Regional Transcript Changes in Early and Late Stages of rd1 Model Mice with Retinitis Pigmentosa. Int. J. Mol. Sci. 2023, 24, 14869. [Google Scholar] [CrossRef]
  185. Zhang, J.; Wang, J.; Zhou, Q.; Chen, Z.; Zhuang, J.; Zhao, X.; Gan, Z.; Wang, Y.; Wang, C.; Molday, R.S.; et al. Spatiotemporally resolved transcriptomics reveals the cellular dynamics of human retinal development. Nat. Commun. 2025, 16, 2307. [Google Scholar] [CrossRef]
  186. Vickovic, S.; Eraslan, G.; Salmén, F.; Klughammer, J.; Stenbeck, L.; Schapiro, D.; Äijö, T.; Bonneau, R.; Bergenstråhle, L.; Navarro, J.F.; et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods 2019, 16, 987–990. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Distribution of single-cell transcriptomics studies in IRDs: (A) Overall distribution of studies across five major IRD categories: retinitis pigmentosa (RP; n = 15; 65%), Leber congenital amaurosis (LCA; n = 3; 13%), enhanced S-cone syndrome (ESCS; n = 2; 9%), Stargardt disease (STGD; n = 2; 9%), and achromatopsia (ACHM; n = 1; 4%). (B) Breakdown of RP studies by specific genetic mutations: RHO (n = 4; 27%), PDE6B (n = 3; 20%), RPGR (n = 2; 13%), and other RP-related mutations (n = 6; 40%). Data from literature search of 23 studies. ACHM: Achromatopsia; ESCS: Enhanced S-cone syndrome; IRD: Inherited retinal dystrophy; LCA: Leber congenital amaurosis; RP: Retinitis pigmentosa; STGD: Stargardt disease.
Figure 1. Distribution of single-cell transcriptomics studies in IRDs: (A) Overall distribution of studies across five major IRD categories: retinitis pigmentosa (RP; n = 15; 65%), Leber congenital amaurosis (LCA; n = 3; 13%), enhanced S-cone syndrome (ESCS; n = 2; 9%), Stargardt disease (STGD; n = 2; 9%), and achromatopsia (ACHM; n = 1; 4%). (B) Breakdown of RP studies by specific genetic mutations: RHO (n = 4; 27%), PDE6B (n = 3; 20%), RPGR (n = 2; 13%), and other RP-related mutations (n = 6; 40%). Data from literature search of 23 studies. ACHM: Achromatopsia; ESCS: Enhanced S-cone syndrome; IRD: Inherited retinal dystrophy; LCA: Leber congenital amaurosis; RP: Retinitis pigmentosa; STGD: Stargardt disease.
Genes 16 01088 g001
Figure 2. Schematic of the retina showing the locations of IRD-associated genes and key pathway dysregulation identified by scRNA-seq studies. Gene positions indicate primary expression sites, with disease mechanisms and findings from single-cell approaches including photoreceptor-specific vulnerabilities, glial responses, and therapeutic targets. Note that MYO7A localisation reflects scRNA-seq expression data, which differs from previous studies localising it primarily to the RPE. AC: Amacrine cell; BC: Bipolar cell; GC: Ganglion cell; HC: Horizontal cell; MG: Müller glia; RPE: Retinal pigment epithelium.
Figure 2. Schematic of the retina showing the locations of IRD-associated genes and key pathway dysregulation identified by scRNA-seq studies. Gene positions indicate primary expression sites, with disease mechanisms and findings from single-cell approaches including photoreceptor-specific vulnerabilities, glial responses, and therapeutic targets. Note that MYO7A localisation reflects scRNA-seq expression data, which differs from previous studies localising it primarily to the RPE. AC: Amacrine cell; BC: Bipolar cell; GC: Ganglion cell; HC: Horizontal cell; MG: Müller glia; RPE: Retinal pigment epithelium.
