Next Article in Journal
Comparative Analysis of Deep Learning Models for Predicting Causative Regulatory Variants
Previous Article in Journal
Transcriptional Activation Mechanisms and Target Genes of the Oncogene Product Tax of Human T-Cell Leukemia Virus Type 1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Therapy of Fuchs Endothelial Corneal Dystrophy: Where Are We? A Review

by
Spela Stunf Pukl
1,2
1
Medical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
2
Eye Hospital, University Clinical Center Ljubljana, 1000 Ljubljana, Slovenia
Genes 2025, 16(10), 1222; https://doi.org/10.3390/genes16101222
Submission received: 20 August 2025 / Revised: 2 October 2025 / Accepted: 14 October 2025 / Published: 15 October 2025
(This article belongs to the Special Issue Genetic Diagnosis and Therapeutics of Eye Diseases)

Abstract

Objectives: The incidence of Fuchs endothelial corneal dystrophy (FECD) is growing, and with it, the unmet need for a corneal transplant. Among alternative treatment modalities, only genetic therapy represents a causal therapy. Methods: Following the SNARA protocol, the PubMed and ClinicalTrials databases were searched using the keywords Fuchs endothelial corneal dystrophy, FECD, genetic therapy, and CRISPR-Cas9. Results: FECD is polyfactorial disease and mutations or polymorphisms in at least 15 different genes were connected to the disease. For the early-onset form of the disease, exclusive connection to mutations in COL8A2 was confirmed, while for the late-onset form, the most characteristic mutation is the expansion of the CTG18.1 triplet in the TCF4 gene, making these two possible targets. While the CRISPR-Cas9 approach represents the mainstay of genetic therapy development recently, the application of this method to FECD contains several obstacles, studied in preclinical settings. DT-168 and the Ad-Cas9-Col8a2gRNA molecules were developed for FECD treatment and preclinically tested, and phase I and II clinical studies for DT-168 are also already being performed. Conclusions: The review of the literature proved that genetic therapy for FECD is at the level of preclinical research and that there are several specific challenges connected to the target genetic mutation as well as the delivery of possible treatment and duration of the effect. Further studies in the field might bring solutions in the future for alternative treatments for this common corneal disease.

1. Introduction

The Fuchs endothelial corneal dystrophy (FECD, MIM 136800) was introduced by Ernst Fuchs in 1910. It is a frequent progressive late-onset genetic and multifactorial disease, affecting primary corneal endothelium. Patients with FECD experience debilitating vision loss due to progressive corneal opacification resulting from endothelial cell loss. Endothelial changes commence in the central cornea, and progress towards the periphery (Figure 1). Dysfunctional endothelium eventually causes corneal edema [1].
FECD represents the number one cause for corneal transplantation in the developed world; endothelial transplantation represents the golden standard of care for patients with FECD. With this procedure, healthy donor endothelium is transplanted selectively in order to regain the endothelial pump function and cornel transparency. An aging population causes growing numbers of FECD, and cornea clinics are challenged due to a constant shortage of corneal tissue and consequent long waiting times for transplantation, which results in the progression of the disease to persistent corneal edema and secondary stromal fibrosis, which is the stage where endothelial transplantation is not sufficient for visual recovery.
This is the major reason which drives several different initiatives for alternatives to transplantation. Clinically, pharmacological endothelial corneal regenerative therapy with Rho kinase inhibitors is being studied in this respect [1] and there were a series of patients already treated with cell therapy [2,3].
Mirroring the other ophthalmologic genetic treatments already approved for clinical use [4], FECD is being studied for genetic treatment. This review article aims to present the current status and future pipeline of genetic therapy for FECD.

2. Methods

To review the current knowledge in the field of genetic therapy for FECD, the Scale for the Assessment of Narrative Review Articles—SNARA protocol was used.
The PubMed and ClinicalTrials databases were searched for preclinical as well as clinical reports.
The keywords used for searching the publications were as follows: Fuchs endothelial corneal dystrophy, FECD, genetic therapy, CRISPR-Cas9 in the period 2000 to 2025.

3. Fuchs Endothelial Corneal Dystrophy

3.1. Clinical Staging and Symptomatic

The main clinical findings in early FECD are guttae on the endothelial side of the cornea surface, which are composed of excessive extracellular matrix. The Descemet membrane is changed and thickened, and there is secondary endothelial cell layer disruption [5]. The main symptom of early stage FECD is blurring of vision; some patients also report glare, and about one third are asymptomatic [6,7].
Despite the absence of epithelial or stromal edema, the light scattering by guttae can cause debilitating symptoms, influence the quality of life, and warrants treatment at an early stage of the disease [6,8].
Later stages of the disease are characterized by corneal edema due to severe endothelial function loss [5].
Firstly, the symptoms are worse in the morning, and later the vision is continuously blurred [6].
The typical presentation of FECD is the late-onset subtype of the disease, which clinically presents between the fifth and seventh decade of life. The early-onset subtype, which typically occurs in the first decade of life, represents less than 5% of cases [7].
FECD is overall more frequent and progressive in females; however, if only the late-onset form is considered, the male population is affected three times more often [7,9].

3.2. Etiology of FECD

The etiology of FECD is complex and heterogenic with genetic, lifestyle, and environmental factors influencing its occurrence and course [10,11,12].

4. Genetics of FECD

4.1. Inheritance of FECD

The inheritance is autosomal dominant with incomplete penetrance and variable expressivity; thus, there is a vast clinical spectrum of age of onset, symptoms severity, and progression even among individuals within a single family [11].
Corneal clinics are even more challenged due to the fact that about half of the cases occur sporadically, and only half are familial [10].

4.2. FECD Genes

There are more than 15 genes with mutations and/or single-nucleotide polymorphisms associated with FECD [10] (Table 1).
The early-onset subtype differs from the late-onset genetically.

4.2.1. Early-Onset Subtype

For the early-onset subtype, the mutations in the COL8A2 gene are characteristic. Mutations causing early-onset FECD have been exclusively linked to the α2 chain of collagen 8 COL8A2 [10].
The COL8A2 gene is the only gene that has been associated with early-onset FECD and is rarely associated with late-onset FECD [13].
A genome-wide study proved different pathogenic variants in COL8A2 on chromosome 1p34.3-p32: R304Q, R434H, Q455K, and R155Q, [13]; the latter was proved as non-pathogenic later [14].
Mutations in the COL8A gene in the early-onset subtype affect the structure of the Descemet’s membrane. The COL8A2 gene encodes the 703 amino acid α2 subunit of collagen VIII, which is a short-chain collagen that is a component of Descemet membrane secreted by CECs [13].