Genes 16 01088 g002
Table 1. Single-cell transcriptomics studies examining pathological changes in disease models of inherited retinal dystrophies.
Table 1. Single-cell transcriptomics studies examining pathological changes in disease models of inherited retinal dystrophies.
IRDMutationStudyModelBiological ReplicatesDisease StageInterventionPlatformMain Dysregulated Pathways or Markers in Disease (or Treatment) Conditions
RPRHOTomita et al. [16]Mouse (RhoP23H)4 MUT and 4 WTActive degeneration (PW7)NoinDrop↓ in PRs: phototransduction, cilium, development, ATP processes, glycolysis; ↑ in Müller cells: PR maintenance, mitochondrial localisation and transport
RPRHOFu et al. [17]Mouse (RhoP23H)3 FGF21-treated and 3 vehicle-treated CTRLActive degeneration (PW10)FGF21 treatmentinDropFGF21-treated Müller cells and astrocytes: axon development↑, synapse formation↑, SRF signalling↑
RPRHOSanthanam et al. [18]Zebrafish1 MUT and 1 WT for V2 and V3 each; 3 fish pooled eachActive degeneration (age 6–10 months)No10xOxidative stress, metabolic reprogramming, misfolded proteins, circadian rhythm disruption, synaptic remodelling, regenerative signalling
RPRHOLin et al. [19]Human iPSC-derived rOrgs1 patient and 1 healthy CTRL at D120 and D270 each; 5–6 rOrgs eachImmature (D120) and mature (D270) stageNo10xVisual perception, PR development, phototransduction, rod maturation, immune and inflammatory signalling
RPPDE6BChen et al. [20] 1Mouse (rd1)MUT and WT at P11, P13, P17; 3 animals per time pointEarly (P11), peak rod degeneration (P13), late rod degeneration (P17)No10xVisual perception, phototransduction, apoptosis, Ca2+ signalling, MAPK pathway, metabolic pathways
RPPDE6BDong et al. [21] 1Mouse (rd1)MUT and WT at P11, P13, P17; 3 animals per time pointEarly (P11), peak rod degeneration (P13), late rod degeneration (P17)No10xEgr1↑ in PRs
RPPDE6BDong et al. [22] 1Mouse (rd1)MUT and WT at P11, P13, P17; 3 animals per time pointEarly (P11), peak rod degeneration (P13), late rod degeneration (P17)No10xHdac1↑ in rods, Parp1↓ in cones
RPPDE6BYan et al. [231Mouse (rd1)MUT and WT at P11, P13, P17; 3 animals per time pointFocus on peak rod degeneration (P13)No10xCa2+ signalling in PRs
RPPDE6BLiao et al. [24]Mouse (rd1)2 MUT and 2 WTPeak rod degeneration (P15)No10xJNK signalling↑, Jun transcription factor↑
RPPDE6BKarademir et al. [25] 2Mouse (rd10)2 MUT and 2 WTPeak rod degeneration (P21)No10xEarly phase: Ca2+ signalling, metabolic disruption, phototransduction, Egr1 activation; late phase: mitochondrial respiratory dysfunction, synaptic remodelling, structural changes, Cebpd activation
RPPDE6BSigurdsson and Grimm [26] 2Mouse (rd10)2 MUT and 2 WTPeak rod degeneration (P21)No10xMüller cell response: gliosis and metabolic markers, immune response, MHC components
RP, CRD, CD, MDRPGRNewton et al. [27]Mouse (RpgrORFd5, RpgrEx3d8)1 MUT and 1 WT for RpgrORFd5 and RpgrEx3d8 eachActive photoreceptor degeneration (18 months)No10x↓ in PRs: phototransduction; ↑ in PRs: PI3K/AKT pathway, autophagy, necroptosis, mitochondrial function, TNF- α /NF- κ B signalling, lysosome biogenesis
RPRPGRLi et al. [28]Human iPSC-derived rOrgs1 patient and 1 healthy CTRL at D40, D90, D150, D200 each; 2–3 rOrgs eachMultiple developmental time points (D40, D90, D150, D200)No10x↑ of PR markers
RPMYO7ALeong et al. [29]Human iPSC-derived rOrgs3 patients and 3 healthy CTRLsEarly/pre-degenerative stage (D245/35 weeks)No10x↑ in rods: cellular and oxidative stress, hydrogen peroxide metabolism; no pathology detected for cones; ↑ in Müller cells: apoptosis
RPCWC27Bertrand et al. [30]Mouse (Cwc27K338fs)1 MUT and 1 WTEarly degenerative stage (4 months)No10x↓ in rods: mitochondrial genes; ↑ in Müller cells: inflammation-related genes
RPPRPF8Atkinson et al. [31]Human iPSC-derived rOrgs4 patients and 4 isogenic CTRLsAdvanced degeneration (D210)No10xMALAT1↑ (degenerating rods), ↓ of cone (ARR3) and rod (NRL) markers, ↓ of phototransduction and mitochondrial genes, ciliary dysfunction
RP, LCACRB1Boon et al. [32] 3Human iPSC-derived rOrgs1 patient; 1st scRNA-seq: 1 patient and 1 healthy CTRL; 2nd scRNA-seq: 1 gene therapy-treated (gene 1) and 1 gene therapy-treated (gene 2) and 1 CTRL-treated; 5–6 rOrgs eachActive photoreceptor degeneration (D230)AAV gene therapy10xUntreated disease: endosomal system dysfunction in rods and Müller cells; phototransduction cascade activation in rods; cell adhesion, protein binding, cell death, iron ion transport in Müller cells; AAV-treated: partial restoration of endosomal system pathways toward control levels
RP, MDPROM1Shigesada et al. [33]Mouse (Prom1−/−)1 light-exposed MUT and 1 dark-reared MUTEarly/pre-symptomatic stage (P11)No10xIgf1↓ in rods and astrocytes, Edn2↑ in rods, glial activation in Müller glia and astrocytes, visual function dysfunction in PRs
RPADIPOR1Lewandowski et al. [34]Mouse (AdiporR1−/−)2 MUT and 2 WT at P19, P25, P30 eachEarly/pre-onset (P18), active/post-onset (P25), advanced (P30) degenerationNo10xPre-onset: translation↑, oxidative phosphorylation↑, mitochondrion organisation↑, RNA splicing↓, protein localisation↓; active and advanced phase: visual perception↓, translation↑; neurodegenerative pathways
LCACEP290Fogerty et al. [35]Zebrafish5 MUT and 5 HET CTRLsActive degeneration (6 months)NoSplitBio↑ in Müller cells: notch3, stat2, yap1, rest-associated genes
LCARPE65Choi et al. [36]Mouse (rd12)4 treated MUT and 4 untreated MUT and 4 WTAdvanced degeneration (2 months)Base editing therapy10xBase editing-treated cones: phototransduction restoration (rescue of phototransduction genes), opsin recovery (Opn1sw), neuroprotection (Mt1↑), cell survival (cell death/stress genes↓)
LCA, CRD, MDCRXKruczek et al. [37]Human iPSC-derived rOrgs2 patients (treated and untreated) and 2 healthy CTRLs; 3–4 rOrgs eachImpaired photoreceptor maturation (D200)AAV gene therapy10xAAV-CRX-treated PRs: partial rescue of RHO, OPN1MW, CABP4
ESCSNR2E3Mullin et al. [38]Human iPSC-derived rOrgs1 patient and 1 isogenic CTRL and 1 healthy CTRL at D40, D80, D120, D160, D260 each; 10 rOrgs eachMultiple time points (D40, D80, D120, D160, D260)AAV gene therapy10xUntreated PRs: phototransduction pathway disruption; “divergent rods”: RHO↓, cone genes↑ (ARR3, PDE6H, GNAT2), rod genes↑ (GNAT1)