4.2.2. Late-Onset Subtype

On the other hand, the far more frequent form, the late-onset FECD, is most characteristically underlined by the expansion of the CTG18.1 triplet in the TCF4 gene [10,40,41,42].
Namely, the mutations in the TCF4 gene are present in 70% of Caucasians with FECD.
Apart from these, SLC4A11 variants were proved to be another possible genetic cause, while the causal role of other candidates, such as ZEB1/TCF8, LOXHD1, and AGBL1, need still to be confirmed [17,43]. Some less common genetic mutations were also described in COL8A2, DMPK, KANK4, LAMC1, ATP1B1, RAD51, FEN1, XRCC1, NEIL1, TGFBI, CLU, PITX2, PTPRG, FASLG, and KCNJ13 [5].
The strongest genetic association with the clinical picture of FECD was proved for the CTG trinucleotide repeat within the transcription factor 4 (TCF4); a repeat length >50 was highly specific for the disease [18,42,43,44]. Following this significant correlation, expansion of this CTG repeat might be important in the mainstay of patients with developed disease. In early stages or clinically nonsignificant grades of pathological endothelial changes, diagnosing the trinucleotide expansion could help to predict the disease and lead to early therapeutic approach with minimally invasive techniques [18,44].
The genetic locus of early-onset and late-onset FECD have been mapped to different chromosomes (Table 2) [45].
TCF4 Gene
Most cases of FECD (late-onset) are caused by the expansion of trinucleotide repeats in the TCF4 gene. The expansion of greater than 40–50 repeats results in the production and accumulation of RNA inside the endothelial cell nuclei in the so-called RNA foci [21,46]. The toxic RNA causes sequestration of muscle blind-like protein MBNL-1 and MBNL2, splicing regulator proteins in nuclear RNA focuses. As a result of its loss of function, several miss-splicing transcripts involved in endothelial corneal cell homeostasis occur, leading to cell dysfunction and deterioration. The severity of the disease correlates to the length of the repeat [47].
As detailed in a comprehensive review of the research implicating CTG18.1 expansion in disease pathogenesis, the proposed mechanisms by which the CTG18.1 expansion may drive or exacerbate the onset of FECD include TCF4 dysregulation, toxic gain-of-function from TCF4 repeat-containing RNA, toxic gain-of-function from repeat-associated non-AUG-dependent translation, and somatic instability of CTG18.1 [41].
The CTG18.1 trinucleotide repeat is the most common genetic background in patients with FECD and can be confirmed in as frequent as 70–80% of individuals with pathological corneal findings [41], which is not the case for any other FECD-associated genetic mutations. Furthermore, the mutation has a direct pathogenic mechanism leading to cell deterioration and has already been studied in clinical settings as a possible diagnostic and prognostic biomarker [12]. The listed facts make this mutation a feasible option for the targeting of genetic therapy.
However, there are also several features specific to this mutation that need to be addressed, before a successful therapeutic strategy can be established. The trinucleotide repeat is placed in the intron 3 of TCF4 and thus does not result in an altered protein but rather, results in toxic RNA, for which a classic genetic gene replacement is not an option.
Large, repetitive DNA tracts also pose different problems for CRISPR-Cas9 editing, due to inefficient binding, possible off-target cleavage, difficulty in completing the excision of a large repeats, and unreliable repair mechanisms in non-dividing endothelial cells [41].
SLC4A11 Gene and Other
The SLC4A11 gene encodes an ion channel that promotes water resorption through the endothelial layer and is an important mediator of solute transport in the cornea.
Mutations in this gene can lead to corneal edema and correlate with FECD.
Similarly, mutations in the ZEB1 gene have been associated with late-onset of FECD and posterior polymorphous corneal dystrophy [48,49].

5. Genetic Treatment for FECD

The cornea seems like a possible target for developing genetic treatments as it is an immune-privileged, avascular, transparent, and relatively accessible tissue.
There are several well-established surgical techniques offering access to and manipulation of cells and layers of the cornea, but an efficient and targeted delivery of genetic material exactly to the corneal endothelial cells represents an additional new challenge. Challenges in establishing possible corneal genetic therapy include but are not limited to achieving sustained non-pathological gene transcription and minimizing off-target effects.

5.1. Established Treatments for FECD

To regain endothelial function and corneal transparency in patients with FECD, endothelial cells need to be repopulated in the areas of the diseased Descemet and dysfunctional or absent endothelial cells.
The golden standard of care is endothelial transplantation (Figure 2a) [50,51,52].
This is successful but technically demanding with surgical risks [53] of possible graft failure or rejection [54,55]. Above all, the constant global tissue shortage [56,57] results in prolonged waiting times for surgery with possible aggravation of the cornea status during the waiting period, resulting in more demanding treatment and worse outcomes [58,59].
Thus, several treatments that do not need a tissue transplantation emerged, including Descemetorhexis only (DSO) (Figure 2c) for candidates with preserved peripheral endothelium (Figure 1) [60,61], pharmacological regeneration [60,61], and cell therapy.

5.2. Genetic Therapy Principles in FECD

Since FECD is linked to specific mutations and the cornea has an immuno-privileged and relatively accessible position, it might be a possible disease for genetic therapy once the biological and technical challenges, including ensuring an efficient and targeted delivery of genetic material to the corneal endothelial cells, sustained gene expression, and minimal off-target effects, are successfully addressed.
Genetic treatment, which will only become clinically accepted following long and precise preclinical research steps, should, in general, be focused on the most prevalent genetic background of the disease.
For genetic therapy of FECD, the most possible target seems to be the TCF4 gene. Namely, the far more prevalent subtype of FECD is the late-onset form and for this subtype, it has been proven to be connected to four genetic loci (FCD1-4), different probable loci, and point mutations in a minimum of three other genes (ZEB1/TCF8, SLC4A11, and LOXHD1). Of these, the trinucleotide repeat in the TCF4 gene was shown to be the genetic cause of the majority (70%) of FECD cases [47].
Traditional gene therapy typically involves delivering functional copies of a gene to compensate for the effects of a mutated or non-functional version [62].
New approaches in molecular biology tend to repair the pathological genetic mutation in situ. Nucleases precisely cut targets in the host genome, which is the beginning of genome-editing.
Of them, the regularly clustered interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR-Cas9) system, which was originally discovered as the adaptive immune system of bacteria, reigns as a preeminent innovative technology [63] in human biomedicine due to its precision, high adaptability, programmability, efficacy, and relative cost effectiveness.
Namely, the gRNA in the Cas9-gRNA complex guides the Cas9 to cleave the DNA at a specific target location(s). Cas9 induces a double-stranded DNA break, followed by activation of cell mechanisms to repair the DNA by either non-homologous end joining (NHEJ) or homology-directed repair (HDR) pathways [64].
The CRISPR-Cas9 system proved successful in treating inherited retinal disorders [65]. Namely, a normal CEP290 gene expression was regained in an in vivo mouse study with Leber congenital amaurosis [66], followed by a clinical trial to evaluate the safety and effectiveness of EDIT-101, which is an experimental gene editing treatment [67]; approved gene therapy was later conducted for patients with biallelic mutations in RPE65 (Luxturna, Hoffmann-La Roche, Basel, Switzerland) [68].
However, use of this technology on the trinucleotide repeat diseases, such as FECD, presents specific and additional challenges.

5.3. Challenges of the CRISPR-Cas9 System in General

The main obstacle to clinical use is the efficient and precise delivery of the CRISPR-Cas9 complex to the target cells.
The golden standard of the delivery system in eye genetic therapy is recombinant adeno-associated viruses (AAVs). They are nonintegrating, with lower risk of unwanted mutation in host genome. There is a potential loss of effect over time, especially in dividing cells. An example of eye genetic treatment, that is AAV-based is the genetic therapy for retinitis pigmentosa, which has been approved by the FDA [69].
The capacity of AAV vectors is very limited and represents a limiting factor for the delivery, especially due to large size of the Cas9 complex [70]. For the genetic treatment of retinal diseases, solutions like smaller caspases [71] or dual-AAV split system [72,73] were introduced.
In any clinical use, the precise modification of genes is one of the most important safety concerns; thus, the possible off-target effects of the CRISPR-Cas9 therapy have to be solved in preclinical research [74]. Several technologies of tissue function have already been developed [75,76,77]. There is also existing software to predict possible off-target binding of RNA-guided endonucleases using sgRNA target sequences and the host genome. However, the significant variability of experimentally predicted verified off-target binding sites was proved [78]. Namely, in the field of ocular genetic CRISPR therapy, off-target control is based on guide selection [79], high-fidelity Cas9 variants [75,80] (like SpCas9, Cas9-HF, HypaCas9, and SniperCas9 [81]), dual-nickase or base/prime editing approaches [82], restricted Cas9 expression in tissue [83], and rigorous preclinical off-target profiling [79,84]. Before any clinical use, the off-target editing is further denied with the aid of preclinical safety studies [85].
Eye and ocular structures and tissues have immune-privileged status; however, due to the reported immunoreactivity to Cas9, validation of the safety of CRISPR-Cas9-based therapeutics in the clinical real world must be performed [69].

Specific Challenges of Applying CRISPR-Cas9 to TCF4

As discussed in Section 4.2.2, the most common mutation found in FECD is the expansion of trinucleotide repeats in the TCF4 gene with consequent production and accumulation of toxic RNA inside endothelial cell nuclei, and sequestration of splicing regulator protein, resulting in cell dysfunction and deterioration. The pathogenic trinucleotide expansion is located in the intron 3—a non-coding region of TCF4—which makes the design of effective editing strategies complicated. Namely, the mutation does not result in an altered protein but rather, a toxic RNA, for which a classic genetic gene replacement is not an option; rather, either the editing of toxic RNA production or regaining the normal expression of TGF4 gene are needed.
Applying the CRISPR-Cas9 to such mutation includes several technical challenges.
The non-coding region often expresses varying genetic structure, which represents a difficulty in finding effective guide sequences [86], resulting in inefficient binding and possible off-target cleavage. While large, repetitive DNA tracts result in difficulty in completing the excision of a large repeats, and unreliable repair mechanisms in non-dividing endothelial cells [41].
The allele-specific editing is also difficult due to the high similarity of wild-type and mutant alleles, which differ only by the repeat length [87,88].
To present a realistic picture, there is limited and indirect preclinical evidence supporting a successful CRISPR-mediated correction of TCF4 expansions in corneal endothelial cells [66,89,90].
The alternative RNA-targeting catalytically inactive or death Cas9 (dCAS9) proved a successful option in reducing toxic nuclear foci and mutant intronic RNA levels in FECD endothelial cells [91]. Other possible therapeutic strategies include the following: an antisense oligonucleotide principle could target the CUG or CAG RNA transcripts from pathological trinucleotide expansion and thus block the formation of toxic foci; the restoration of normal splicing could, as another strategy, be obtained via overexpression of the splicing factor (MBNL proteins); small molecule therapeutics could induce disruption of RNA hairpin; and some other possibilities were also suggested [41].

5.4. Real World Genetic Treatment for FECD

There are no clinically approved genetic treatments for FECD, nor are there any clinical trials ongoing yet.
With the latest evidence demonstrating that the pathogenic CTG trinucleotide repeat expansions in TCF4 is a frequent genetic background in patients with the clinical picture of FECD, the CRISPR-Cas9 technology for FECD holds significant promise in two aspects [47]: on one hand, as analogous to emerging genetic treatments for trinucleotide repeat expansion disorders such as Huntington’s disease or myotonic dystrophy; and on the other hand, similar to approved genetic therapy for other eye disorders, such as retinal.
There was a patent registered proposing the cornea as a site for CRISPR–Cas9-based therapeutic approaches [92].

5.4.1. DT-168

A small molecule named DT-168 was developed in the form of topical corneal therapy for FECD and aimed to inhibit the transcription of CTG18.1 into pathogenic TCF4 RNA, which transcribes into splicing proteins initiating transcript mis-splicing (spliceopathy), which eventually results in endothelial cell death [93].
Corneal endothelial cells were cultured from donors with a clinical diagnosis of FECD who underwent endothelial keratoplasty and were exposed to the DT-168 molecule. DT-168 significantly reduced nuclear foci to levels seen in cells cultured from healthy individuals. Exposition to DT-168 molecule also improved mis-splicing in vitro in different genes. Subsequent animal experiments treating rabbit eyes twice daily for two weeks proved good tolerability and even corneal distribution; it also proved persistent corneal concentration for 24 h, with negligible plasma levels of the molecule [93].
In conclusion, DT-168 in eye drops could be useful as disease-modifying therapy attaching the most frequent genetic change in FECD.
Results of a phase I clinical study on healthy volunteers are expected in October 2025; however, there are preliminary reports of good tolerability without serious adverse events and there are no treatment discontinuations due to AEs in the trial.
A phase 2 biomarker trial of DT-168 was set to study safety, tolerability, and corneal endothelium biomarkers in FECD patients scheduled for corneal transplant surgery [94].

5.4.2. Ad-Cas9-Col8a2gRNA

An animal study was performed proving efficient knock down of mutant COL8A2 expression in endothelial cells of FECD model mice [95].
Namely, adenovirus vector was used to introduce Cas9 gene and RNA-Ad-Cas9-Col8a2gRNA as a single intraocular injection. Using this postnatal gene editing prevented endothelial cell loss and improved their pump function without adverse effects on histological or electroretinographically exams [95].
This study proved that Ad-Cas9-Col8a2gRNA disruption of the Col8a2 start codon restores the normal endothelial cell phenotype in adult post-mitotic non-reproducing cells and could represent a medical genetic therapy of adult-onset diseases like FECD.

6. Discussion

Compared to the retina, the cornea, which is the outermost tissue of the eye, has the advantage of being easily accessible to different treatment delivery options such as drops [93] or intracameral injections [95]. There are also different possibilities of precise treatment evaluation via slit lamp clinical exam and advanced diagnostic methods such as specular or corneal confocal microscopy of endothelial cells, corneal optical coherence tomography, etc. The application of CRISPR/Cas9 ocular genome treatments in clinical use is on the rise, and corneal diseases will hopefully become part of this in the near future [96,97].
FECD is a devastating disease, which represents one of the most frequent reasons for the need for corneal transplantation. This procedure, however, is not readily accessible to patients in need due to global corneal tissue shortage, while procedures without donor tissue, such as DSO, are only suitable for patients with early-grade disease. Thus, a non-surgical, readily accessible, and economically affordable treatment is needed. The inherited mutations in FECD are well-researched, the cornea represents an easy to approach and immune-privileged tissue and seems very suitable for CRISPR-Cas9 therapy.
There are already molecules developed that can stop the pathological vicious circle in FECD-affected post-mitotic endothelial cells, such as the DT-168 or the RNA-Ad-Cas9-Col8a2gRNA, and have been tested in preclinical, and some as well in phase I clinical studies, and have entered phase II clinical studies [93,95]. However, there are still certain challenges as compared to already approved genetic therapies for retinal diseases [68,98]. Namely, their delivery and duration of effect, as well as clinical efficiency due to the polygenic nature of FECD are challenges to be solved.
Due to their post-mitotic state, corneal endothelial cells might be more difficult to be reached by adenoviral vectors [99], while herpes viral vector or non-viral vectors appear to be not efficient enough [100,101] to reach the endothelial cell interior.
FECD develops due to toxic gain-of-function after trinucleotide repeat resulting in protein misfolding, which is different to the retinal pigment epithelium (RPE) loss of function in the RPE65-mediated inherited retinal dystrophy [68]. FECD patients thus need long-term suppression of toxic gene activity while one-time treatment is enough to repair the pathological gene with Luxturna [68].
The polyfactorial nature of FECD will continue to be the main clinical challenge, which will eventually demand a reliable genetic test to select patients suitable for genetic treatment.

7. Conclusions

There are well-established treatments for the FECD but due to rising numbers of patients unmet by the tissue supply shortage and with the debilitating effect on vision and quality of life of untreated patients, the need for advanced treatment is clear.
Results of ongoing research in genetic treatment possibilities are optimistic; however, the review of the literature proved that they are still at the preclinical stage for FECD and that there are several specific challenges connected that still need to be solved before realistic clinical usage is expected.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AAVadeno-associated viruses
Cas9CRISPR-associated 9
CECscorneal endothelial cells
CRISPRclustered regularly interspaced short palindromic repeats
DMEKDescemet membrane endothelial keratoplasty
DSOdescemetorhexis only
FECDFuchs endothelial corneal dystrophy
RPEretinal pigment epithelium

References

  1. Yi, L.Y.; Hsieh, H.H.; Lin, Z.Q.; Hung, K.F.; Sun, Y.C. Exploring the Role of ROCK Inhibition in Corneal Edema Through Crosstalk Between Epithelial and Endothelial Cells. J. Ophthalmol. 2024, 5, 9381303. [Google Scholar] [CrossRef] [PubMed]
  2. Kinoshita, S.; Koizumi, N.; Ueno, M.; Okumura, N.; Imai, K.; Tanaka, H.; Yamamoto, Y.; Nakamura, T.; Inatomi, T.; Bush, J.; et al. Injection of Cultured Cells with a ROCK Inhibitor for Bullous Keratopathy. N. Engl. J. Med. 2018, 15, 995–1003. [Google Scholar] [CrossRef] [PubMed]
  3. Kinoshita, S.; Ueno, M.; Toda, M.; Imai, K.; Tomioka, Y.; Numa, K.; Tanaka, H.; Inatomi, T.; Kameda, T.; Tsujikawa, A.; et al. Long-Term Corneal Rejuvenation after Transplantation of Cultured Human Corneal Endothelial Cells. Ophthalmology 2025, 132, 1105–1113. [Google Scholar] [CrossRef]
  4. Leroy, B.P.; Fischer, M.D.; Flannery, J.G.; MacLaren, R.E.; Dalkara, D.; Scholl, H.P.N.; Chung, D.C.; Spera, C.; Viriato, D.; Banhazi, J. Gene Therapy for Inherited Retinal Disease: Long-Term Durability of Effect. Ophthalmic Res. 2023, 66, 179–196. [Google Scholar] [CrossRef] [PubMed]
  5. Ong Tone, S.; Kocaba, V.; Böhm, M.; Wylegala, A.; White, T.L.; Jurkunas, U.V. Fuchs endothelial corneal dystrophy: The vicious cycle of Fuchs pathogenesis. Prog. Retin. Eye Res. 2021, 80, 100863. [Google Scholar] [CrossRef]
  6. Oie, Y.; Watanabe, S.; Nishida, K. Evaluation of Visual Quality in Patients with Fuchs Endothelial Corneal Dystrophy. Cornea 2016, 35 (Suppl. S1), S55–S58. [Google Scholar] [CrossRef]
  7. Bhadra, P.; Sahoo, S.; Sahu, S.K.; Priyadarshini, S.; Mohanty, A.; Das, S. Demographic profile and clinical characteristics of Fuchs’ endothelial corneal dystrophy in a tertiary eye care center. Indian J. Ophthalmol. 2023, 71, 505–509. [Google Scholar] [CrossRef]
  8. Watanabe, S.; Oie, Y.; Fujimoto, H.; Soma, T.; Koh, S.; Tsujikawa, M.; Maeda, N.; Nishida, K. Relationship between corneal guttae and quality of vision in patients with mild fuchs’ endothelial corneal dystrophy. Ophthalmology 2015, 122, 2103–2109. [Google Scholar] [CrossRef]
  9. Krachmer, J.H.; Purcell, J.J., Jr.; Young, C.W.; Bucher, K.D. Corneal endothelial dystrophy. A study of 64 families. Arch. Ophthalmol. 1978, 96, 2036–2039. [Google Scholar] [CrossRef]
  10. Zhang, J.; McGhee, C.N.J.; Patel, D.V. The Molecular Basis of Fuchs’ Endothelial Corneal Dystrophy. Mol. Diagn. Ther. 2019, 23, 97–112. [Google Scholar] [CrossRef]
  11. Price, M.O.; Mehta, J.S.; Jurkunas, U.V.; Price, F.W., Jr. Corneal endothelial dysfunction: Evolving understanding and treatment options. Prog. Retin. Eye Res. 2021, 82, 100904. [Google Scholar] [CrossRef] [PubMed]
  12. Nanda, G.G.; Alone, D.P. REVIEW: Current understanding of the pathogenesis of Fuchs’ endothelial corneal dystrophy. Mol. Vis. 2019, 5, 295–310. [Google Scholar]
  13. Biswas, S.; Munier, F.L.; Yardley, J.; Hart-Holden, N.; Perveen, R.; Cousin, P.; Sutphin, J.E.; Noble, B.; Batterbury, M.; Kielty, C.; et al. Missense mutations in COL8A2, the gene encoding the alpha2 chain of type VIII collagen, cause two forms of corneal endothelial dystrophy. Hum. Mol. Genet. 2001, 10, 2415–2423. [Google Scholar] [CrossRef] [PubMed]
  14. Kobayashi, A.; Fujiki, K.; Murakami, A.; Kato, T.; Chen, L.Z.; Onoe, H.; Nakayasu, K.; Sakurai, M.; Takahashi, M.; Sugiyama, K.; et al. Analysis of COL8A2 gene mutation in Japanese patients with Fuchs’ endothelial dystrophy and posterior polymorphous dystrophy. Jpn. J. Ophthalmol. 2004, 48, 195–198. [Google Scholar] [CrossRef] [PubMed]
  15. Gottsch, J.D.; Sundin, O.H.; Rencs, E.V.; Emmert, D.G.; Stark, W.J.; Cheng, C.J.; Schmidt, G.W. Analysis and documentation of progression of Fuchs corneal dystrophy with retroillumination photography. Cornea 2006, 25, 485–489. [Google Scholar] [CrossRef]
  16. Liskova, P.; Prescott, Q.; Bhattacharya, S.S.; Tuft, S.J. British family with early-onset Fuchs’ endothelial corneal dystrophy associated with p.L450W mutation in the COL8A2 gene. Br. J. Ophthalmol. 2007, 91, 1717–1718. [Google Scholar] [CrossRef]
  17. Mootha, V.V.; Gong, X.; Ku, H.C.; Xing, C. Association and familial segregation of CTG18.1 trinucleotide repeat expansion of TCF4 gene in Fuchs’ endothelial corneal dystrophy. Investig. Ophthalmol. Vis. Sci. 2014, 55, 33–42. [Google Scholar] [CrossRef]
  18. Wieben, E.D.; Aleff, R.A.; Tosakulwong, N.; Butz, M.L.; Highsmith, W.E.; Edwards, A.O.; Baratz, K.H. A common trinucleotide repeat expansion within the transcription factor 4 (TCF4, E2-2) gene predicts Fuchs corneal dystrophy. PLoS ONE 2012, 7, e49083. [Google Scholar] [CrossRef]
  19. Baratz, K.H.; Tosakulwong, N.; Ryu, E.; Brown, W.L.; Branham, K.; Chen, W.; Tran, K.D.; Schmid-Kubista, K.E.; Heckenlively, J.R.; Swaroop, A.; et al. E2-2 protein and Fuchs’s corneal dystrophy. N. Engl. J. Med. 2010, 363, 1016–1024. [Google Scholar] [CrossRef]
  20. Afshari, N.A.; Igo RPJr Morris, N.J.; Stambolian, D.; Sharma, S.; Pulagam, V.L.; Dunn, S.; Stamler, J.F.; Truitt, B.J.; Rimmler, J.; Kuot, A.; et al. Genome-wide association study identifies three novel loci in Fuchs endothelial corneal dystrophy. Nat. Commun. 2017, 8, 14898. [Google Scholar] [CrossRef]
  21. Mootha, V.V.; Hansen, B.; Rong, Z.; Mammen, P.P.; Zhou, Z.; Xing, C.; Gong, X. Fuchs’ Endothelial Corneal Dystrophy and RNA Foci in Patients with Myotonic Dystrophy. Investig. Ophthalmol. Vis. Sci. 2017, 58, 4579–4585. [Google Scholar] [CrossRef]
  22. Winkler, N.S.; Milone, M.; Martinez-Thompson, J.M.; Raja, H.; Aleff, R.A.; Patel, S.V.; Fautsch, M.P.; Wieben, E.D.; Baratz, K.H. Fuchs’ Endothelial Corneal Dystrophy in Patients with Myotonic Dystrophy, Type 1. Investig. Ophthalmol. Vis. Sci. 2018, 59, 3053–3057. [Google Scholar] [CrossRef] [PubMed]
  23. Vithana, E.N.; Morgan, P.E.; Ramprasad, V.; Tan, D.T.; Yong, V.H.; Venkataraman, D.; Venkatraman, A.; Yam, G.H.; Nagasamy, S.; Law, R.W.; et al. SLC4A11 mutations in Fuchs endothelial corneal dystrophy. Hum. Mol. Genet. 2008, 17, 656–666. [Google Scholar] [CrossRef] [PubMed]
  24. Riazuddin, S.A.; Vithana, E.N.; Seet, L.F.; Liu, Y.; Al-Saif, A.; Koh, L.W.; Heng, Y.M.; Aung, T.; Meadows, D.N.; Eghrari, A.O.; et al. Missense mutations in the sodium borate cotransporter SLC4A11 cause late-onset Fuchs corneal dystrophy. Hum. Mutat. 2010, 31, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
  25. Soumittra, N.; Loganathan, S.K.; Madhavan, D.; Ramprasad, V.L.; Arokiasamy, T.; Sumathi, S.; Karthiyayini, T.; Rachapalli, S.R.; Kumaramanickavel, G.; Casey, J.R.; et al. Biosynthetic and functional defects in newly identified SLC4A11 mutants and absence of COL8A2 mutations in Fuchs endothelial corneal dystrophy. J. Hum. Genet. 2014, 59, 444–453. [Google Scholar] [CrossRef]
  26. Mehta, J.S.; Vithana, E.N.; Tan, D.T.; Yong, V.H.; Yam, G.H.; Law, R.W.; Chong, W.G.; Pang, C.P.; Aung, T. Analysis of the posterior polymorphous corneal dystrophy 3 gene, TCF8, in late-onset Fuchs endothelial corneal dystrophy. Investig. Ophthalmol. Vis. Sci. 2008, 49, 184–188. [Google Scholar] [CrossRef]
  27. Gupta, R.; Kumawat, B.L.; Paliwal, P.; Tandon, R.; Sharma, N.; Sen, S.; Kashyap, S.; Nag, T.C.; Vajpayee, R.B.; Sharma, A. Association of ZEB1 and TCF4 rs613872 changes with late onset Fuchs endothelial corneal dystrophy in patients from northern India. Mol. Vis. 2015, 21, 1252–1260. [Google Scholar]
  28. Riazuddin, S.A.; Parker, D.S.; McGlumphy, E.J.; Oh, E.C.; Iliff, B.W.; Schmedt, T.; Jurkunas, U.; Schleif, R.; Katsanis, N.; Gottsch, J.D. Mutations in LOXHD1, a recessive-deafness locus, cause dominant late-onset Fuchs corneal dystrophy. Am. J. Hum. Genet. 2012, 90, 533–539. [Google Scholar] [CrossRef]
  29. Riazuddin, S.A.; Vasanth, S.; Katsanis, N.; Gottsch, J.D. Mutations in AGBL1 cause dominant late-onset Fuchs corneal dystrophy and alter protein-protein interaction with TCF4. Am. J. Hum. Genet. 2013, 93, 758–764. [Google Scholar] [CrossRef]
  30. Wieben, E.D.; Aleff, R.A.; Tang, X.; Kalari, K.R.; Maguire, L.J.; Patel, S.V.; Baratz, K.H.; Fautsch, M.P. Gene expression in the corneal endothelium of Fuchs endothelial corneal dystrophy patients with and without expansion of a trinucleotide repeat in TCF4. PLoS ONE 2018, 13, e0200005. [Google Scholar] [CrossRef]
  31. Synowiec, E.; Wojcik, K.A.; Izdebska, J.; Binczyk, E.; Blasiak, J.; Szaflik, J.; Szaflik, J.P. Polymorphisms of the homologous recombination gene RAD51 in keratoconus and Fuchs endothelial corneal dystrophy. Dis. Markers 2013, 35, 353–362. [Google Scholar] [CrossRef]
  32. Wojcik, K.A.; Synowiec, E.; Polakowski, P.; Głowacki, S.; Izdebska, J.; Lloyd, S.; Galea, D.; Blasiak, J.; Szaflik, J.; Szaflik, J.P. Polymorphism of the flap endonuclease 1 gene in keratoconus and Fuchs endothelial corneal dystrophy. Int. J. Mol. Sci. 2014, 15, 14786–14802. [Google Scholar] [CrossRef] [PubMed]
  33. Wójcik, K.A.; Synowiec, E.; Polakowski, P.; Błasiak, J.; Szaflik, J.; Szaflik, J.P. Variation in DNA Base Excision Repair Genes in Fuchs Endothelial Corneal Dystrophy. Med. Sci. Monit. 2015, 21, 2809–2827. [Google Scholar] [CrossRef] [PubMed]
  34. Synowiec, E.; Wojcik, K.A.; Izdebska, J.; Binczyk, E.; Szaflik, J.; Blasiak, J.; Szaflik, J.P. Polymorphism of the LIG3 gene in keratoconus and Fuchs endothelial corneal dystrophy. Cell Mol. Biol. 2015, 61, 56–63. [Google Scholar] [PubMed]
  35. Kuot, A.; Hewitt, A.W.; Griggs, K.; Klebe, S.; Mills, R.; Jhanji, V.; Craig, J.E.; Sharma, S.; Burdon, K.P. Association of TCF4 and CLU polymorphisms with Fuchs’ endothelial dystrophy and implication of CLU and TGFBI proteins in the disease process. Eur. J. Hum. Genet. 2012, 20, 632–638. [Google Scholar] [CrossRef]
  36. Kniestedt, C.; Taralczak, M.; Thiel, M.A.; Stuermer, J.; Baumer, A.; Gloor, B.P. A novel PITX2 mutation and a polymorphism in a 5-generation family with Axenfeld-Rieger anomaly and coexisting Fuchs’ endothelial dystrophy. Ophthalmology 2006, 113, e1–e8. [Google Scholar] [CrossRef]
  37. Li, Y.J.; Minear, M.A.; Qin, X.; Rimmler, J.; Hauser, M.A.; Allingham, R.R.; Igo, R.P.; Lass, J.H.; Iyengar, S.K.; Klintworth, G.K.; et al. Mitochondrial polymorphism A10398G and Haplogroup I are associated with Fuchs’ endothelial corneal dystrophy. Investig. Ophthalmol. Vis. Sci. 2014, 55, 4577–4584. [Google Scholar] [CrossRef]
  38. Li, Q.J.; Ashraf, M.F.; Shen, D.F.; Green, W.R.; Stark, W.J.; Chan, C.C.; O’Brien, T.P. The role of apoptosis in the pathogenesis of Fuchs endothelial dystrophy of the cornea. Arch. Ophthalmol. 2001, 119, 1597–1604. [Google Scholar] [CrossRef]
  39. Hejtmancik, J.F.; Jiao, X.; Li, A.; Sergeev, Y.V.; Ding, X.; Sharma, A.K.; Chan, C.C.; Medina, I.; Edwards, A.O. Mutations in KCNJ13 cause autosomal-dominant snowflake vitreoretinal degeneration. Am. J. Hum. Genet. 2008, 82, 174–180. [Google Scholar] [CrossRef]
  40. Foja, S.; Luther, M.; Hoffmann, K.; Rupprecht, A.; Gruenauer-Kloevekorn, C. CTG18.1 repeat expansion may reduce TCF4 gene expression in corneal endothelial cells of German patients with Fuchs’ dystrophy. Graefes Arch. Clin. Exp. Ophthalmol. 2017, 255, 1621–1631. [Google Scholar] [CrossRef]
  41. Fautsch, M.P.; Wieben, E.D.; Baratz, K.H.; Bhattacharyya, N.; Sadan, A.N.; Hafford-Tear, N.J.; Tuft, S.J.; Davidson, A.E. TCF4-mediated Fuchs endothelial corneal dystrophy: Insights into a common trinucleotide repeat-associated disease. Prog. Retin. Eye Res. 2021, 81, 100883. [Google Scholar] [CrossRef]
  42. Xing, C.; Gong, X.; Hussain, I.; Khor, C.C.; Tan, D.T.; Aung, T.; Mehta, J.S.; Vithana, E.N.; Mootha, V.V. Transethnic replication of association of CTG18.1 repeat expansion of TCF4 gene with Fuchs’ corneal dystrophy in Chinese implies common causal variant. Investig. Ophthalmol. Vis. Sci. 2014, 55, 7073–7078. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Vasanth, S.; Eghrari, A.O.; Gapsis, B.C.; Wang, J.; Haller, N.F.; Stark, W.J.; Katsanis, N.; Riazuddin, S.A.; Gottsch, J.D. Expansion of CTG18.1 Trinucleotide Repeat in TCF4 Is a Potent Driver of Fuchs’ Corneal Dystrophy. Investig. Ophthalmol. Vis. Sci. 2015, 56, 4531–4536. [Google Scholar] [CrossRef] [PubMed]
  44. Tsedilina, T.R.; Sharova, E.; Iakovets, V.; Skorodumova, L.O. Systematic review of SLC4A11, ZEB1, LOXHD1, and AGBL1 variants in the development of Fuchs’ endothelial corneal dystrophy. Front. Med. 2023, 10, 1153122. [Google Scholar] [CrossRef] [PubMed]
  45. Weiss, J.S.; Møller, H.U.; Aldave, A.J.; Seitz, B.; Bredrup, C.; Kivelä, T.; Munier, F.L.; Rapuano, C.J.; Nischal, K.K.; Kim, E.K.; et al. IC3D classification of corneal dystrophies-edition 2. Cornea 2015, 34, 117–159, Erratum in Cornea 2015, 34, e32; Erratum in Cornea 2022, 41, e23. [Google Scholar] [CrossRef]
  46. Du, J.; Aleff, R.A.; Soragni, E.; Kalari, K.; Nie, J.; Tang, X.; Davila, J.; Kocher, J.P.; Patel, S.V.; Gottesfeld, J.M.; et al. RNA toxicity and missplicing in the common eye disease fuchs endothelial corneal dystrophy. J. Biol. Chem. 2015, 290, 5979–5990. [Google Scholar] [CrossRef]
  47. Wieben, E.D.; Aleff, R.A.; Tang, X.; Butz, M.L.; Kalari, K.R.; Highsmith, E.W.; Jen, J.; Vasmatzis, G.; Patel, S.V.; Maguire, L.J.; et al. Trinucleotide Repeat Expansion in the Transcription Factor 4 (TCF4) Gene Leads to Widespread mRNA Splicing Changes in Fuchs’ Endothelial Corneal Dystrophy. Investig. Ophthalmol. Vis. Sci. 2017, 58, 343–352. [Google Scholar] [CrossRef]
  48. Gurnani, B.; Somani, A.N.; Moshirfar, M.; Patel, B.C. Fuchs Endothelial Dystrophy. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  49. Malhotra, D.; Loganathan, S.K.; Chiu, A.M.; Lukowski, C.M.; Casey, J.R. Human Corneal Expression of SLC4A11, a Gene Mutated in Endothelial Corneal Dystrophies. Sci. Rep. 2019, 9, 9681. [Google Scholar] [CrossRef]
  50. Tourabaly, M.; Knoeri, J.; Georgeon, C.; Borderie, V. Review of the Literature: Surgery Indications for Fuchs’ Endothelial Corneal Dystrophy. J. Clin. Med. 2025, 14, 2365. [Google Scholar] [CrossRef]
  51. Hurley, D.J.; Murtagh, P.; Guerin, M. Ultrathin Descemet Stripping Automated Endothelial Keratoplasty (UT-DSAEK) versus Descemet Membrane Endothelial Keratoplasty (DMEK)-a systematic review and meta-analysis. Eye 2023, 37, 3026–3032. [Google Scholar] [CrossRef]
  52. Stuart, A.J.; Romano, V.; Virgili, G.; Shortt, A.J. Descemet’s membrane endothelial keratoplasty (DMEK) versus Descemet’s stripping automated endothelial keratoplasty (DSAEK) for corneal endothelial failure. Cochrane Database Syst. Rev. 2018, 6, CD012097. [Google Scholar] [CrossRef]
  53. Gorovoy, M.S. DMEK Complications. Cornea 2014, 33, 101–104. [Google Scholar] [CrossRef] [PubMed]
  54. Mechels, K.B.; Greenwood, M.D.; Sudhagoni, R.G.; Berdahl, J.P. Influences on rebubble rate in Descemet’s membrane endothelial keratoplasty. Clin. Ophthalmol. 2017, 11, 2139–2144. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Guerra, F.P.; Anshu, A.; Price, M.O.; Giebel, A.W.; Price, F.W. Descemet’s membrane endothelial keratoplasty: Prospective study of 1-year visual outcomes, graft survival, and endothelial cell loss. Ophthalmology 2011, 118, 2368–2373. [Google Scholar] [CrossRef] [PubMed]
  56. Machin, H.; Sutton, G.; Baird, P.N. Examining Corneal Tissue Exportation Fee and Its Impact on Equitable Allocation. Cornea 2022, 41, 390–395. [Google Scholar] [CrossRef]
  57. Gain, P.; Jullienne, R.; He, Z.; Aldossary, M.; Acquart, S.; Cognasse, F.; Thuret, G. Global Survey of Corneal Transplantation and Eye Banking. JAMA Ophthalmol. 2016, 134, 167–173. [Google Scholar] [CrossRef]
  58. Schrittenlocher, S.; Bachmann, B.; Tiurbe, A.M.; Tuac, O.; Velten, K.; Schmidt, D.; Cursiefen, C. Impact of preoperative visual acuity on Descemet Membrane Endothelial Keratoplasty (DMEK) outcome. Graefes Arch. Clin. Exp. Ophthalmol. 2019, 257, 321–329. [Google Scholar] [CrossRef]
  59. Friedrich, M.; Son, H.S.; Lind, J.; Hammer, M.; Chychko, L.; Yildirim, T.M.; Auffarth, G.U.; Augustin, V.A. Preoperative edema severity affects outcomes after Descemet membrane endothelial keratoplasty for Fuchs endothelial corneal dystrophy: A cohort study. Eye Vis. 2025, 12, 9. [Google Scholar] [CrossRef]
  60. Garcerant, D.; Hirnschall, N.; Toalster, N.; Zhu, M.; Wen, L.; Moloney, G. Descemet’s stripping without endothelial keratoplasty. Curr. Opin. Ophthalmol. 2019, 30, 275–285. [Google Scholar] [CrossRef]
  61. Borkar, D.S.; Veldman, P.; Colby, K.A. Treatment of Fuchs Endothelial Dystrophy by Descemet Stripping without Endothelial Keratoplasty. Cornea 2016, 35, 1267–1273. [Google Scholar] [CrossRef]
  62. Williams, K.A.; Irani, Y.D. Gene Therapy and Gene Editing for the Corneal Dystrophies. Asia Pac. J. Ophthalmol. 2016, 5, 312–316. [Google Scholar] [CrossRef]
  63. Tetsch, L. The adaptive bacterial immune system CRISPR-Cas and its therapeutic potential. Med. Monatsschr. Pharm. 2017, 40, 17–23. [Google Scholar] [PubMed]
  64. Hsu, P.D.; Lander, E.S.; Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 2014, 157, 1262–1278. [Google Scholar] [CrossRef] [PubMed]
  65. Cabral, T.; DiCarlo, J.E.; Justus, S.; Sengillo, J.D.; Xu, Y.; Tsang, S.H. CRISPR applications in ophthalmologic genome surgery. Curr. Opin. Ophthalmol. 2017, 28, 252–259. [Google Scholar] [CrossRef] [PubMed]
  66. Maeder, M.L.; Stefanidakis, M.; Wilson, C.J.; Baral, R.; Barrera, L.A.; Bounoutas, G.S.; Bumcrot, D.; Chao, H.; Ciulla, D.M.; DaSilva, J.A.; et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat. Med. 2019, 25, 229–233. [Google Scholar] [CrossRef] [PubMed]
  67. Pierce, E.A.; Aleman, T.S.; Jayasundera, K.T.; Ashimatey, B.S.; Kim, K.; Rashid, A.; Jaskolka, M.C.; Myers, R.L.; Lam, B.L.; Bailey, S.T.; et al. Gene Editing for CEP290-Associated Retinal Degeneration. N. Engl. J. Med. 2024, 390, 1972–1984. [Google Scholar] [CrossRef]
  68. Russell, S.; Bennett, J.; Wellman, J.A.; Chung, D.C.; Yu, Z.F.; Tillman, A.; Wittes, J.; Pappas, J.; Elci, O.; McCague, S.; et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: A randomised, controlled, open-label, phase 3 trial. Lancet 2017, 390, 849–860, Erratum in Lancet 2017, 390, 848. [Google Scholar] [CrossRef]
  69. Dunbar, C.E.; High, K.A.; Joung, J.K.; Kohn, D.B.; Ozawa, K.; Sadelain, M. Gene therapy comes of age. Science 2018, 359, eaan4672. [Google Scholar] [CrossRef]
  70. Wang, D.; Zhang, F.; Gao, G. CRISPR-Based Therapeutic Genome Editing: Strategies and In Vivo Delivery by AAV Vectors. Cell 2020, 181, 136–150. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  71. Nishiguchi, K.M.; Fujita, K.; Miya, F.; Katayama, S.; Nakazawa, T. Single AAV-mediated mutation replacement genome editing in limited number of photoreceptors restores vision in mice. Nat. Commun. 2020, 11, 482. [Google Scholar] [CrossRef]
  72. Li, F.; Wing, K.; Wang, J.H.; Luu, C.D.; Bender, J.A.; Chen, J.; Wang, Q.; Lu, Q.; Nguyen Tran, M.T.; Young, K.M.; et al. Comparison of CRISPR/Cas Endonucleases for in vivo Retinal Gene Editing. Front. Cell. Neurosci. 2020, 14, 570917. [Google Scholar] [CrossRef]
  73. Gallego, C.; Gonçalves, M.A.F.V.; Wijnholds, J. Novel Therapeutic Approaches for the Treatment of Retinal Degenerative Diseases: Focus on CRISPR/Cas-Based Gene Editing. Front. Neurosci. 2020, 14, 838. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  74. Fu, Y.; Foden, J.A.; Khayter, C.; Maeder, M.L.; Reyon, D.; Joung, J.K.; Sander, J.D. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 2013, 31, 822–826. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, X.; Yang, D.; Zhang, J.; Xu, J.; Chen, Y.E. Recent Advances in Improving Gene-Editing Specificity through CRISPR-Cas9 Nuclease Engineering. Cells 2022, 11, 2186. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  76. Cai, W.; Luo, T.; Mao, L.; Wang, M. Spatiotemporal Delivery of CRISPR/Cas9 Genome Editing Machinery Using Stimuli-Responsive Vehicles. Angew. Chem. Int. Ed. Engl. 2021, 60, 8596–8606. [Google Scholar] [CrossRef]
  77. Hasanzadeh, A.; Noori, H.; Jahandideh, A.; Haeri Moghaddam, N.; Kamrani Mousavi, S.M.; Nourizadeh, H.; Saeedi, S.; Karimi, M.; Hamblin, M.R. Smart Strategies for Precise Delivery of CRISPR/Cas9 in Genome Editing. ACS Appl. Bio Mater. 2022, 5, 413–437. [Google Scholar] [CrossRef]
  78. Tsai, S.Q.; Zheng, Z.; Nguyen, N.T.; Liebers, M.; Topkar, V.V.; Thapar, V.; Wyvekens, N.; Khayter, C.; Iafrate, A.J.; Le, L.P.; et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 2015, 33, 187–197. [Google Scholar] [CrossRef]
  79. Quinn, J.; Musa, A.; Kantor, A.; McClements, M.E.; Cehajic-Kapetanovic, J.; MacLaren, R.E.; Xue, K. Genome-Editing Strategies for Treating Human Retinal Degenerations. Hum. Gene Ther. 2021, 32, 247–259. [Google Scholar] [CrossRef]
  80. Naeem, M.; Majeed, S.; Hoque, M.Z.; Ahmad, I. Latest Developed Strategies to Minimize the Off-Target Effects in CRISPR-Cas-Mediated Genome Editing. Cells 2020, 9, 1608. [Google Scholar] [CrossRef]
  81. Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.; Joung, J.K. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef]
  82. Klermund, J.; Rhiel, M.; Kocher, T.; Chmielewski, K.O.; Bischof, J.; Andrieux, G.; El Gaz, M.; Hainzl, S.; Boerries, M.; Cornu, T.I.; et al. On- and off-target effects of paired CRISPR-Cas nickase in primary human cells. Mol. Ther. 2024, 32, 1298–1310. [Google Scholar] [CrossRef]
  83. Sundaresan, Y.; Yacoub, S.; Kodati, B.; Amankwa, C.E.; Raola, A.; Zode, G. Therapeutic applications of CRISPR/Cas9 gene editing technology for the treatment of ocular diseases. FEBS J. 2023, 290, 5248–5269. [Google Scholar] [CrossRef]
  84. Koo, T.; Lee, J.; Kim, J.S. Measuring and Reducing Off-Target Activities of Programmable Nucleases Including CRISPR-Cas9. Mol. Cells 2015, 38, 475–481. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  85. Hernández-Juárez, J.; Rodríguez-Uribe, G.; Borooah, S. Toward the Treatment of Inherited Diseases of the Retina Using CRISPR-Based Gene Editing. Front. Med. 2021, 8, 698521. [Google Scholar] [CrossRef] [PubMed]
  86. Müller Paul, H.; Istanto, D.D.; Heldenbrand, J.; Hudson, M.E. CROPSR: An automated platform for complex genome-wide CRISPR gRNA design and validation. BMC Bioinform. 2022, 23, 74. [Google Scholar] [CrossRef] [PubMed]
  87. Monteys, A.M.; Ebanks, S.A.; Keiser, M.S.; Davidson, B.L. CRISPR/Cas9 Editing of the Mutant Huntingtin Allele In Vitro and In Vivo. Mol. Ther. 2017, 25, 12–23. [Google Scholar] [CrossRef]
  88. Christie, K.A.; Robertson, L.J.; Conway, C.; Blighe, K.; DeDionisio, L.A.; Chao-Shern, C.; Kowalczyk, A.M.; Marshall, J.; Turnbull, D.; Nesbit, M.A.; et al. Mutation-Independent Allele-Specific Editing by CRISPR-Cas9, a Novel Approach to Treat Autosomal Dominant Disease. Mol. Ther. 2020, 28, 1846–1857. [Google Scholar] [CrossRef]
  89. Oyama, Y.; Ito, S.; Yuasa, T.; Ueda, M.; Chiba, S.; Nakagawa, T.; Izumi, A.; Ikawa, M.; Koizumi, N.; Okumura, N. Generation of a Mouse Model of Fuchs Endothelial Corneal Dystrophy by Knock-in of CTG Trinucleotide Repeat Expansion in the TCF4 Gene. Investig. Ophthalmol. Vis. Sci. 2025, 66, 18. [Google Scholar] [CrossRef]
  90. Okumura, N.; Hayashi, R.; Nakano, M.; Yoshii, K.; Tashiro, K.; Sato, T.; Blake, D.J.; Aleff, R.; Butz, M.; Highsmith, E.W.; et al. Effect of Trinucleotide Repeat Expansion on the Expression of TCF4 mRNA in Fuchs’ Endothelial Corneal Dystrophy. Investig. Ophthalmol. Vis. Sci. 2019, 60, 779–786. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  91. Rong, Z.; Gong, X.; Hulleman, J.D.; Corey, D.R.; Mootha, V.V. Trinucleotide Repeat-Targeting dCas9 as a Therapeutic Strategy for Fuchs’ Endothelial Corneal Dystrophy. Transl. Vis. Sci. Technol. 2020, 9, 47. [Google Scholar] [CrossRef]
  92. Jun, A.S.; Ranganathan, V.; Zack, D. CRISPR/CAS9-Based Treatments. WO2017004616A1, 5 January 2017. Available online: https://patentimages.storage.googleapis.com/f1/03/93/8e4d244e564db4/WO2017004616A1.pdf (accessed on 1 August 2025).
  93. Powers, A.; Cheung, K.; Osgood, N.; Kudwa, A.; Shepard, P.; Datta, S.; Safadi, M.; Sinha, S.; Kerr, J.; Zhang, C.; et al. Pharmacological and molecular features of DT-168, a topical GeneTACTM small molecule being developed as potential treatment for Fuchs Endothelial Corneal Dystrophy caused by CTG repeat expansions in the TCF4 gene. Investig. Ophthalmol. Vis. Sci. 2023, 64, 1333. [Google Scholar]
  94. Fuchs Endothelial Corneal Dystrophy Program. Available online: https://www.designtx.com/our-programs/#fuchs-endothelial-corneal-dystrophy (accessed on 1 August 2025).
  95. Uehara, H.; Zhang, X.; Pereira, F.; Narendran, S.; Choi, S.; Bhuvanagiri, S.; Liu, J.; Kumar, S.R.; Bohner, A.; Carroll, L.; et al. Start codon disruption with CRISPR/Cas9 prevents murine Fuchs’ endothelial corneal dystrophy. eLife 2021, 10, e55637. [Google Scholar] [CrossRef]
  96. Hu, X.; Zhang, B.; Li, X.; Li, M.; Wang, Y.; Dan, H.; Zhou, J.; Wei, Y.; Ge, K.; Li, P.; et al. The application and progression of CRISPR/Cas9 technology in ophthalmological diseases. Eye 2023, 37, 607–617. [Google Scholar] [CrossRef]
  97. Gowda, D.A.A.; Birappa, G.; Rajkumar, S.; Ajaykumar, C.B.; Srikanth, B.; Kim, S.L.; Singh, V.; Jayachandran, A.; Lee, J.; Ramakrishna, S. Recent progress in CRISPR/Cas9 system for eye disorders. Prog. Mol. Biol. Transl. Sci. 2025, 210, 21–46. [Google Scholar] [CrossRef] [PubMed]
  98. Rim, M.H.; Dean, C.; Aliaj, E.; Karas, B.L.; Barada, F.; Levitsky, A.M. Recent and anticipated novel drug approvals (3Q 2024 through 2Q 2025). Am. J. Health Syst. Pharm. 2024, 81, 1103–1108. [Google Scholar] [CrossRef]
  99. Mohan, R.R.; Martin, L.M.; Sinha, N.R. Novel insights into gene therapy in the cornea. Exp. Eye Res. 2021, 202, 108361. [Google Scholar] [CrossRef] [PubMed]
  100. Hudde, T.; Rayner, S.A.; De Alwis, M.; Thrasher, A.J.; Smith, J.; Coffin, R.S.; George, A.J.; Larkin, D.F. Adeno-associated and herpes simplex viruses as vectors for gene transfer to the corneal endothelium. Cornea 2000, 19, 369–373. [Google Scholar] [CrossRef]
  101. Klebe, S.; Sykes, P.J.; Coster, D.J.; Bloom, D.C.; Williams, K.A. Gene transfer to ovine corneal endothelium. Clin. Exp. Ophthalmol. 2001, 29, 316–322. [Google Scholar] [CrossRef]
Figure 1. Photograph of cornea in a patient with Fuchs endothelial corneal dystrophy; grade 3; endothelial corneal changes are present in the central cornea (a). Specular microscopy of corneal endothelium in the (b) center showing the image of changed and dysfunctional endothelial cells, and the (c) periphery with normal endothelium. The patient with normal periphery can be treated with Descemet stripping only (DSO) procedure. Source: unpublished collection from archive of Eye Hospital University Clinical Center Ljubljana.
Figure 1. Photograph of cornea in a patient with Fuchs endothelial corneal dystrophy; grade 3; endothelial corneal changes are present in the central cornea (a). Specular microscopy of corneal endothelium in the (b) center showing the image of changed and dysfunctional endothelial cells, and the (c) periphery with normal endothelium. The patient with normal periphery can be treated with Descemet stripping only (DSO) procedure. Source: unpublished collection from archive of Eye Hospital University Clinical Center Ljubljana.
Genes 16 01222 g001
Figure 2. Surgical procedures for FECD—(a,b) Descemet membrane endothelial keratoplasty (DMEK) intraoperative view: (a) 8 mm descemetorhexis on recipient eye, (b) endothelial graft was inserted into the anterior chamber, (c) central diseased endothelium was removed during Descemetorhexis only (DSO)—retroillumination view of the transparent cornea after healing due to peripheral endothelial cell movement into the central part of the cornea. Source: archive, unpublished, collection from archive of Eye Hospital University Clinical Center Ljubljana.
Figure 2. Surgical procedures for FECD—(a,b) Descemet membrane endothelial keratoplasty (DMEK) intraoperative view: (a) 8 mm descemetorhexis on recipient eye, (b) endothelial graft was inserted into the anterior chamber, (c) central diseased endothelium was removed during Descemetorhexis only (DSO)—retroillumination view of the transparent cornea after healing due to peripheral endothelial cell movement into the central part of the cornea. Source: archive, unpublished, collection from archive of Eye Hospital University Clinical Center Ljubljana.
Genes 16 01222 g002
Table 1. Different genes of early and late-onset FECD.
Table 1. Different genes of early and late-onset FECD.
FECDGeneMutation—Significance or ACMG Classification *: 5–1References
Early-onsetCOL8A2Q455K-5
L450W-5
R155Q-1
R304Q-3
R434H-3
[13,14,15,16]
Late-onsetTCF4CTG repeat within third intron—pathogenic above >40–50 repeats
rs613872-3
rsl 7595731-3
rs9954153-3
rs2286812-3
rs784257-3
[17,18,19,20]
DMPKCTG repeat within the 3′ UTR—not common[21,22]
SLC4A11E399K-4
G709E-4
T754M-4
c.99–100delTC-4
E167D-3
R282P-3
Y526C-3
V575M-3
G583D-3
G742R-3
G834S-3
W240S-3
V507I-3
T434I-3
[23,24,25]
TCF8/ZEB1
(chr 10p11.22)
N78T-4
Q810P-4
Q840P-4
A905G-4
P649A-4
N696S-3
rs77516068-3
rsl49166539-3
IVS2+276-3
S234S-3
E733K-3
A818V-3
L947Stop-3
[24,26,27]
L0XHD1R547C-4
rs450997-3
[20,28]
AGBL1R1028Stop-3
c.2969G>C-3
[29]
KANK4
(chr 1q25.3)
rs79742895-3[20]
LAMC1rs3768617-3[20,30]
LINC00970/ATP1B1rsl022114-3[20]
DNA Repair enzymes
RAD51
FEN1
XRCC1
NEIL1
LIG3
c.−61G>T/rs1801321)-3
c.−98G>C/rs1801320-3
rs4246215-4
c.H96A>G-4
g.46438521G>C-3rslO52536-3
rs3135967-3
[31,32,33,34]
Extracellular Matrix
TGFBI
CLU
COL8A2
one haplotype-3
rs 17466684-4
R304Q-3
R434H-3
[34,35,36]
Mitochondrial
ND3
TSPOAP1
A10398G-1
Variants-3,4
[18,37]
Other
PITX2
PTPRG
FASLG
KCNJ13
g.20913G>T-3
rs7640737-3
rs 10490775
c.−671A>G-3
R162W-4
[19,36,38,39]
* ACMG American College of Medical Genetics and genomics classification for each variant (Class 5: pathogenic; Class 4: likely pathogenic; Class 3: variant of uncertain significance [VUS]; or Class 1: benign polymorphism).
Table 2. Chromosome loci of FECD.
Table 2. Chromosome loci of FECD.
FECDLocus on ChromosomeReference
Early-onset1p34.3-p32 (FECD1)[45]
Late-onset13pter-q12.13 (FECD2)
18q21.2-q21.3 (FECD3)
20p13-p12 (FECD4)
5q33.1-q35.2 (FECD5)
10p11.2 (FECD6)
9p24.1-p22.1 (FECD7)
15q25 (FECD8)
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

Stunf Pukl, S. Genetic Therapy of Fuchs Endothelial Corneal Dystrophy: Where Are We? A Review. Genes 2025, 16, 1222. https://doi.org/10.3390/genes16101222

AMA Style

Stunf Pukl S. Genetic Therapy of Fuchs Endothelial Corneal Dystrophy: Where Are We? A Review. Genes. 2025; 16(10):1222. https://doi.org/10.3390/genes16101222

Chicago/Turabian Style

Stunf Pukl, Spela. 2025. "Genetic Therapy of Fuchs Endothelial Corneal Dystrophy: Where Are We? A Review" Genes 16, no. 10: 1222. https://doi.org/10.3390/genes16101222

APA Style

Stunf Pukl, S. (2025). Genetic Therapy of Fuchs Endothelial Corneal Dystrophy: Where Are We? A Review. Genes, 16(10), 1222. https://doi.org/10.3390/genes16101222

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