ESCSNR2E3Aísa-Marín et al. [39]Mouse (Nr2e3  Δ 27, Nr2e3  Δ E8)P40: 2 Δ E8 MUT and 2 WT; P80: 2 Δ 27 MUT and 1 WTEarly-onset, but stable retinal dysfunction ( Δ 27 at P80); pre-symptomatic stage of progressive late-onset retinal degeneration ( Δ E8 at P40)No10xFormation of “hybrid photoreceptors”, phototransduction↑, autophagy and necrosis↑, stress response↑, homeostasis↓, mitochondria and synaptic function↓
STGDABCA4Watson et al. [40]Human iPSC-derived rOrgs3 patients and 2 healthy CTRLs; at least 25 rOrgs eachImpaired photoreceptor maturation (D200)No10xStress response pathways (mTOR signalling, mitochondrial dysfunction, oxidative phosphorylation), phototransduction disruption, cell cycle dysregulation, and impaired maturation
STGDABCA4Zhao et al. [41]Human iPSC-derived rOrgs1 patient and 1 healthy CTRLMultiple time points (D40, D90, D150, D200, D260)No10xNo specific pathways or markers reported
ACHMPDE6CMiller et al. [42]Mouse (Pde6ccpfl1)1 treated MUT and 1 untreated MUTPeak cone degeneration (P24)H3K27 demethylase inhibitor treatmentSMART-seq2 / MARS-seqGSK-J4-treated cones: oxidative phosphorylation↓, mitochondrial dysfunction↓, endoplasmic reticulum stress↓
1 Studies used same scRNA-seq dataset. GSE ID: GSE212183. 2 Studies used same scRNA-seq dataset. GSE ID: GSE183206. 3 Authors performed two separate scRNA-seq experiments on P128 patient samples as well as analyses on organoids from P116 and P117 patients to support their findings from P128 samples (not included in this table). Abbreviations: ↓: Downregulated; ↑: Upregulated; AAV: Adeno-associated virus; CD: Cone dystrophy; CRD: Cone-rod dystrophy; CTRL: Control; D: Day; EGR1: Early growth response 1; ESCS: Enhanced S-cone syndrome; FGF21: Fibroblast growth factor 21; GSE ID: Gene expression omnibus series identifier; H3K27: Histone H3 lysine 27; HDAC: Histone deacetylase; HET: Heterozygous; iPSCs: Induced pluripotent stem cells; LCA: Leber congenital amaurosis; MAPK: Mitogen-activated protein kinase; MD: Macular dystrophy; MHC: Major histocompatibility complex; MUT: Mutant; P: Postnatal day; PARP: Poly (ADP-ribose) polymerase; PR: Photoreceptor(s); PW: Postnatal week; rOrgs: Retinal organoids; RP: Retinitis pigmentosa; scRNA-seq: Single-cell RNA sequencing; SRF: Serum response factor; V: Version; WT: Wild-type.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nguyen, L.; Vallejos, C.A.; Mill, P.; Megaw, R. Single-Cell Transcriptomics in Inherited Retinal Dystrophies: Current Findings and Emerging Perspectives. Genes 2025, 16, 1088. https://doi.org/10.3390/genes16091088

AMA Style

Nguyen L, Vallejos CA, Mill P, Megaw R. Single-Cell Transcriptomics in Inherited Retinal Dystrophies: Current Findings and Emerging Perspectives. Genes. 2025; 16(9):1088. https://doi.org/10.3390/genes16091088

Chicago/Turabian Style

Nguyen, Linda, Catalina A. Vallejos, Pleasantine Mill, and Roly Megaw. 2025. "Single-Cell Transcriptomics in Inherited Retinal Dystrophies: Current Findings and Emerging Perspectives" Genes 16, no. 9: 1088. https://doi.org/10.3390/genes16091088

APA Style

Nguyen, L., Vallejos, C. A., Mill, P., & Megaw, R. (2025). Single-Cell Transcriptomics in Inherited Retinal Dystrophies: Current Findings and Emerging Perspectives. Genes, 16(9), 1088. https://doi.org/10.3390/genes16091088

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop