Gene Therapy Approaches to Treat Retinitis Pigmentosa: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Ophthalmology
Contributor: , , , , , ,

Retinitis pigmentosa (RP) is a group of hereditary diseases that result in the progressive degeneration of the retina's photoreceptor cells, primarily starting with the rods. This gradual loss of vision is the most common form of inherited retinal dystrophy, and it imposes a significant burden on both individuals and society. RP is a leading cause of visual disability and blindness in people under 60, affecting more than 1.5 million individuals worldwide. The most common symptoms of RP include nyctalopia and gradual peripheral vision loss, which may ultimately lead to complete blindness.

  • retinitis pigmentosa
  • ER stress
  • retinal degeneration
  • photoreceptor cell death
  • therapeutictarget
  • neuroprotection
  • optogenetics
  • gene therapy
  • stem cell therapy
  • preclinical studies

Gene Therapy

Overview of Gene Therapy Methods

Gene therapy has made significant advancements in the treatment of inherited retinal diseases, particularly in the field of RP [34]. Gene therapy approaches for RP are generally based on two strategies, which are influenced by the disease's inheritance pattern. In recessive RP, where there is a loss of function of the affected protein, the gene complementation approach is applied. On the other hand, gene therapy approaches for dominant RP typically involve gene suppression, with or without gene complementation.

To achieve RP management's therapeutic goal, various gene therapy techniques have been developed using viral or non-viral vectors. This section provides an overview of the primary gene therapy methods currently being studied for the development of therapeutic approaches for RP management.

 

Viral Vectors

Gene therapy using viral vectors for the treatment of retinitis pigmentosa (RP) has been extensively researched, with several vectors reaching clinical studies [35]. Three primary vector strategies based on viruses have been studied: adenoviruses (Ad), adeno-associated viruses (AAV), and lentiviruses (LV) [36]. Recently, AAV has gained focus due to its potential. AAV is a nonenveloped icosahedral virus that was described in 1996 and is relatively small in size (25 nm) [37] (pp. 1–23). The small size of AAV enables the efficient targeting of retinal layers, which makes it a favored option in gene therapy for the treatment of inherited retinopathies (IR). Additionally, AAV exhibits low immunogenicity, allowing a prolonged expression of the target gene after single-dose treatment and safety in second-dose administration in the subretinal space [38]. Moreover, AAV is non-pathogenic and has not yet been linked to human diseases, although it can be present [36]. On the other hand, It is important to note that pre-immunity towards Ad-based vectors exists due to their high seroprevalence, which can compromise Ad-based gene therapies [39].

AAV vectors have a simple genome that lacks auto-replication and expression [36]. AAV-mediated DNA editing is performed through the clathrin-mediated endocytosis pathway, and once in the nucleus, the DNA strand undergoes second-strand synthesis followed by gene transcription. The AAV genome can integrate into the human genome on chromosome 19 at the AAVS1 position [40]. AAV is highly transduction efficient and provides stable expression, making it an attractive option for gene therapy [41]. However, the use of single AAV vectors has limitations in the context of RP, including a cloning capacity that is limited to non-large genomes (4.7 kb), a relatively slow onset of transgene expression due to the need for second-strand synthesis, and highly invasive procedures requiring subretinal injections [38]. Researchers have developed novel approaches to address these challenges for treating IRD, such as splitting the coding sequence into separate AAV vectors to reconstitute full-length transgene protein expression in transduced cells, increasing the cloning capacity of AAV up to 14 kb using triple vectors [46], and using dual vectors to treat Stargardt disease in Abca4−/− mice, which improved disease phenotype [47]. However, using multiple AAV vectors for treating monogenic diseases is challenging due to the requirement of laborious post-translational modifications such as recombination and splicing [42]. Nonetheless, novel AAV capsids have been developed that allow for intravitreal gene therapy, which could alleviate the burden associated with intraretinal injections [48–50].

 

CRISPR-Cas Gene Editing System

The novel CRISPR-Cas9 gene editing system is a widely used tool for gene expression and suppression [51,52]. In retinal diseases, it has successfully silenced dominant mutations via the NHEJ pathway [53,54]. However, clinical application faces a major challenge due to lower performance rates, with success highly dependent on sgRNA design, cell target type, delivery method, and host factors. Despite this, even a small percentage of gene modification through CRISPR-Cas9 can improve phenotype and hold significant promise for IRD.

Recent research has shown a growing interest in RNA-guided gene editing methods [55]. The Cas13 tool is an RNAtargeting endonuclease that can implement gene modifications during transcriptional activity, unlike the DNA-editing CRISPR-Cas9 system [56,57]. Cas13-based RNA editing has successfully shown the restoration of protein product functions in monogenic disease therapy [55]. However, the potential for non-specific collateral RNA cleavage is a major drawback associated with the Cas13 tool [58–60], along with a recent finding that it could be neurotoxic, impeding neuronal development in vitro [61]. While CRISPR-Cas13 methods represent a novel approach for the treatment of retinal diseases, further studies are required to assess their potential and safety [62].

 

RNA Replacement

RNA replacement therapies involve the depletion of endogenous mutant RNAs. Noncoding RNAs (ncRNAs) play a crucial role in regulating gene expression and can be categorized into microRNA (miRNA), small interfering RNA (siRNA), and short hairpin RNA (shRNA) based on their regulatory elements [63]. MiRNA represses mRNA translation and destabilizes mRNA by binding to its target sites [63]. RNA interference (RNAi) regulates protein-coding gene expression [64], and mirtrons, a subtype of miRNA, can be used in gene therapy for retinitis pigmentosa (RP) treatment [65]. SiRNA binds with high specificity, and shRNA regulates DNA delivery instead of RNA effector molecule regulation [63]. Antisense oligonucleotides (ASO) are synthetic single-stranded RNAs that promote RNA cleavage and have therapeutic effects in inherited retinal diseases [63]. SiRNA is vulnerable to degradation by serological enzymes, but shRNA is more potent and has a sustained effect [67].

 

Targeting Retinitis Pigmentosa Pathogenesis

In preclinical studies, various genetic mutations have been implicated in the development of RP, affecting apoptotic and non-apoptotic cell death pathways, inflammatory responses, and metabolic pathways (Table 1) [68]. Unfortunately, there is no curative treatment available for RP, and current approaches focus on developing targeted gene therapies for cell rescue, as well as molecular targets to facilitate cell replacement and maintenance of artificial retina [69].

Management of RP has been the subject of numerous studies, which have reviewed current treatment options, focusing on clinical trials or FDA-approved treatments [68,69]. Despite the genetic heterogeneity of RP, new molecular findings from preclinical studies may offer potential targets for drug development. In this section, we will review these findings, as well as advances in mutation-specific gene therapy.

 

Physiological Signaling Pathways of the Retina

Retinal dystrophy is influenced by multiple genes. Most genetic mutations associated with RP lead to the dysfunction of photoreceptors or RPE. Among these, various mutations have been identified as playing a key role in RP development [89-91], with some mutations currently in clinical trial phases for drug development. Most genetic mutations associated with RP lead to the dysfunction of photoreceptors or RPE [92].

 

Phototransduction

Photoreceptors, including rod and cone cells, are specialized neurons responsible for the initial step of vision: phototransduction. Rod cells are responsible for dim light vision, whereas cone cells allow for color vision. Each photoreceptor is composed of five distinct regions: the outer segment (OS), connecting cilium (CC), inner segment (IS), nuclear region, and synaptic region. The OS of rod cells contains stacked disks, which significantly increase the membrane surface area and promote a high density of visual pigments such as rhodopsin. Rhodopsin is a G-protein-coupled receptor synthesized in the endoplasmic reticulum that allows phototransduction once activated by light. Activation of rhodopsin initiates GTP-mediated signaling, leading to the hydrolysis and reduction of cGMP in the cytosol of photoreceptors. The lower levels of cGMP cause the closure of CNG ion channels, which inhibits calcium and sodium influx, resulting in the hyperpolarization of rod photoreceptors and the subsequent inhibition of glutamate release at the synapse.

 

Photoreceptor Cell Death

Photoreceptor cell death is a critical aspect of the pathogenesis of inherited retinal diseases such as RP, leading to visual loss. Apoptosis pathway activation, which involves three caspase-dependent pathways, has been identified as the most studied cell death mechanism in retinal degeneration. The intrinsic apoptotic pathway is mediated by pro-apoptotic Bcl-2 family proteins, which induce pore formation in the mitochondrial membrane, leading to cytochrome c release. Cytochrome c forms a complex with APAF1 and procaspase9, called the apoptosome, leading to caspase-9 activation, which ultimately induces irreversible DNA fragmentation via activation of caspase-3. However, inflammasome activation by NOD-like receptors (NLRs) was shown to significantly contribute to photoreceptor cell death through immunity activation and executioner caspase cleavage, demonstrating the importance of the immune system in the pathogenesis of retinal degeneration.

Research has demonstrated the significance of unfolded protein response (UPR) signaling through the ER stress pathway in retinal degeneration. When there is an accumulation of unfolded or misfolded proteins in the cytosol, the UPR pathway is activated, mediated by three main pathways involving ER transmembrane proteins: IRE1α, eIF2 kinase-PERK, and ATF6 (Figure 3) [104-107]. Uncontrolled activation of the ER stress pathway can lead to cell apoptosis [102], while in normal conditions, inhibitory chaperone molecules such as glucose regulatory protein 78 (GRP78/BiP) maintain ER transmembrane protein homeostasis [108].

RP pathogenesis involves various regulated necrosis pathways, including necroptosis, ferroptosis, and pyroptosis. Necroptosis is activated by TNFα or reactive oxygen species (ROS) stimulation, and through ischemia-induced cell death, leading to RIPK1-RIPK3-MLKL activation and membrane alteration, and eventually inducing inflammatory response [109]. Ferroptosis involves lipid peroxidation and intracellular iron accumulation, leading to ROS level elevation. Pyroptosis leads to caspase activation and causes cell membrane lysis and inflammatory response, similar to the apoptosis pathway [99].

 

Autosomal Dominant-Linked Mutations

Autosomal dominant disorders are highly studied due to their severity as a single allele mutation can induce a change in phenotype. Mutations associated with adRP make up 50-75% of cases, prompting the need for new therapeutic approaches targeting these genes [110]. The pathogenesis of adRP involves numerous genes, with RHO being the most studied. In this section, we will review new advances in other molecular targets.

 

Rhodopsin-Induced Autosomal Dominant Retinitis Pigmentosa

RP is often caused by mutations in the RHO gene, affecting about 25% of patients [111]. To find a treatment for rhodopsin-associated RP, researchers have focused on understanding the pathogenesis and biomolecular changes caused by RHO mutations using cell lines and animal models, primarily rodents. With valid and robust animal models in place, scientific interest has shifted to developing gene therapy vectors.

In preclinical phases, gene therapy vectors for RP primarily target adRP, utilizing advances in mammalian models with rhodopsin mutations. However, recent in vivo models have revealed important insights into the pathogenesis of RP, including the inhibition of Müller glial cell proliferation in Xenopus laevis tadpoles with a CRISPRcas9-induced rho mutation [70], and the rapid destruction of the outer nuclear layer due to rhodopsin accumulation in a P347L transgenic rat strain [72]. In addition, the ER stress pathway was activated in P347L transgenic rats, as evidenced by significant upregulation of CHOP and BiP mRNA expression [72].

The P347S mutation in rhodopsin has been shown to impair transport to the photoreceptor outer segment in transgenic mice and pigs. Strategies to suppress the expression of the mutant transcript and replace it with a wild-type copy have shown promise in restoring retinal morphology and function. Specifically, transcriptional repression with the KRAB domain and zinc-finger DNA-binding domains has successfully decreased transcript levels of the mutant while preserving endogenous murine RHO gene expression in transgenic mice. RNA replacement therapies using shRNA delivery have also successfully replaced mutant rhodopsin RNA transcripts, leading to increased rhodopsin levels and improved ERG responses. The CRISPR-cas9 system has demonstrated success in allele-specific disruption of the RHOS334 gene in S334ter-3 rats, leading to increased retinal preservation and visual acuity. Additionally, NRL and NR2E3 gene disruptions with dual subretinal injection of AAV-cas9 vector have shown promising results in RHO-P347S/Rd10 and Rd10/Rd1 mice, respectively. Dysregulation of miRNA levels has also been implicated in RP pathogenesis, and AAV-mediated delivery of pre-miR-204 and AAV-mirtron vector in RHO-P247S transgenic mice and Nrl.GFP/+, RhoP23H/+ mice, respectively, have shown success in delaying degeneration and attenuating microglia activation and photoreceptor cell death. Lastly, inhibition of ceramide de novo synthesis with Myriocin has been shown to support cell survival and preserve ERG responses in Tvrm4 mice carrying a Rhodopsin gene mutation.

The T17M rhodopsin mutation activates the unfolded protein response (UPR) pathway, leading to ER stress and photoreceptor cell death [71,121]. In transgenic C57BL6 mice, T17M rhodopsin expression upregulated IL-1β, IL-6, and NFκB and the IB1A microglia markers, indicating UPR pathway activation and loss of photoreceptor function and retinal structure [71]. Knockdown of ATF4, a UPR pathway mediator, delayed retinal degeneration in T17M-induced RP mice, with double knockdowns having a more significant therapeutic effect, likely due to the attenuation of pEIF2α, ATF6, and CHOP expression [122].

 

Non-Rhodopsin-Related Autosomal Dominant Mutations

Mutations in pre-mRNA processing factors (PRPFs) are linked to up to 20% of adRP cases [123]. PRPFs are involved in pre-mRNA splicing, ciliogenesis, and DNA damage repair pathways. Prpf31 gene augmentation using AAV-associated CRISPR-cas9 vector gene augmentation restored the integrity of retinal structure in mice [79]. Similarly, Rpgr gene-editing using sgRNA (cas9) and AAV vectors induced photoreceptor preservation in Rpgr−/yCas9+/WT mice [83].

Mutations in the RP1 gene, which is involved in adRP, disrupt protein transport within photoreceptors, cilial structure maintenance, and disc membrane stability, leading to photoreceptor cell death. Recently, dual Cas9/sgRNA technology has been shown to successfully reduce RP1 expression in edited Hap1-EF1a-RP1 cells, which could lead to further clinical trials.

 

Autosomal Recessive-Linked Mutations

Rhodopsin-Induced Autosomal Recessive Retinitis Pigmentosa

RHO mutations are associated with both adRP and arRP, with only a few linked to the latter. The five known variants for rhodopsin-associated arRP are E150K, W161ter, E249ter, and M2531. The missense mutation E150K impairs rhodopsin stabilization, leading to retinal degeneration due to a disorganized photoreceptor disc structure and impaired phagocytosis. Homozygous E150K mice exhibit higher retinal immune activation. Two nonsense mutations (E249ter and W161ter) were also found to be involved in arRP, with lower levels of rhodopsin mRNA detected in human cells transfected with the rhodopsin nonsense mutants. The M2531 rhodopsin mutants were shown to be linked to the asymptomatic arRP form in heterozygous patients from consanguineous Turkish descents. Further studies are required to better understand the functions and impacts of autosomal recessive mutations in RP development.

 

Non-Rhodopsin-Related Autosomal Recessive Mutations

CNGB1 sequence variants are associated with arRP, which accounts for almost 4% of arRP cases [132]. A recent literature review has linked a total of 62 genetic variants to inherited retinal diseases [132]. Restoring CNGB1 expression in Cngb1−/− mice using an AAV vector has been shown to improve vision. CNGB1-expressing mice had better performance in a rod-dependent vision-guided behavior test [84], and retinal degeneration was significantly delayed, with morphology preserved [84].

TULP1 mutation causes early-onset retinal degeneration, but its pathogenesis remains largely unknown [133]. To investigate the pathogenic mechanism of various retinal diseases, the zebrafish model has gained popularity in recent years [134]. Zebrafish models were generated with single-knockout (Tulp1+/−) and double-knockout for Tulp1, which significantly altered ciliogenesis by downregulating tektin2, a microtubular component [85]. AAV-mediated TULP1 gene expression editing successfully restored TULP1 mRNA and protein levels [86]. However, in Tulp1−/− mice, the gene editing process was not sufficient to improve the thickness of the outer nuclear layer [86].

Generating new animal models is a crucial step in developing gene therapy treatments. Beryozkin and colleagues have recently generated a knockout mouse model for Fam161a deficiency, which displays retinal degeneration and enhanced molecular generative markers such as microglia [87]. Homozygous Fam161a p.Arg512∗ have also been shown to have altered visual acuity due to complete loss of the outer nuclear layer and photoreceptor cell death [88].

The retinitis pigmentosa GTPase regulator (RPGR) gene is the most commonly studied gene and has been shown to cause recessive RP [135]. Promising clinical trials are being conducted for therapeutic approaches using AAV vectors, while preclinical phase studies are ongoing to develop other gene therapy methods. In the rd9 mice model, AAV-guided CRISPR-cas9 vector injection has been used to restore the reading frame of RPGRORF15 [82].

 

Identification of Gene Targets Involved in Retinitis Pigmentosa for Novel Gene Therapy Treatments

The identification of novel genes involved in RP pathogenesis is essential for future gene therapy development. Gene sequencing studies of large patient cohorts have identified potential gene targets. Genome sequencing studies in the past five years have highlighted the involvement of multiple genes in RP pathogenesis [136-138]. For instance, SLC66A1 and SLC39A12 were identified as novel targets in inherited retinal disease cases by analyzing sequence variants of SLC genes in Israeli and Palestinian cohorts [139]. SLC7A14 was also shown to be involved in retinal development as SLC7A14 knockout mice exhibited retinal degeneration and altered visual function [140]. In addition, rare heterozygous variants of AGBL5, p.Arg281Cys and p.Arg487*, were potentially implicated in RP [141]. However, further research is necessary to elucidate the pathophysiology of these novel genes in RP. The Retinal Information Network’s website (https://web.sph.uth.edu/RetNet/ (accessed on 20 January 2023)) provides an updated list of genes and mapped loci causing retinal diseases.

 

[1][2][3][4][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79][80][81][82][83][84][85][86][87][88][89][90][91][92][93][94][95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117][118][119][120][121][122][123][124][125][126][127][128][129][130][131][132][133][134][135][136][137][138][139][140][141][142][143][144][145][146][147][148][149][150][151][152][153][154][155][156][157][158][159][160][161][162][163][164][165][166][167][168][169][170][171][172][173][174][175][176][177][178][179][180][181][182][183][184][185][186][187][188][189][190][191][192][193][194][195][196][197][198][199][200][201][202][203][204][205][206][207][208][209][210][211][212][213][214][215][216][217][218][219][220][221][222][223][224][225][226][227][228][229][230][231][232][233][234][235][236][237][238][239][240]

This entry is adapted from the peer-reviewed paper 10.3390/pharmaceutics15020685

References

  1. 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]
  2. Retinitis Pigmentosa: Study Guide. Available online: https://www.aao.org/Assets/bcc226cb-7ac7-443c-897e-9b20afdfacfb/637153836910470000/r38u-pdf?inline=1&fbclid=IwAR2oP_lD1pWSAROpIfa9poqhZzeSf29_PvZztBZMSQfWKNqTbv1b6t0BXJU (accessed on 15 December 2022).
  3. O’Neal, T.B.; Luther, E.E. Retinitis Pigmentosa. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  4. Hartong, D.T.; Berson, E.L.; Dryja, T.P. Retinitis Pigmentosa. Lancet 2006, 368, 1795–1809. [Google Scholar] [CrossRef] [PubMed]
  5. Kimberling, W.J.; Hildebrand, M.S.; Shearer, A.E.; Jensen, M.L.; Halder, J.A.; Trzupek, K.; Cohn, E.S.; Weleber, R.G.; Stone, E.M.; Smith, R.J.H. Frequency of Usher Syndrome in Two Pediatric Populations: Implications for Genetic Screening of Deaf and Hard of Hearing Children. Genet. Med. Off. J. Am. Coll. Med. Genet. 2010, 12, 512–516. [Google Scholar] [CrossRef] [PubMed][Green Version]
  6. Weihbrecht, K. Bardet-Biedl Syndrome. In Genetics and Genomics of Eye Disease; Elsevier: Amsterdam, The Netherlands, 2020; pp. 117–136. ISBN 978-0-12-816222-4. [Google Scholar]
  7. Von Sallmann, L.; Gelderman, A.H.; Laster, L. Ocular Histopathologic Changes in a Case of A-Beta-Lipoproteinemia (Bassen-Kornzweig Syndrome). Doc. Ophthalmol. Adv. Ophthalmol. 1969, 26, 451–460. [Google Scholar] [CrossRef] [PubMed]
  8. Grant, C.A.; Berson, E.L. Treatable Forms of Retinitis Pigmentosa Associated with Systemic Neurological Disorders. Int. Ophthalmol. Clin. 2001, 41, 103–110. [Google Scholar] [CrossRef]
  9. Gibberd, F.B.; Billimoria, J.D.; Goldman, J.M.; Clemens, M.E.; Evans, R.; Whitelaw, M.N.; Retsas, S.; Sherratt, R.M. Heredopathia Atactica Polyneuritiformis: Refsum’s Disease. Acta Neurol. Scand. 1985, 72, 1–17. [Google Scholar] [CrossRef]
  10. Berson, E.L.; Sandberg, M.A.; Rosner, B.; Birch, D.G.; Hanson, A.H. Natural Course of Retinitis Pigmentosa over a Three-Year Interval. Am. J. Ophthalmol. 1985, 99, 240–251. [Google Scholar] [CrossRef]
  11. Wang, A.L.; Knight, D.K.; Vu, T.-T.T.; Mehta, M.C. Retinitis Pigmentosa: Review of Current Treatment. Int. Ophthalmol. Clin. 2019, 59, 263–280. [Google Scholar] [CrossRef]
  12. Rayapudi, S.; Schwartz, S.G.; Wang, X.; Chavis, P. Vitamin A and Fish Oils for Retinitis Pigmentosa. Cochrane Libr. Cochrane Rev. 2013, 2013, CD008428. [Google Scholar] [CrossRef][Green Version]
  13. Berson, E.L. Retinitis Pigmentosa. The Friedenwald Lecture. Investig. Ophthalmol. Vis. Sci. 1993, 34, 1659–1676. [Google Scholar]
  14. Berson, E.L.; Rosner, B.; Sandberg, M.A.; Weigel-DiFranco, C.; Moser, A.; Brockhurst, R.J.; Hayes, K.C.; Johnson, C.A.; Anderson, E.J.; Gaudio, A.R.; et al. Further Evaluation of Docosahexaenoic Acid in Patients with Retinitis Pigmentosa Receiving Vitamin A Treatment: Subgroup Analyses. Arch. Ophthalmol. 2004, 122, 1306–1314. [Google Scholar] [CrossRef] [PubMed]
  15. Berson, E.L.; Rosner, B.; Sandberg, M.A.; Weigel-DiFranco, C.; Brockhurst, R.J.; Hayes, K.C.; Johnson, E.J.; Anderson, E.J.; Johnson, C.A.; Gaudio, A.R.; et al. Clinical Trial of Lutein in Patients with Retinitis Pigmentosa Receiving Vitamin A. Arch. Ophthalmol. 2010, 128, 403–411. [Google Scholar] [CrossRef] [PubMed][Green Version]
  16. Russell, R.M. The Vitamin A Spectrum: From Deficiency to Toxicity. Am. J. Clin. Nutr. 2000, 71, 878–884. [Google Scholar] [CrossRef] [PubMed][Green Version]
  17. Pierce, E.A.; Bujakowska, K.M.; Place, E.; Navarro-Gomez, D.; Maher, M.; Weigel-DFranco, C.; Comander, J. The Effect Of Vitamin A On Progression Of Retinitis Pigmentosa Is Not Determined By The Underlying Genetic Cause Of Disease. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2011. [Google Scholar]
  18. Hajali, M.; Fishman, G.A.; Anderson, R.J. The Prevalence of Cystoid Macular Oedema in Retinitis Pigmentosa Patients Determined by Optical Coherence Tomography. Br. J. Ophthalmol. 2008, 92, 1065–1068. [Google Scholar] [CrossRef] [PubMed]
  19. Ozdemir, H.; Karacorlu, M.; Karacorlu, S. Intravitreal Triamcinolone Acetonide for Treatment of Cystoid Macular Oedema in Patients with Retinitis Pigmentosa. Acta Ophthalmol. Scand. 2005, 83, 248–251. [Google Scholar] [CrossRef]
  20. Bakthavatchalam, M.; Lai, F.H.P.; Rong, S.S.; Ng, D.S.; Brelen, M.E. Treatment of Cystoid Macular Edema Secondary to Retinitis Pigmentosa: A Systematic Review. Surv. Ophthalmol. 2018, 63, 329–339. [Google Scholar] [CrossRef]
  21. Gallenga, C.E.; Lonardi, M.; Pacetti, S.; Violanti, S.S.; Tassinari, P.; Di Virgilio, F.; Tognon, M.; Perri, P. Molecular Mechanisms Related to Oxidative Stress in Retinitis Pigmentosa. Antioxidants 2021, 10, 848. [Google Scholar] [CrossRef]
  22. Naash, M.L.; Peachey, N.S.; Li, Z.Y.; Gryczan, C.C.; Goto, Y.; Blanks, J.; Milam, A.H.; Ripps, H. Light-Induced Acceleration of Photoreceptor Degeneration in Transgenic Mice Expressing Mutant Rhodopsin. Investig. Ophthalmol. Vis. Sci. 1996, 37, 775–782. [Google Scholar]
  23. Kutsyr, O.; Sánchez-Sáez, X.; Martínez-Gil, N.; de Juan, E.; Lax, P.; Maneu, V.; Cuenca, N. Gradual Increase in Environmental Light Intensity Induces Oxidative Stress and Inflammation and Accelerates Retinal Neurodegeneration. Investig. Ophthalmol. Vis. Sci. 2020, 61, 1. [Google Scholar] [CrossRef]
  24. Berson, E.L.; Rabin, A.R.; Mehaffey, L. Advances in Night Vision Technology. A Pocketscope for Patients with Retinitis Pigmentosa. Arch. Ophthalmol. 1973, 90, 427–431. [Google Scholar] [CrossRef] [PubMed]
  25. Hartong, D.T.; Jorritsma, F.F.; Neve, J.J.; Melis-Dankers, B.J.M.; Kooijman, A.C. Improved Mobility and Independence of Night-Blind People Using Night-Vision Goggles. Investig. Ophthalmol. Vis. Sci. 2004, 45, 1725–1731. [Google Scholar] [CrossRef] [PubMed][Green Version]
  26. Mancil, R.M.; Mancil, G.L.; King, E.; Legault, C.; Munday, J.; Alfieri, S.; Nowakowski, R.; Blasch, B.B. Improving Nighttime Mobility in Persons with Night Blindness Caused by Retinitis Pigmentosa: A Comparison of Two Low-Vision Mobility Devices. J. Rehabil. Res. Dev. 2005, 42, 471–486. [Google Scholar] [CrossRef] [PubMed][Green Version]
  27. Ikeda, Y.; Suzuki, E.; Kuramata, T.; Kozaki, T.; Koyama, T.; Kato, Y.; Murakami, Y.; Enaida, H.; Ishibashi, T. Development and Evaluation of a Visual Aid Using See-through Display for Patients with Retinitis Pigmentosa. Jpn. J. Ophthalmol. 2015, 59, 43–47. [Google Scholar] [CrossRef]
  28. Humayun, M.S.; Dorn, J.D.; da Cruz, L.; Dagnelie, G.; Sahel, J.-A.; Stanga, P.E.; Cideciyan, A.V.; Duncan, J.L.; Eliott, D.; Filley, E.; et al. Interim Results from the International Trial of Second Sight’s Visual Prosthesis. Ophthalmology 2012, 119, 779–788. [Google Scholar] [CrossRef][Green Version]
  29. da Cruz, L.; Dorn, J.D.; Humayun, M.S.; Dagnelie, G.; Handa, J.; Barale, P.-O.; Sahel, J.-A.; Stanga, P.E.; Hafezi, F.; Safran, A.B.; et al. Five-Year Safety and Performance Results from the Argus II Retinal Prosthesis System Clinical Trial. Ophthalmology 2016, 123, 2248–2254. [Google Scholar] [CrossRef][Green Version]
  30. Arevalo, J.F.; Al Rashaed, S.; Alhamad, T.A.; Al Kahtani, E.; Al-Dhibi, H.A.; for the KKESH Collaborative Retina Study Group; Mura, M.; Al Kahtani, E.; Nowilaty, S.; Al Rashaed, S.; et al. Argus II Retinal Prosthesis for Retinitis Pigmentosa in the Middle East: The 2015 Pan-American Association of Ophthalmology Gradle Lecture. Int. J. Retin. Vitr. 2021, 7, 65. [Google Scholar] [CrossRef]
  31. 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. [Google Scholar] [CrossRef]
  32. Maguire, A.M.; Russell, S.; Wellman, J.A.; Chung, D.C.; Yu, Z.-F.; Tillman, A.; Wittes, J.; Pappas, J.; Elci, O.; Marshall, K.A.; et al. Efficacy, Safety, and Durability of Voretigene Neparvovec-Rzyl in RPE65 Mutation-Associated Inherited Retinal Dystrophy: Results of Phase 1 and 3 Trials. Ophthalmology 2019, 126, 1273–1285. [Google Scholar] [CrossRef][Green Version]
  33. Maguire, A.M.; Russell, S.; Chung, D.C.; Yu, Z.-F.; Tillman, A.; Drack, A.V.; Simonelli, F.; Leroy, B.P.; Reape, K.Z.; High, K.A.; et al. Durability of Voretigene Neparvovec for Biallelic RPE65-Mediated Inherited Retinal Disease: Phase 3 Results at 3 and 4 Years. Ophthalmology 2021, 128, 1460–1468. [Google Scholar] [CrossRef]
  34. Dhurandhar, D.; Sahoo, N.; Mariappan, I.; Narayanan, R. Gene Therapy in Retinal Diseases: A Review. Indian J. Ophthalmol. 2021, 69, 2257–2265. [Google Scholar] [CrossRef] [PubMed]
  35. Ong, T.; Pennesi, M.E.; Birch, D.G.; Lam, B.L.; Tsang, S.H. Adeno-Associated Viral Gene Therapy for Inherited Retinal Disease. Pharm. Res. 2019, 36, 34. [Google Scholar] [CrossRef] [PubMed]
  36. Bulcha, J.T.; Wang, Y.; Ma, H.; Tai, P.W.L.; Gao, G. Viral Vector Platforms within the Gene Therapy Landscape. Signal Transduct. Target. Ther. 2021, 6, 53. [Google Scholar] [CrossRef] [PubMed]
  37. Berns, K.I.; Giraud, C. Biology of Adeno-Associated Virus. In Adeno-Associated Virus (AAV) Vectors in Gene Therapy; Berns, K.I., Giraud, C., Eds.; Current Topics in Microbiology and Immunology; Springer: Berlin/Heidelberg, Germany, 1996; Volume 218, pp. 1–23. ISBN 978-3-642-80209-6. [Google Scholar]
  38. Trapani, I.; Puppo, A.; Auricchio, A. Vector Platforms for Gene Therapy of Inherited Retinopathies. Prog. Retin. Eye Res. 2014, 43, 108–128. [Google Scholar] [CrossRef][Green Version]
  39. Chen, H.; Xiang, Z.Q.; Li, Y.; Kurupati, R.K.; Jia, B.; Bian, A.; Zhou, D.M.; Hutnick, N.; Yuan, S.; Gray, C.; et al. Adenovirus-Based Vaccines: Comparison of Vectors from Three Species of Adenoviridae. J. Virol. 2010, 84, 10522–10532. [Google Scholar] [CrossRef][Green Version]
  40. Kotin, R.M.; Menninger, J.C.; Ward, D.C.; Berns, K.I. Mapping and Direct Visualization of a Region-Specific Viral DNA Integration Site on Chromosome 19q13-Qter. Genomics 1991, 10, 831–834. [Google Scholar] [CrossRef]
  41. Qu, Y.; Liu, Y.; Noor, A.; Tran, J.; Li, R. Characteristics and Advantages of Adeno-Associated Virus Vector-Mediated Gene Therapy for Neurodegenerative Diseases. Neural Regen. Res. 2019, 14, 931. [Google Scholar] [CrossRef]
  42. Trapani, I.; Tornabene, P.; Auricchio, A. Large Gene Delivery to the Retina with AAV Vectors: Are We There Yet? Gene Ther. 2021, 28, 220–222. [Google Scholar] [CrossRef] [PubMed]
  43. Patel, A.; Zhao, J.; Duan, D.; Lai, Y. Design of AAV Vectors for Delivery of Large or Multiple Transgenes. In Adeno-Associated Virus Vectors; Castle, M.J., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2019; Volume 1950, pp. 19–33. ISBN 978-1-4939-9138-9. [Google Scholar]
  44. Tornabene, P.; Trapani, I. Can Adeno-Associated Viral Vectors Deliver Effectively Large Genes? Hum. Gene Ther. 2020, 31, 47–56. [Google Scholar] [CrossRef]
  45. Carvalho, L.S.; Turunen, H.T.; Wassmer, S.J.; Luna-Velez, M.V.; Xiao, R.; Bennett, J.; Vandenberghe, L.H. Evaluating Efficiencies of Dual AAV Approaches for Retinal Targeting. Front. Neurosci. 2017, 11, 503. [Google Scholar] [CrossRef]
  46. Maddalena, A.; Tornabene, P.; Tiberi, P.; Minopoli, R.; Manfredi, A.; Mutarelli, M.; Rossi, S.; Simonelli, F.; Naggert, J.K.; Cacchiarelli, D.; et al. Triple Vectors Expand AAV Transfer Capacity in the Retina. Mol. Ther. 2018, 26, 524–541. [Google Scholar] [CrossRef] [PubMed][Green Version]
  47. McClements, M.E.; Barnard, A.R.; Singh, M.S.; Charbel Issa, P.; Jiang, Z.; Radu, R.A.; MacLaren, R.E. An AAV Dual Vector Strategy Ameliorates the Stargardt Phenotype in Adult Abca4−/− Mice. Hum. Gene Ther. 2019, 30, 590–600. [Google Scholar] [CrossRef] [PubMed]
  48. Pavlou, M.; Schön, C.; Occelli, L.M.; Rossi, A.; Meumann, N.; Boyd, R.F.; Bartoe, J.T.; Siedlecki, J.; Gerhardt, M.J.; Babutzka, S.; et al. Novel AAV Capsids for Intravitreal Gene Therapy of Photoreceptor Disorders. EMBO Mol. Med. 2021, 13, e13392. [Google Scholar] [CrossRef]
  49. Kevany, B.; Suh, S.; Lu, J.; Padegimas, L.; Palczewski, K.; Miller, T. Novel AAV Capsids Demonstrate Strong Retinal Expression in Non-Human Primates After Intravitreal Administration. Investig. Ophthalmol. Vis. Sci. 2019, 60, 2900. [Google Scholar]
  50. Katada, Y.; Kobayashi, K.; Tsubota, K.; Kurihara, T. Evaluation of AAV-DJ Vector for Retinal Gene Therapy. PeerJ 2019, 7, e6317. [Google Scholar] [CrossRef]
  51. Leonova, E.I.; Gainetdinov, R.R. CRISPR/Cas9 Technology in Translational Biomedicine|Cell Physiol Biochem. Cell. Physiol. Biochem. 2020, 54, 354–370. [Google Scholar]
  52. Doudna, J.A.; Charpentier, E. The New Frontier of Genome Engineering with CRISPR-Cas9. Science 2014, 346, 1258096. [Google Scholar] [CrossRef]
  53. Peddle, C.F.; MacLaren, R.E. The Application of CRISPR/Cas9 for the Treatment of Retinal Diseases. Yale J. Biol. Med. 2017, 90, 533–541. [Google Scholar]
  54. Ahmad, I. CRISPR/Cas9-A Promising Therapeutic Tool to Cure Blindness: Current Scenario and Future Prospects. Int. J. Mol. Sci. 2022, 23, 11482. [Google Scholar] [CrossRef]
  55. Reshetnikov, V.V.; Chirinskaite, A.V.; Sopova, J.V.; Ivanov, R.A.; Leonova, E.I. Cas-Based Systems for RNA Editing in Gene Therapy of Monogenic Diseases: In Vitro and in Vivo Application and Translational Potential. Front. Cell Dev. Biol. 2022, 10, 903812. [Google Scholar] [CrossRef]
  56. Hu, Y.; Chen, Y.; Xu, J.; Wang, X.; Luo, S.; Mao, B.; Zhou, Q.; Li, W. Metagenomic Discovery of Novel CRISPR-Cas13 Systems. Cell Discov. 2022, 8, 107. [Google Scholar] [CrossRef] [PubMed]
  57. Botto, C.; Dalkara, D.; El-Amraoui, A. Progress in Gene Editing Tools and Their Potential for Correcting Mutations Underlying Hearing and Vision Loss. Front. Genome Ed. 2021, 3, 737632. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, Q.; Liu, X.; Zhou, J.; Yang, C.; Wang, G.; Tan, Y.; Wu, Y.; Zhang, S.; Yi, K.; Kang, C. The CRISPR-Cas13a Gene-Editing System Induces Collateral Cleavage of RNA in Glioma Cells. Adv. Sci. 2019, 6, 1901299. [Google Scholar] [CrossRef] [PubMed][Green Version]
  59. Li, Y.; Xu, J.; Guo, X.; Li, Z.; Cao, L.; Liu, S.; Guo, Y.; Wang, G.; Luo, Y.; Zhang, Z.; et al. The Collateral Activity of RfxCas13d Can Induce Lethality in a RfxCas13d Knock-in Mouse Model. Genome Biol. 2023, 24, 20. [Google Scholar] [CrossRef]
  60. Ai, Y.; Liang, D.; Wilusz, J.E. CRISPR/Cas13 Effectors Have Differing Extents of off-Target Effects That Limit Their Utility in Eukaryotic Cells. Nucleic Acids Res. 2022, 50, e65. [Google Scholar] [CrossRef]
  61. Wu, Q.-W.; Kapfhammer, J.P. The Bacterial Enzyme Cas13 Interferes with Neurite Outgrowth from Cultured Cortical Neurons. Toxins 2021, 13, 262. [Google Scholar] [CrossRef]
  62. Fry, L.E.; McClements, M.E.; MacLaren, R.E. Comparison of CRISPR-Cas13 RNA Editing Tools for Inherited Retinal Disease. Investig. Ophthalmol. Vis. Sci. 2022, 63, 3845. [Google Scholar]
  63. Gemayel, M.C.; Bhatwadekar, A.D.; Ciulla, T. RNA Therapeutics for Retinal Diseases. Expert Opin. Biol. Ther. 2021, 21, 603–613. [Google Scholar] [CrossRef]
  64. Hannon, G.J. RNA Interference. Nature 2002, 418, 244–251. [Google Scholar] [CrossRef]
  65. Orlans, H.O.; McClements, M.E.; Barnard, A.R.; Martinez-Fernandez de la Camara, C.; MacLaren, R.E. Mirtron-Mediated RNA Knockdown/Replacement Therapy for the Treatment of Dominant Retinitis Pigmentosa. Nat. Commun. 2021, 12, 4934. [Google Scholar] [CrossRef]
  66. Bajan, S.; Hutvagner, G. RNA-Based Therapeutics: From Antisense Oligonucleotides to MiRNAs. Cells 2020, 9, 137. [Google Scholar] [CrossRef] [PubMed][Green Version]
  67. Knott, S.R.V.; Maceli, A.; Erard, N.; Chang, K.; Marran, K.; Zhou, X.; Gordon, A.; Demerdash, O.E.; Wagenblast, E.; Kim, S.; et al. A Computational Algorithm to Predict ShRNA Potency. Mol. Cell 2014, 56, 796–807. [Google Scholar] [CrossRef] [PubMed][Green Version]
  68. Liu, W.; Liu, S.; Li, P.; Yao, K. Retinitis Pigmentosa: Progress in Molecular Pathology and Biotherapeutical Strategies. Int. J. Mol. Sci. 2022, 23, 4883. [Google Scholar] [CrossRef] [PubMed]
  69. Cross, N.; van Steen, C.; Zegaoui, Y.; Satherley, A.; Angelillo, L. Current and Future Treatment of Retinitis Pigmentosa. Clin. Ophthalmol. 2022, 16, 2909–2921. [Google Scholar] [CrossRef] [PubMed]
  70. Parain, K.; Lourdel, S.; Donval, A.; Chesneau, A.; Borday, C.; Bronchain, O.; Locker, M.; Perron, M. CRISPR/Cas9-Mediated Models of Retinitis Pigmentosa Reveal Differential Proliferative Response of Müller Cells between Xenopus Laevis and Xenopus Tropicalis. Cells 2022, 11, 807. [Google Scholar] [CrossRef]
  71. Rana, T.; Shinde, V.M.; Starr, C.R.; Kruglov, A.A.; Boitet, E.R.; Kotla, P.; Zolotukhin, S.; Gross, A.K.; Gorbatyuk, M.S. An Activated Unfolded Protein Response Promotes Retinal Degeneration and Triggers an Inflammatory Response in the Mouse Retina. Cell Death Dis. 2014, 5, e1578. [Google Scholar] [CrossRef][Green Version]
  72. Inoue, C.; Takeuchi, T.; Shiota, A.; Kondo, M.; Nshizawa, Y. A Rat Model for Retinitis Pigmentosa with Rapid Retinal Degeneration Enables Drug Evaluation in Vivo. Biol. Proced. Online 2021, 23, 11. [Google Scholar] [CrossRef]
  73. Marrocco, E.; Maritato, R.; Botta, S.; Esposito, M.; Surace, E.M. Challenging Safety and Efficacy of Retinal Gene Therapies by Retinogenesis. Int. J. Mol. Sci. 2021, 22, 5767. [Google Scholar] [CrossRef]
  74. Bakondi, B.; Lv, W.; Lu, B.; Jones, M.K.; Tsai, Y.; Kim, K.J.; Levy, R.; Akhtar, A.A.; Breunig, J.J.; Svendsen, C.N.; et al. In Vivo CRISPR/Cas9 Gene Editing Corrects Retinal Dystrophy in the S334ter-3 Rat Model of Autosomal Dominant Retinitis Pigmentosa. Mol. Ther. 2016, 24, 556–563. [Google Scholar] [CrossRef][Green Version]
  75. Karali, M.; Guadagnino, I.; Marrocco, E.; De Cegli, R.; Carissimo, A.; Pizzo, M.; Casarosa, S.; Conte, I.; Surace, E.M.; Banfi, S. AAV-MiR-204 Protects from Retinal Degeneration by Attenuation of Microglia Activation and Photoreceptor Cell Death. Mol. Ther.-Nucleic Acids 2020, 19, 144–156. [Google Scholar] [CrossRef]
  76. Cuevas, E.; Holder, D.L.; Alshehri, A.H.; Tréguier, J.; Lakowski, J.; Sowden, J.C. NRL−/− Gene Edited Human Embryonic Stem Cells Generate Rod-Deficient Retinal Organoids Enriched in S-Cone-like Photoreceptors. Stem Cells 2021, 39, 414–428. [Google Scholar] [CrossRef] [PubMed]
  77. Yu, W.; Mookherjee, S.; Chaitankar, V.; Hiriyanna, S.; Kim, J.-W.; Brooks, M.; Ataeijannati, Y.; Sun, X.; Dong, L.; Li, T.; et al. Nrl Knockdown by AAV-Delivered CRISPR/Cas9 Prevents Retinal Degeneration in Mice. Nat. Commun. 2017, 8, 14716. [Google Scholar] [CrossRef] [PubMed][Green Version]
  78. Zhu, J.; Ming, C.; Fu, X.; Duan, Y.; Hoang, D.A.; Rutgard, J.; Zhang, R.; Wang, W.; Hou, R.; Zhang, D.; et al. Gene and Mutation Independent Therapy via CRISPR-Cas9 Mediated Cellular Reprogramming in Rod Photoreceptors. Cell Res. 2017, 27, 830–833. [Google Scholar] [CrossRef] [PubMed][Green Version]
  79. Xi, Z.; Vats, A.; Sahel, J.-A.; Chen, Y.; Byrne, L.C. Gene Augmentation Prevents Retinal Degeneration in a CRISPR/Cas9-Based Mouse Model of PRPF31 Retinitis Pigmentosa. Nat. Commun. 2022, 13, 7695. [Google Scholar] [CrossRef] [PubMed]
  80. Collin, C.; Liu, Q. CRISPR/Cas9-Based Genome Editing Approaches for RP1 Associated Autosomal Dominant Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2021, 62, 1486. [Google Scholar]
  81. Roman-Sanchez, R.; Wensel, T.G.; Wilson, J.H. Nonsense Mutations in the Rhodopsin Gene That Give Rise to Mild Phenotypes Trigger MRNA Degradation in Human Cells by Nonsense-Mediated Decay. Exp. Eye Res. 2016, 145, 444–449. [Google Scholar] [CrossRef] [PubMed][Green Version]
  82. Gumerson, J.D.; Alsufyani, A.; Yu, W.; Lei, J.; Sun, X.; Dong, L.; Wu, Z.; Li, T. Restoration of RPGR Expression in Vivo Using CRISPR/Cas9 Gene Editing. Gene Ther. 2022, 29, 81–93. [Google Scholar] [CrossRef]
  83. Hu, S.; Du, J.; Chen, N.; Jia, R.; Zhang, J.; Liu, X.; Yang, L. In Vivo CRISPR/Cas9-Mediated Genome Editing Mitigates Photoreceptor Degeneration in a Mouse Model of X-Linked Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2020, 61, 31. [Google Scholar] [CrossRef][Green Version]
  84. Michalakis, S.; Koch, S.; Sothilingam, V.; Garrido, M.G.; Tanimoto, N.; Schulze, E.; Becirovic, E.; Koch, F.; Seide, C.; Beck, S.C.; et al. Gene Therapy Restores Vision and Delays Degeneration in the CNGB1−/− Mouse Model of Retinitis Pigmentosa. In Retinal Degenerative Diseases; Ash, J.D., Grimm, C., Hollyfield, J.G., Anderson, R.E., LaVail, M.M., Bowes Rickman, C., Eds.; Advances in Experimental Medicine and Biology; Springer: New York, NY, USA, 2014; Volume 801, pp. 733–739. ISBN 978-1-4614-3208-1. [Google Scholar]
  85. Jia, D.; Gao, P.; Lv, Y.; Huang, Y.; Reilly, J.; Sun, K.; Han, Y.; Hu, H.; Chen, X.; Zhang, Z.; et al. Tulp1 Deficiency Causes Early-Onset Retinal Degeneration through Affecting Ciliogenesis and Activating Ferroptosis in Zebrafish. Cell Death Dis. 2022, 13, 962. [Google Scholar] [CrossRef]
  86. Palfi, A.; Yesmambetov, A.; Millington-Ward, S.; Shortall, C.; Humphries, P.; Kenna, P.F.; Chadderton, N.; Farrar, G.J. AAV-Delivered Tulp1 Supplementation Therapy Targeting Photoreceptors Provides Minimal Benefit in Tulp1−/− Retinas. Front. Neurosci. 2020, 14, 891. [Google Scholar] [CrossRef]
  87. Beryozkin, A.; Matsevich, C.; Obolensky, A.; Kostic, C.; Arsenijevic, Y.; Wolfrum, U.; Banin, E.; Sharon, D. A New Mouse Model for Retinal Degeneration Due to Fam161a Deficiency. Sci. Rep. 2021, 11, 2030. [Google Scholar] [CrossRef] [PubMed]
  88. Matsevich, C.; Gopalakrishnan, P.; Obolensky, A.; Banin, E.; Sharon, D.; Beryozkin, A. Retinal Structure and Function in a Knock-in Mouse Model for the FAM161A-p.Arg523∗ Human Nonsense Pathogenic Variant. Ophthalmol. Sci. 2023, 3, 100229. [Google Scholar] [CrossRef] [PubMed]
  89. García Bohórquez, B.; Aller, E.; Rodríguez Muñoz, A.; Jaijo, T.; García García, G.; Millán, J.M. Updating the Genetic Landscape of Inherited Retinal Dystrophies. Front. Cell Dev. Biol. 2021, 9, 645600. [Google Scholar] [CrossRef] [PubMed]
  90. Nash, B.M.; Wright, D.C.; Grigg, J.R.; Bennetts, B.; Jamieson, R.V. Retinal Dystrophies, Genomic Applications in Diagnosis and Prospects for Therapy. Transl. Pediatr. 2015, 4, 139–163. [Google Scholar] [CrossRef]
  91. Perea-Romero, I.; Gordo, G.; Iancu, I.F.; Del Pozo-Valero, M.; Almoguera, B.; Blanco-Kelly, F.; Carreño, E.; Jimenez-Rolando, B.; Lopez-Rodriguez, R.; Lorda-Sanchez, I.; et al. Genetic Landscape of 6089 Inherited Retinal Dystrophies Affected Cases in Spain and Their Therapeutic and Extended Epidemiological Implications. Sci. Rep. 2021, 11, 1526. [Google Scholar] [CrossRef] [PubMed]
  92. Parmeggiani, F.S.; Sorrentino, F.; Ponzin, D.; Barbaro, V.; Ferrari, S.; Di Iorio, E. Retinitis Pigmentosa: Genes and Disease Mechanisms. Curr. Genom. 2011, 12, 238–249. [Google Scholar] [CrossRef]
  93. Palczewski, K. Chemistry and Biology of Vision. J. Biol. Chem. 2012, 287, 1612–1619. [Google Scholar] [CrossRef][Green Version]
  94. Lenahan, C.; Sanghavi, R.; Huang, L.; Zhang, J.H. Rhodopsin: A Potential Biomarker for Neurodegenerative Diseases. Front. Neurosci. 2020, 14, 326. [Google Scholar] [CrossRef][Green Version]
  95. Kandori, H.; Shichida, Y.; Yoshizawa, T. Photoisomerization in Rhodopsin. Biochemistry 2001, 66, 1197–1209. [Google Scholar] [CrossRef]
  96. Palczewski, K. G Protein-Coupled Receptor Rhodopsin. Annu. Rev. Biochem. 2006, 75, 743–767. [Google Scholar] [CrossRef][Green Version]
  97. Kramer, R.H.; Molokanova, E. Modulation of Cyclic-Nucleotide-Gated Channels and Regulation of Vertebrate Phototransduction. J. Exp. Biol. 2001, 204, 2921–2931. [Google Scholar] [CrossRef] [PubMed]
  98. BIO254: Phototransduction—OpenWetWare. Available online: https://openwetware.org/mediawiki/index.php?title=BIO254:Phototransduction&oldid=85424 (accessed on 12 February 2023).
  99. Newton, F.; Megaw, R. Mechanisms of Photoreceptor Death in Retinitis Pigmentosa. Genes 2020, 11, 1120. [Google Scholar] [CrossRef]
  100. Choudhury, S.; Bhootada, Y.; Gorbatyuk, O.; Gorbatyuk, M. Caspase-7 Ablation Modulates UPR, Reprograms TRAF2-JNK Apoptosis and Protects T17M Rhodopsin Mice from Severe Retinal Degeneration. Cell Death Dis. 2013, 4, e528. [Google Scholar] [CrossRef] [PubMed][Green Version]
  101. Comitato, A.; Sanges, D.; Rossi, A.; Humphries, M.M.; Marigo, V. Activation of Bax in Three Models of Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2014, 55, 3555. [Google Scholar] [CrossRef] [PubMed][Green Version]
  102. Kulbay, M.; Paimboeuf, A.; Ozdemir, D.; Bernier, J. Review of Cancer Cell Resistance Mechanisms to Apoptosis and Actual Targeted Therapies. J. Cell. Biochem. 2022, 123, 1736–1761. [Google Scholar] [CrossRef]
  103. Zhao, L.; Hou, C.; Yan, N. Neuroinflammation in Retinitis Pigmentosa: Therapies Targeting the Innate Immune System. Front. Immunol. 2022, 13, 1059947. [Google Scholar] [CrossRef] [PubMed]
  104. Read, A.; Schröder, M. The Unfolded Protein Response: An Overview. Biology 2021, 10, 384. [Google Scholar] [CrossRef]
  105. Junjappa, R.P.; Patil, P.; Bhattarai, K.R.; Kim, H.-R.; Chae, H.-J. IRE1α Implications in Endoplasmic Reticulum Stress-Mediated Development and Pathogenesis of Autoimmune Diseases. Front. Immunol. 2018, 9, 1289. [Google Scholar] [CrossRef][Green Version]
  106. Teske, B.F.; Wek, S.A.; Bunpo, P.; Cundiff, J.K.; McClintick, J.N.; Anthony, T.G.; Wek, R.C. The EIF2 Kinase PERK and the Integrated Stress Response Facilitate Activation of ATF6 during Endoplasmic Reticulum Stress. Mol. Biol. Cell 2011, 22, 4390–4405. [Google Scholar] [CrossRef]
  107. Hillary, R.F.; FitzGerald, U. A Lifetime of Stress: ATF6 in Development and Homeostasis. J. Biomed. Sci. 2018, 25, 48. [Google Scholar] [CrossRef][Green Version]
  108. Wang, M.; Wey, S.; Zhang, Y.; Ye, R.; Lee, A.S. Role of the Unfolded Protein Response Regulator GRP78/BiP in Development, Cancer, and Neurological Disorders. Antioxid. Redox Signal. 2009, 11, 2307–2316. [Google Scholar] [CrossRef] [PubMed][Green Version]
  109. Dhuriya, Y.K.; Sharma, D. Necroptosis: A Regulated Inflammatory Mode of Cell Death. J. Neuroinflammation 2018, 15, 199. [Google Scholar] [CrossRef] [PubMed][Green Version]
  110. Daiger, S.P.; Bowne, S.J.; Sullivan, L.S. Genes and Mutations Causing Autosomal Dominant Retinitis Pigmentosa. Cold Spring Harb. Perspect. Med. 2014, 5, a017129. [Google Scholar] [CrossRef] [PubMed][Green Version]
  111. Massengill, M.T.; Lewin, A.S. Gene Therapy for Rhodopsin-Associated Autosomal Dominant Retinitis Pigmentosa. Int. Ophthalmol. Clin. 2021, 61, 79–96. [Google Scholar] [CrossRef] [PubMed]
  112. Li, T.; Snyder, W.K.; Olsson, J.E.; Dryja, T.P. Transgenic Mice Carrying the Dominant Rhodopsin Mutation P347S: Evidence for Defective Vectorial Transport of Rhodopsin to the Outer Segments. Proc. Natl. Acad. Sci. USA 1996, 93, 14176–14181. [Google Scholar] [CrossRef] [PubMed][Green Version]
  113. Kraft, T.W.; Allen, D.; Petters, R.M.; Hao, Y.; Peng, Y.-W.; Wong, F. Altered Light Responses of Single Rod Photoreceptors in Transgenic Pigs Expressing P347L or P347S Rhodopsin. Mol. Vis. 2005, 11, 1246–1256. [Google Scholar] [PubMed]
  114. Mussolino, C.; Sanges, D.; Marrocco, E.; Bonetti, C.; Di Vicino, U.; Marigo, V.; Auricchio, A.; Meroni, G.; Surace, E.M. Zinc-Finger-Based Transcriptional Repression of Rhodopsin in a Model of Dominant Retinitis Pigmentosa. EMBO Mol. Med. 2011, 3, 118–128. [Google Scholar] [CrossRef][Green Version]
  115. Chadderton, N.; Millington-Ward, S.; Palfi, A.; O’Reilly, M.; Tuohy, G.; Humphries, M.M.; Li, T.; Humphries, P.; Kenna, P.F.; Farrar, G.J. Improved Retinal Function in a Mouse Model of Dominant Retinitis Pigmentosa Following AAV-Delivered Gene Therapy. Mol. Ther. 2009, 17, 593–599. [Google Scholar] [CrossRef]
  116. Palfi, A.; Millington-Ward, S.; Chadderton, N.; O’Reilly, M.; Goldmann, T.; Humphries, M.M.; Li, T.; Wolfrum, U.; Humphries, P.; Kenna, P.F.; et al. Adeno-Associated Virus-Mediated Rhodopsin Replacement Provides Therapeutic Benefit in Mice with a Targeted Disruption of the Rhodopsin Gene. Hum. Gene Ther. 2010, 21, 311–323. [Google Scholar] [CrossRef]
  117. Millington-Ward, S.; Chadderton, N.; O’Reilly, M.; Palfi, A.; Goldmann, T.; Kilty, C.; Humphries, M.; Wolfrum, U.; Bennett, J.; Humphries, P.; et al. Suppression and Replacement Gene Therapy for Autosomal Dominant Disease in a Murine Model of Dominant Retinitis Pigmentosa. Mol. Ther. 2011, 19, 642–649. [Google Scholar] [CrossRef]
  118. Moore, S.M.; Skowronska-Krawczyk, D.; Chao, D.L. Targeting of the NRL Pathway as a Therapeutic Strategy to Treat Retinitis Pigmentosa. J. Clin. Med. 2020, 9, 2224. [Google Scholar] [CrossRef] [PubMed]
  119. Conte, I.; Hadfield, K.D.; Barbato, S.; Carrella, S.; Pizzo, M.; Bhat, R.S.; Carissimo, A.; Karali, M.; Porter, L.F.; Urquhart, J.; et al. MiR-204 Is Responsible for Inherited Retinal Dystrophy Associated with Ocular Coloboma. Proc. Natl. Acad. Sci. USA 2015, 112, E3236–E3245. [Google Scholar] [CrossRef] [PubMed][Green Version]
  120. Piano, I.; D’Antongiovanni, V.; Novelli, E.; Biagioni, M.; Dei Cas, M.; Paroni, R.C.; Ghidoni, R.; Strettoi, E.; Gargini, C. Myriocin Effect on Tvrm4 Retina, an Autosomal Dominant Pattern of Retinitis Pigmentosa. Front. Neurosci. 2020, 14, 372. [Google Scholar] [CrossRef] [PubMed]
  121. Jiang, H.; Xiong, S.; Xia, X. Retinitis Pigmentosa-Associated Rhodopsin Mutant T17M Induces Endoplasmic Reticulum (ER) Stress and Sensitizes Cells to ER Stress-Induced Cell Death. Mol. Med. Rep. 2014, 9, 1737–1742. [Google Scholar] [CrossRef][Green Version]
  122. Bhootada, Y.; Gully, C.; Gorbatyuk, M.S. Controlling PERK-ATF4-CHOP Branch of the UPR Is the Key to Reverse Retinal Degeneration of T17M Retina. Investig. Ophthalmol. Vis. Sci. 2015, 56, 5409. [Google Scholar]
  123. 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]
  124. Bowne, S. Mutations in the RP1 Gene Causing Autosomal Dominant Retinitis Pigmentosa. Hum. Mol. Genet. 1999, 8, 2121–2128. [Google Scholar] [CrossRef][Green Version]
  125. Liu, Q.; Zhou, J.; Daiger, S.P.; Farber, D.B.; Heckenlively, J.R.; Smith, J.E.; Sullivan, L.S.; Zuo, J.; Milam, A.H.; Pierce, E.A. Identification and Subcellular Localization of the RP1 Protein in Human and Mouse Photoreceptors. Investig. Ophthalmol. Vis. Sci. 2002, 43, 22–32. [Google Scholar]
  126. Liu, Q.; Saveliev, A.; Hu, J.; Cao, H.; Auricchio, A.; Pierce, E.A. The Degeneration of Photoreceptor Cells in the Rp1 Form of Retinitis Pigmentosa Is Caused by a Dominant Mechanism. Investig. Ophthalmol. Vis. Sci. 2009, 50, 2285. [Google Scholar]
  127. Athanasiou, D.; Aguila, M.; Bellingham, J.; Li, W.; McCulley, C.; Reeves, P.J.; Cheetham, M.E. The Molecular and Cellular Basis of Rhodopsin Retinitis Pigmentosa Reveals Potential Strategies for Therapy. Prog. Retin. Eye Res. 2018, 62, 1–23. [Google Scholar] [CrossRef][Green Version]
  128. Saqib, M.A.N.; Nikopoulos, K.; Ullah, E.; Sher Khan, F.; Iqbal, J.; Bibi, R.; Jarral, A.; Sajid, S.; Nishiguchi, K.M.; Venturini, G.; et al. Homozygosity Mapping Reveals Novel and Known Mutations in Pakistani Families with Inherited Retinal Dystrophies. Sci. Rep. 2015, 5, 9965. [Google Scholar] [CrossRef] [PubMed][Green Version]
  129. Van Schil, K.; Karlstetter, M.; Aslanidis, A.; Dannhausen, K.; Azam, M.; Qamar, R.; Leroy, B.P.; Depasse, F.; Langmann, T.; De Baere, E. Autosomal Recessive Retinitis Pigmentosa with Homozygous Rhodopsin Mutation E150K and Non-Coding Cis-Regulatory Variants in CRX-Binding Regions of SAMD7. Sci. Rep. 2016, 6, 21307. [Google Scholar] [CrossRef] [PubMed][Green Version]
  130. Zhang, N.; Kolesnikov, A.V.; Jastrzebska, B.; Mustafi, D.; Sawada, O.; Maeda, T.; Genoud, C.; Engel, A.; Kefalov, V.J.; Palczewski, K. Autosomal Recessive Retinitis Pigmentosa E150K Opsin Mice Exhibit Photoreceptor Disorganization. J. Clin. Investig. 2013, 123, 121–137. [Google Scholar] [CrossRef] [PubMed][Green Version]
  131. Collin, R.W.J.; van den Born, L.I.; Klevering, B.J.; de Castro-Miró, M.; Littink, K.W.; Arimadyo, K.; Azam, M.; Yazar, V.; Zonneveld, M.N.; Paun, C.C.; et al. High-Resolution Homozygosity Mapping Is a Powerful Tool to Detect Novel Mutations Causative of Autosomal Recessive RP in the Dutch Population. Investig. Ophthalmol. Vis. Sci. 2011, 52, 2227. [Google Scholar] [CrossRef][Green Version]
  132. Nassisi, M.; Smirnov, V.M.; Solis Hernandez, C.; Mohand-Saïd, S.; Condroyer, C.; Antonio, A.; Kühlewein, L.; Kempf, M.; Kohl, S.; Wissinger, B.; et al. CNGB1-related Rod-cone Dystrophy: A Mutation Review and Update. Hum. Mutat. 2021, 42, 641–666. [Google Scholar] [CrossRef] [PubMed]
  133. Jacobson, S.G.; Cideciyan, A.V.; Huang, W.C.; Sumaroka, A.; Roman, A.J.; Schwartz, S.B.; Luo, X.; Sheplock, R.; Dauber, J.M.; Swider, M.; et al. TULP1 Mutations Causing Early-Onset Retinal Degeneration: Preserved but Insensitive Macular Cones. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5354. [Google Scholar] [CrossRef][Green Version]
  134. Chhetri, J.; Jacobson, G.; Gueven, N. Zebrafish—On the Move towards Ophthalmological Research. Eye 2014, 28, 367–380. [Google Scholar] [CrossRef][Green Version]
  135. Martinez-Fernandez De La Camara, C.; Nanda, A.; Salvetti, A.P.; Fischer, M.D.; MacLaren, R.E. Gene Therapy for the Treatment of X-Linked Retinitis Pigmentosa. Expert Opin. Orphan Drugs 2018, 6, 167–177. [Google Scholar] [CrossRef][Green Version]
  136. Koyanagi, Y.; Akiyama, M.; Nishiguchi, K.M.; Momozawa, Y.; Kamatani, Y.; Takata, S.; Inai, C.; Iwasaki, Y.; Kumano, M.; Murakami, Y.; et al. Genetic Characteristics of Retinitis Pigmentosa in 1204 Japanese Patients. J. Med. Genet. 2019, 56, 662–670. [Google Scholar] [CrossRef]
  137. González-del Pozo, M.; Fernández-Suárez, E.; Martín-Sánchez, M.; Bravo-Gil, N.; Méndez-Vidal, C.; Rodríguez-de la Rúa, E.; Borrego, S.; Antiñolo, G. Unmasking Retinitis Pigmentosa Complex Cases by a Whole Genome Sequencing Algorithm Based on Open-Access Tools: Hidden Recessive Inheritance and Potential Oligogenic Variants. J. Transl. Med. 2020, 18, 73. [Google Scholar] [CrossRef][Green Version]
  138. Birtel, J.; Gliem, M.; Mangold, E.; Müller, P.L.; Holz, F.G.; Neuhaus, C.; Lenzner, S.; Zahnleiter, D.; Betz, C.; Eisenberger, T.; et al. Next-Generation Sequencing Identifies Unexpected Genotype-Phenotype Correlations in Patients with Retinitis Pigmentosa. PLoS ONE 2018, 13, e0207958. [Google Scholar] [CrossRef] [PubMed][Green Version]
  139. Millo, T.; Rivera, A.; Obolensky, A.; Marks-Ohana, D.; Xu, M.; Li, Y.; Wilhelm, E.; Gopalakrishnan, P.; Gross, M.; Rosin, B.; et al. Identification of Autosomal Recessive Novel Genes and Retinal Phenotypes in Members of the Solute Carrier (SLC) Superfamily. Genet. Med. 2022, 24, 1523–1535. [Google Scholar] [CrossRef] [PubMed]
  140. Jin, Z.-B.; Huang, X.-F.; Lv, J.-N.; Xiang, L.; Li, D.-Q.; Chen, J.; Huang, C.; Wu, J.; Lu, F.; Qu, J. SLC7A14 Linked to Autosomal Recessive Retinitis Pigmentosa. Nat. Commun. 2014, 5, 3517. [Google Scholar] [CrossRef] [PubMed][Green Version]
  141. Branham, K.; Matsui, H.; Biswas, P.; Guru, A.A.; Hicks, M.; Suk, J.J.; Li, H.; Jakubosky, D.; Long, T.; Telenti, A.; et al. Establishing the Involvement of the Novel Gene AGBL5 in Retinitis Pigmentosa by Whole Genome Sequencing. Physiol. Genom. 2016, 48, 922–927. [Google Scholar] [CrossRef] [PubMed][Green Version]
  142. Jindal, N.; Banik, A.; Prabhakar, S.; Vaiphie, K.; Anand, A. Alteration of Neurotrophic Factors After Transplantation of Bone Marrow Derived Lin-ve Stem Cell in NMDA-Induced Mouse Model of Retinal Degeneration. J. Cell. Biochem. 2017, 118, 1699–1711. [Google Scholar] [CrossRef]
  143. Kashani, A.H.; Uang, J.; Mert, M.; Rahhal, F.; Chan, C.; Avery, R.L.; Dugel, P.; Chen, S.; Lebkowski, J.; Clegg, D.O.; et al. Surgical Method for Implantation of a Biosynthetic Retinal Pigment Epithelium Monolayer for Geographic Atrophy: Experience from a Phase 1/2a Study. Ophthalmol. Retin. 2020, 4, 264–273. [Google Scholar] [CrossRef]
  144. Mandai, M.; Fujii, M.; Hashiguchi, T.; Sunagawa, G.A.; Ito, S.; Sun, J.; Kaneko, J.; Sho, J.; Yamada, C.; Takahashi, M. IPSC-Derived Retina Transplants Improve Vision in Rd1 End-Stage Retinal-Degeneration Mice. Stem Cell Rep. 2017, 8, 69–83. [Google Scholar] [CrossRef][Green Version]
  145. Salas, A.; Duarri, A.; Fontrodona, L.; Ramírez, D.M.; Badia, A.; Isla-Magrané, H.; Ferreira-de-Souza, B.; Zapata, M.Á.; Raya, Á.; Veiga, A.; et al. Cell Therapy with HiPSC-Derived RPE Cells and RPCs Prevents Visual Function Loss in a Rat Model of Retinal Degeneration. Mol. Ther.-Methods Clin. Dev. 2021, 20, 688–702. [Google Scholar] [CrossRef]
  146. Surendran, H.; Nandakumar, S.; Reddy, K.V.B.; Stoddard, J.; Mohan, K.V.; Upadhyay, P.K.; McGill, T.J.; Pal, R. Transplantation of Retinal Pigment Epithelium and Photoreceptors Generated Concomitantly via Small Molecule-Mediated Differentiation Rescues Visual Function in Rodent Models of Retinal Degeneration. Stem Cell Res. Ther. 2021, 12, 70. [Google Scholar] [CrossRef]
  147. Lin, B.; McLelland, B.T.; Aramant, R.B.; Thomas, B.B.; Nistor, G.; Keirstead, H.S.; Seiler, M.J. Retina Organoid Transplants Develop Photoreceptors and Improve Visual Function in RCS Rats With RPE Dysfunction. Investig. Ophthalmol. Vis. Sci. 2020, 61, 34. [Google Scholar] [CrossRef]
  148. Duarri, A.; Rodríguez-Bocanegra, E.; Martínez-Navarrete, G.; Biarnés, M.; García, M.; Ferraro, L.L.; Kuebler, B.; Aran, B.; Izquierdo, E.; Aguilera-Xiol, E.; et al. Transplantation of Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium in a Swine Model of Geographic Atrophy. Int. J. Mol. Sci. 2021, 22, 10497. [Google Scholar] [CrossRef] [PubMed]
  149. Thomas, B.B.; Lin, B.; Martinez-Camarillo, J.C.; Zhu, D.; McLelland, B.T.; Nistor, G.; Keirstead, H.S.; Humayun, M.S.; Seiler, M.J. Co-Grafts of Human Embryonic Stem Cell Derived Retina Organoids and Retinal Pigment Epithelium for Retinal Reconstruction in Immunodeficient Retinal Degenerate Royal College of Surgeons Rats. Front. Neurosci. 2021, 15, 752958. [Google Scholar] [CrossRef] [PubMed]
  150. Ferroni, L.; Gardin, C.; Tocco, I.; Epis, R.; Casadei, A.; Vindigni, V.; Mucci, G.; Zavan, B. Potential for Neural Differentiation of Mesenchymal Stem Cells. Adv. Biochem. Eng. Biotechnol. 2013, 129, 89–115. [Google Scholar] [CrossRef]
  151. Bassi, Ê.J.; de Almeida, D.C.; Moraes-Vieira, P.M.M.; Câmara, N.O.S. Exploring the Role of Soluble Factors Associated with Immune Regulatory Properties of Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2012, 8, 329–342. [Google Scholar] [CrossRef] [PubMed]
  152. Soleymaninejadian, E.; Pramanik, K.; Samadian, E. Immunomodulatory Properties of Mesenchymal Stem Cells: Cytokines and Factors. Am. J. Reprod. Immunol. 2012, 67, 1–8. [Google Scholar] [CrossRef][Green Version]
  153. Ding, S.L.S.; Kumar, S.; Mok, P.L. Cellular Reparative Mechanisms of Mesenchymal Stem Cells for Retinal Diseases. Int. J. Mol. Sci. 2017, 18, 1406. [Google Scholar] [CrossRef][Green Version]
  154. Brown, C.; Agosta, P.; McKee, C.; Walker, K.; Mazzella, M.; Alamri, A.; Svinarich, D.; Chaudhry, G.R. Human Primitive Mesenchymal Stem Cell-Derived Retinal Progenitor Cells Improved Neuroprotection, Neurogenesis, and Vision in Rd12 Mouse Model of Retinitis Pigmentosa. Stem Cell Res. Ther. 2022, 13, 148. [Google Scholar] [CrossRef]
  155. Kolomeyer, A.M.; Zarbin, M.A. Trophic Factors in the Pathogenesis and Therapy for Retinal Degenerative Diseases. Surv. Ophthalmol. 2014, 59, 134–165. [Google Scholar] [CrossRef]
  156. Talcott, K.E.; Ratnam, K.; Sundquist, S.M.; Lucero, A.S.; Lujan, B.J.; Tao, W.; Porco, T.C.; Roorda, A.; Duncan, J.L. Longitudinal Study of Cone Photoreceptors during Retinal Degeneration and in Response to Ciliary Neurotrophic Factor Treatment. Investig. Ophthalmol. Vis. Sci. 2011, 52, 2219. [Google Scholar] [CrossRef]
  157. Moisseiev, E.; Smit-McBride, Z.; Oltjen, S.; Zhang, P.; Zawadzki, R.J.; Motta, M.; Murphy, C.J.; Cary, W.; Annett, G.; Nolta, J.A.; et al. Intravitreal Administration of Human Bone Marrow CD34+ Stem Cells in a Murine Model of Retinal Degeneration. Investig. Ophthalmol. Vis. Sci. 2016, 57, 4125. [Google Scholar] [CrossRef]
  158. Soleimannejad, M.; Ebrahimi-Barough, S.; Nadri, S.; Riazi-Esfahani, M.; Soleimani, M.; Tavangar, S.M.; Ai, J. Retina Tissue Engineering by Conjunctiva Mesenchymal Stem Cells Encapsulated in Fibrin Gel: Hypotheses on Novel Approach to Retinal Diseases Treatment. Med. Hypotheses 2017, 101, 75–77. [Google Scholar] [CrossRef] [PubMed]
  159. Zhang, J.; Li, P.; Zhao, G.; He, S.; Xu, D.; Jiang, W.; Peng, Q.; Li, Z.; Xie, Z.; Zhang, H.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Protect Retina in a Mouse Model of Retinitis Pigmentosa by Anti-Inflammation through MiR-146a-Nr4a3 Axis. Stem Cell Res. Ther. 2022, 13, 394. [Google Scholar] [CrossRef] [PubMed]
  160. Pan, T.; Shen, H.; Yuan, S.; Lu, G.; Zhang, Y.; Wang, H.; Zhao, Y.; Sun, X.; Liu, Q. Combined Transplantation With Human Mesenchymal Stem Cells Improves Retinal Rescue Effect of Human Fetal RPE Cells in Retinal Degeneration Mouse Model. Investig. Ophthalmol. Vis. Sci. 2020, 61, 9. [Google Scholar] [CrossRef] [PubMed]
  161. Wang, N.-K.; Tosi, J.; Kasanuki, J.M.; Chou, C.L.; Kong, J.; Parmalee, N.; Wert, K.J.; Allikmets, R.; Lai, C.-C.; Chien, C.-L.; et al. Transplantation of Reprogrammed Embryonic Stem Cells Improves Visual Function in a Mouse Model for Retinitis Pigmentosa. Transplantation 2010, 89, 911–919. [Google Scholar] [CrossRef]
  162. Klassen, H.J.; Ng, T.F.; Kurimoto, Y.; Kirov, I.; Shatos, M.; Coffey, P.; Young, M.J. Multipotent Retinal Progenitors Express Developmental Markers, Differentiate into Retinal Neurons, and Preserve Light-Mediated Behavior. Investig. Ophthalmol. Vis. Sci. 2004, 45, 4167. [Google Scholar] [CrossRef]
  163. Qiu, G.; Seiler, M.J.; Thomas, B.B.; Wu, K.; Radosevich, M.; Sadda, S.R. Revisiting Nestin Expression in Retinal Progenitor Cells in Vitro and after Transplantation in Vivo. Exp. Eye Res. 2007, 84, 1047–1059. [Google Scholar] [CrossRef][Green Version]
  164. MacLaren, R.E.; Pearson, R.A.; MacNeil, A.; Douglas, R.H.; Salt, T.E.; Akimoto, M.; Swaroop, A.; Sowden, J.C.; Ali, R.R. Retinal Repair by Transplantation of Photoreceptor Precursors. Nature 2006, 444, 203–207. [Google Scholar] [CrossRef]
  165. He, X.-Y.; Zhao, C.-J.; Xu, H.; Chen, K.; Bian, B.-S.-J.; Gong, Y.; Weng, C.-H.; Zeng, Y.-X.; Fu, Y.; Liu, Y.; et al. Synaptic Repair and Vision Restoration in Advanced Degenerating Eyes by Transplantation of Retinal Progenitor Cells. Stem Cell Rep. 2021, 16, 1805–1817. [Google Scholar] [CrossRef]
  166. Stern, J.H.; Tian, Y.; Funderburgh, J.; Pellegrini, G.; Zhang, K.; Goldberg, J.L.; Ali, R.R.; Young, M.; Xie, Y.; Temple, S. Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell 2018, 22, 834–849. [Google Scholar] [CrossRef][Green Version]
  167. Ellingford, J.M.; Barton, S.; Bhaskar, S.; O’Sullivan, J.; Williams, S.G.; Lamb, J.A.; Panda, B.; Sergouniotis, P.I.; Gillespie, R.L.; Daiger, S.P.; et al. Molecular Findings from 537 Individuals with Inherited Retinal Disease. J. Med. Genet. 2016, 53, 761–767. [Google Scholar] [CrossRef][Green Version]
  168. Bi, A.; Cui, J.; Ma, Y.-P.; Olshevskaya, E.; Pu, M.; Dizhoor, A.M.; Pan, Z.-H. Ectopic Expression of a Microbial-Type Rhodopsin Restores Visual Responses in Mice with Photoreceptor Degeneration. Neuron 2006, 50, 23–33. [Google Scholar] [CrossRef] [PubMed][Green Version]
  169. Tomita, H.; Sugano, E.; Fukazawa, Y.; Isago, H.; Sugiyama, Y.; Hiroi, T.; Ishizuka, T.; Mushiake, H.; Kato, M.; Hirabayashi, M.; et al. Visual Properties of Transgenic Rats Harboring the Channelrhodopsin-2 Gene Regulated by the Thy-1.2 Promoter. PLoS ONE 2009, 4, e7679. [Google Scholar] [CrossRef] [PubMed][Green Version]
  170. Simon, C.-J.; Sahel, J.-A.; Duebel, J.; Herlitze, S.; Dalkara, D. Opsins for Vision Restoration. Biochem. Biophys. Res. Commun. 2020, 527, 325–330. [Google Scholar] [CrossRef] [PubMed]
  171. Zhang, F.; Vierock, J.; Yizhar, O.; Fenno, L.E.; Tsunoda, S.; Kianianmomeni, A.; Prigge, M.; Berndt, A.; Cushman, J.; Polle, J.; et al. The Microbial Opsin Family of Optogenetic Tools. Cell 2011, 147, 1446–1457. [Google Scholar] [CrossRef] [PubMed][Green Version]
  172. Deisseroth, K. Optogenetics: 10 Years of Microbial Opsins in Neuroscience. Nat. Neurosci. 2015, 18, 1213–1225. [Google Scholar] [CrossRef][Green Version]
  173. Gradinaru, V.; Zhang, F.; Ramakrishnan, C.; Mattis, J.; Prakash, R.; Diester, I.; Goshen, I.; Thompson, K.R.; Deisseroth, K. Molecular and Cellular Approaches for Diversifying and Extending Optogenetics. Cell 2010, 141, 154–165. [Google Scholar] [CrossRef][Green Version]
  174. Bedbrook, C.N.; Yang, K.K.; Rice, A.J.; Gradinaru, V.; Arnold, F.H. Machine Learning to Design Integral Membrane Channelrhodopsins for Efficient Eukaryotic Expression and Plasma Membrane Localization. PLoS Comput. Biol. 2017, 13, e1005786. [Google Scholar] [CrossRef][Green Version]
  175. Garita-Hernandez, M.; Guibbal, L.; Toualbi, L.; Routet, F.; Chaffiol, A.; Winckler, C.; Harinquet, M.; Robert, C.; Fouquet, S.; Bellow, S.; et al. Optogenetic Light Sensors in Human Retinal Organoids. Front. Neurosci. 2018, 12, 789. [Google Scholar] [CrossRef][Green Version]
  176. Gaub, B.M.; Berry, M.H.; Holt, A.E.; Reiner, A.; Kienzler, M.A.; Dolgova, N.; Nikonov, S.; Aguirre, G.D.; Beltran, W.A.; Flannery, J.G.; et al. Restoration of Visual Function by Expression of a Light-Gated Mammalian Ion Channel in Retinal Ganglion Cells or ON-Bipolar Cells. Proc. Natl. Acad. Sci. USA 2014, 111, E5574–E5583. [Google Scholar] [CrossRef][Green Version]
  177. Ameline, B.; Tshilenge, K.-T.; Weber, M.; Biget, M.; Libeau, L.; Caplette, R.; Mendes-Madeira, A.; Provost, N.; Guihal, C.; Picaud, S.; et al. Long-Term Expression of Melanopsin and Channelrhodopsin Causes No Gross Alterations in the Dystrophic Dog Retina. Gene Ther. 2017, 24, 735–741. [Google Scholar] [CrossRef]
  178. Nikonov, S.; Aravand, P.; Lyubarsky, A.; Nikonov, R.; Luo, A.J.; Wei, Z.; Maguire, A.M.; Phelps, N.T.; Shpylchak, I.; Willett, K.; et al. Restoration of Vision and Retinal Responses After Adeno-Associated Virus–Mediated Optogenetic Therapy in Blind Dogs. Transl. Vis. Sci. Technol. 2022, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  179. Chaffiol, A.; Caplette, R.; Jaillard, C.; Brazhnikova, E.; Desrosiers, M.; Dubus, E.; Duhamel, L.; Macé, E.; Marre, O.; Benoit, P.; et al. A New Promoter Allows Optogenetic Vision Restoration with Enhanced Sensitivity in Macaque Retina. Mol. Ther. 2017, 25, 2546–2560. [Google Scholar] [CrossRef] [PubMed][Green Version]
  180. Ganjawala, T.H.; Lu, Q.; Fenner, M.D.; Abrams, G.W.; Pan, Z.-H. Improved CoChR Variants Restore Visual Acuity and Contrast Sensitivity in a Mouse Model of Blindness under Ambient Light Conditions. Mol. Ther. 2019, 27, 1195–1205. [Google Scholar] [CrossRef] [PubMed]
  181. Tabata, K.; Sugano, E.; Hatakeyama, A.; Watanabe, Y.; Suzuki, T.; Ozaki, T.; Fukuda, T.; Tomita, H. Phototoxicities Caused by Continuous Light Exposure Were Not Induced in Retinal Ganglion Cells Transduced by an Optogenetic Gene. Int. J. Mol. Sci. 2021, 22, 6732. [Google Scholar] [CrossRef]
  182. Watanabe, Y.; Sugano, E.; Tabata, K.; Hatakeyama, A.; Sakajiri, T.; Fukuda, T.; Ozaki, T.; Suzuki, T.; Sayama, T.; Tomita, H. Development of an Optogenetic Gene Sensitive to Daylight and Its Implications in Vision Restoration. Npj Regen. Med. 2021, 6, 64. [Google Scholar] [CrossRef]
  183. Weis, W.I.; Kobilka, B.K. The Molecular Basis of G Protein-Coupled Receptor Activation. Annu. Rev. Biochem. 2018, 87, 897–919. [Google Scholar] [CrossRef]
  184. Katada, Y.; Yoshida, K.; Serizawa, N.; Kobayashi, K.; Neghisi, K.; Okano, H.; Kandori, H.; Tsubota, K.; Kurihara, T. High-Sensitivity Vision Restoration via Ectopic Expression of Chimeric Rhodopsin in Mice. bioRxiv 2020. [CrossRef]
  185. Buch, P.; MacLaren, R.; Ali, R. Neuroprotective Gene Therapy for the Treatment of Inherited Retinal Degeneration. Curr. Gene Ther. 2007, 7, 434–445. [Google Scholar] [CrossRef]
  186. Delplace, V.; Ortin-Martinez, A.; Tsai, E.L.S.; Amin, A.N.; Wallace, V.; Shoichet, M.S. Controlled Release Strategy Designed for Intravitreal Protein Delivery to the Retina. J. Control. Release 2019, 293, 10–20. [Google Scholar] [CrossRef]
  187. Gupta, V.; You, Y.; Gupta, V.; Klistorner, A.; Graham, S. TrkB Receptor Signalling: Implications in Neurodegenerative, Psychiatric and Proliferative Disorders. Int. J. Mol. Sci. 2013, 14, 10122–10142. [Google Scholar] [CrossRef][Green Version]
  188. Pardue, M.T.; Allen, R.S. Neuroprotective Strategies for Retinal Disease. Prog. Retin. Eye Res. 2018, 65, 50–76. [Google Scholar] [CrossRef] [PubMed]
  189. Wen, R.; Tao, W.; Li, Y.; Sieving, P.A. CNTF and Retina. Prog. Retin. Eye Res. 2012, 31, 136–151. [Google Scholar] [CrossRef] [PubMed][Green Version]
  190. Ghasemi, M.; Alizadeh, E.; Saei Arezoumand, K.; Fallahi Motlagh, B.; Zarghami, N. Ciliary Neurotrophic Factor (CNTF) Delivery to Retina: An Overview of Current Research Advancements. Artif. Cells Nanomed. Biotechnol. 2017, 46, 1694–1707. [Google Scholar] [CrossRef]
  191. Chew, E.Y.; Clemons, T.E.; Jaffe, G.J.; Johnson, C.A.; Farsiu, S.; Lad, E.M.; Guymer, R.; Rosenfeld, P.; Hubschman, J.-P.; Constable, I.; et al. Effect of Ciliary Neurotrophic Factor on Retinal Neurodegeneration in Patients with Macular Telangiectasia Type 2. Ophthalmology 2019, 126, 540–549. [Google Scholar] [CrossRef] [PubMed]
  192. Bielmeier, C.B.; Roth, S.; Schmitt, S.I.; Boneva, S.K.; Schlecht, A.; Vallon, M.; Tamm, E.R.; Ergün, S.; Neueder, A.; Braunger, B.M. Transcriptional Profiling Identifies Upregulation of Neuroprotective Pathways in Retinitis Pigmentosa. Int. J. Mol. Sci. 2021, 22, 6307. [Google Scholar] [CrossRef] [PubMed]
  193. Bramall, A.N.; Szego, M.J.; Pacione, L.R.; Chang, I.; Diez, E.; D’Orleans-Juste, P.; Stewart, D.J.; Hauswirth, W.W.; Yanagisawa, M.; McInnes, R.R. Endothelin-2-Mediated Protection of Mutant Photoreceptors in Inherited Photoreceptor Degeneration. PLoS ONE 2013, 8, e58023. [Google Scholar] [CrossRef][Green Version]
  194. Froger, N.; Matonti, F.; Roubeix, C.; Forster, V.; Ivkovic, I.; Brunel, N.; Baudouin, C.; Sahel, J.-A.; Picaud, S. VEGF Is an Autocrine/Paracrine Neuroprotective Factor for Injured Retinal Ganglion Neurons. Sci. Rep. 2020, 10, 12409. [Google Scholar] [CrossRef]
  195. Tao, W.; Wen, R.; Goddard, M.B.; Sherman, S.D.; O’Rourke, P.J.; Stabila, P.F.; Bell, W.J.; Dean, B.J.; Kauper, K.A.; Budz, V.A.; et al. Encapsulated Cell-Based Delivery of CNTF Reduces Photoreceptor Degeneration in Animal Models of Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2002, 43, 3292–3298. [Google Scholar]
  196. Yang, J.-Y.; Lu, B.; Feng, Q.; Alfaro, J.S.; Chen, P.-H.; Loscalzo, J.; Wei, W.-B.; Zhang, Y.-Y.; Lu, S.-J.; Wang, S. Retinal Protection by Sustained Nanoparticle Delivery of Oncostatin M and Ciliary Neurotrophic Factor Into Rodent Models of Retinal Degeneration. Transl. Vis. Sci. Technol. 2021, 10, 6. [Google Scholar] [CrossRef]
  197. Fernández-Sánchez, L.; Lax, P.; Pinilla, I.; Martín-Nieto, J.; Cuenca, N. Tauroursodeoxycholic Acid Prevents Retinal Degeneration in Transgenic P23H Rats. Investig. Ophthalmol. Vis. Sci. 2011, 52, 4998. [Google Scholar] [CrossRef][Green Version]
  198. Olivares-González, L.; Velasco, S.; Campillo, I.; Rodrigo, R. Retinal Inflammation, Cell Death and Inherited Retinal Dystrophies. Int. J. Mol. Sci. 2021, 22, 2096. [Google Scholar] [CrossRef] [PubMed]
  199. Omura, T.; Asari, M.; Yamamoto, J.; Oka, K.; Hoshina, C.; Maseda, C.; Awaya, T.; Tasaki, Y.; Shiono, H.; Yonezawa, A.; et al. Sodium Tauroursodeoxycholate Prevents Paraquat-Induced Cell Death by Suppressing Endoplasmic Reticulum Stress Responses in Human Lung Epithelial A549 Cells. Biochem. Biophys. Res. Commun. 2013, 432, 689–694. [Google Scholar] [CrossRef] [PubMed]
  200. Zhang, T.; Baehr, W.; Fu, Y. Chemical Chaperone TUDCA Preserves Cone Photoreceptors in a Mouse Model of Leber Congenital Amaurosis. Investig. Ophthalmol. Vis. Sci. 2012, 53, 3349. [Google Scholar] [CrossRef][Green Version]
  201. Li, K.; Li, J.; Zheng, J.; Qin, S. Reactive Astrocytes in Neurodegenerative Diseases. Aging Dis. 2019, 10, 664. [Google Scholar] [CrossRef][Green Version]
  202. Eltony, S.A.; Mohaseb, H.S.; Ahmed, A.A.; Sayed, M.M. Can Metformin Modulate the Retinal Degenerative Changes in a Rat Model of Retinitis Pigmentosa? Tissue Cell 2022, 76, 101786. [Google Scholar] [CrossRef] [PubMed]
  203. Kilicarslan, I.; Zanetti, L.; Novelli, E.; Schwarzer, C.; Strettoi, E.; Koschak, A. Knockout of CaV1.3 L-Type Calcium Channels in a Mouse Model of Retinitis Pigmentosa. Sci. Rep. 2021, 11, 15146. [Google Scholar] [CrossRef]
  204. Aloe, L.; Rocco, M.L.; Bianchi, P.; Manni, L. Nerve Growth Factor: From the Early Discoveries to the Potential Clinical Use. J. Transl. Med. 2012, 10, 239. [Google Scholar] [CrossRef][Green Version]
  205. Li, B.; Ning, B.; Yang, F.; Guo, C. Nerve Growth Factor Promotes Retinal Neurovascular Unit Repair: A Review. Curr. Eye Res. 2022, 47, 1095–1105. [Google Scholar] [CrossRef]
  206. Wiatrak, B.; Kubis-Kubiak, A.; Piwowar, A.; Barg, E. PC12 Cell Line: Cell Types, Coating of Culture Vessels, Differentiation and Other Culture Conditions. Cells 2020, 9, 958. [Google Scholar] [CrossRef]
  207. Rocco, M.L.; Balzamino, B.O.; Petrocchi Passeri, P.; Micera, A.; Aloe, L. Effect of Purified Murine NGF on Isolated Photoreceptors of a Rodent Developing Retinitis Pigmentosa. PLoS ONE 2015, 10, e0124810. [Google Scholar] [CrossRef][Green Version]
  208. Rocco, M.L.; Calzà, L.; Aloe, L. NGF and Retinitis Pigmentosa: Structural and Molecular Studies. In Recent Advances in NGF and Related Molecules; Calzà, L., Aloe, L., Giardino, L., Eds.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2021; Volume 1331, pp. 255–263. ISBN 978-3-030-74045-0. [Google Scholar]
  209. Cocchiaro, P.; Di Donato, V.; Rubbini, D.; Mastropasqua, R.; Allegretti, M.; Mantelli, F.; Aramini, A.; Brandolini, L. Intravitreal Administration of RhNGF Enhances Regenerative Processes in a Zebrafish Model of Retinal Degeneration. Front. Pharmacol. 2022, 13, 822359. [Google Scholar] [CrossRef]
  210. Bielmeier, C.B.; Schmitt, S.I.; Kleefeldt, N.; Boneva, S.K.; Schlecht, A.; Vallon, M.; Tamm, E.R.; Hillenkamp, J.; Ergün, S.; Neueder, A.; et al. Deficiency in Retinal TGFβ Signaling Aggravates Neurodegeneration by Modulating Pro-Apoptotic and MAP Kinase Pathways. Int. J. Mol. Sci. 2022, 23, 2626. [Google Scholar] [CrossRef]
  211. Eastlake, K.; Lamb, W.D.B.; Luis, J.; Khaw, P.T.; Jayaram, H.; Limb, G.A. Prospects for the Application of Müller Glia and Their Derivatives in Retinal Regenerative Therapies. Prog. Retin. Eye Res. 2021, 85, 100970. [Google Scholar] [CrossRef]
  212. 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]
  213. Rasmussen, M.; Zhou, J.; Schwede, F.; Ekström, P. Enhanced CGMP Interactor Rap Guanine Exchange Factor 4 (EPAC2) Expression and Activity in Degenerating Photoreceptors: A Neuroprotective Response? Int. J. Mol. Sci. 2022, 23, 4619. [Google Scholar] [CrossRef]
  214. Ozawa, Y.; Toda, E.; Homma, K.; Osada, H.; Nagai, N.; Tsubota, K.; Okano, H. Effects of Epigenetic Modification of PGC-1α by a Chemical Chaperon on Mitochondria Biogenesis and Visual Function in Retinitis Pigmentosa. Cells 2022, 11, 1497. [Google Scholar] [CrossRef]
  215. Campochiaro, P.A.; Mir, T.A. The Mechanism of Cone Cell Death in Retinitis Pigmentosa. Prog. Retin. Eye Res. 2018, 62, 24–37. [Google Scholar] [CrossRef]
  216. Komeima, K.; Rogers, B.S.; Lu, L.; Campochiaro, P.A. Antioxidants Reduce Cone Cell Death in a Model of Retinitis Pigmentosa. Proc. Natl. Acad. Sci. USA 2006, 103, 11300–11305. [Google Scholar] [CrossRef] [PubMed][Green Version]
  217. Lee, S.Y.; Usui, S.; Zafar, A.; Oveson, B.C.; Jo, Y.-J.; Lu, L.; Masoudi, S.; Campochiaro, P.A. N-Acetylcysteine Promotes Long-Term Survival of Cones in a Model of Retinitis Pigmentosa. J. Cell. Physiol. 2011, 226, 1843–1849. [Google Scholar] [CrossRef] [PubMed]
  218. Yoshida, N.; Ikeda, Y.; Notomi, S.; Ishikawa, K.; Murakami, Y.; Hisatomi, T.; Enaida, H.; Ishibashi, T. Laboratory Evidence of Sustained Chronic Inflammatory Reaction in Retinitis Pigmentosa. Ophthalmology 2013, 120, e5–e12. [Google Scholar] [CrossRef] [PubMed]
  219. Johansson, I.; Monsen, V.T.; Pettersen, K.; Mildenberger, J.; Misund, K.; Kaarniranta, K.; Schønberg, S.; Bjørkøy, G. The Marine N-3 PUFA DHA Evokes Cytoprotection against Oxidative Stress and Protein Misfolding by Inducing Autophagy and NFE2L2 in Human Retinal Pigment Epithelial Cells. Autophagy 2015, 11, 1636–1651. [Google Scholar] [CrossRef] [PubMed][Green Version]
  220. 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] [PubMed][Green Version]
  221. Morshedian, A.; Kaylor, J.J.; Ng, S.Y.; Tsan, A.; Frederiksen, R.; Xu, T.; Yuan, L.; Sampath, A.P.; Radu, R.A.; Fain, G.L.; et al. Light-Driven Regeneration of Cone Visual Pigments through a Mechanism Involving RGR Opsin in Müller Glial Cells. Neuron 2019, 102, 1172–1183.e5. [Google Scholar] [CrossRef] [PubMed]
  222. Ozaki, T.; Nakazawa, M.; Kudo, T.; Hirano, S.; Suzuki, K.; Ishiguro, S. Protection of Cone Photoreceptor M-Opsin Degradation with 9-Cis-β-Carotene-Rich Alga Dunaliella Bardawil in Rpe65(−/−) Mouse Retinal Explant Culture. Curr. Eye Res. 2014, 39, 1221–1231. [Google Scholar] [CrossRef] [PubMed]
  223. Pawlyk, B.S.; Li, T.; Scimeca, M.S.; Sandberg, M.A.; Berson, E.L. Absence of Photoreceptor Rescue with D-Cis-Diltiazem in the Rd Mouse. Investig. Ophthalmol. Vis. Sci. 2002, 43, 1912–1915. [Google Scholar]
  224. Yang, J.; Weimer, R.M.; Kallop, D.; Olsen, O.; Wu, Z.; Renier, N.; Uryu, K.; Tessier-Lavigne, M. Regulation of Axon Degeneration after Injury and in Development by the Endogenous Calpain Inhibitor Calpastatin. Neuron 2013, 80, 1175–1189. [Google Scholar] [CrossRef][Green Version]
  225. Yamamoto, S.; Sugawara, T.; Murakami, A.; Nakazawa, M.; Nao-I, N.; Machida, S.; Wada, Y.; Mashima, Y.; Myake, Y. Topical Isopropyl Unoprostone for Retinitis Pigmentosa: Microperimetric Results of the Phase 2 Clinical Study. Ophthalmol. Ther. 2012, 1, 5. [Google Scholar] [CrossRef][Green Version]
  226. Sahaboglu, A.; Vidal-Gil, L.; Sancho-Pelluz, J. Release of Retinal Extracellular Vesicles in a Model of Retinitis Pigmentosa. In Retinal Degenerative Diseases; Bowes Rickman, C., Grimm, C., Anderson, R.E., Ash, J.D., LaVail, M.M., Hollyfield, J.G., Eds.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2019; Volume 1185, pp. 431–436. ISBN 978-3-030-27377-4. [Google Scholar]
  227. Knickelbein, J.E.; Liu, B.; Arakelyan, A.; Zicari, S.; Hannes, S.; Chen, P.; Li, Z.; Grivel, J.-C.; Chaigne-Delalande, B.; Sen, H.N.; et al. Modulation of Immune Responses by Extracellular Vesicles From Retinal Pigment Epithelium. Investig. Ophthalmol. Vis. Sci. 2016, 57, 4101. [Google Scholar] [CrossRef][Green Version]
  228. Ma, M.; Li, B.; Zhang, M.; Zhou, L.; Yang, F.; Ma, F.; Shao, H.; Li, Q.; Li, X.; Zhang, X. Therapeutic Effects of Mesenchymal Stem Cell-Derived Exosomes on Retinal Detachment. Exp. Eye Res. 2020, 191, 107899. [Google Scholar] [CrossRef]
  229. Liu, Y.; Zhou, T.; Yang, Z.; Sun, X.; Huang, Z.; Deng, X.; He, C.; Liu, X. Bone Marrow Mesenchymal Stem Cells-Derived Exosomes Attenuate Neuroinflammation and Promote Survival of Photoreceptor in Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3108. [Google Scholar]
  230. Odagiu, L.; May, J.; Boulet, S.; Baldwin, T.A.; Labrecque, N. Role of the Orphan Nuclear Receptor NR4A Family in T-Cell Biology. Front. Endocrinol. 2020, 11, 624122. [Google Scholar] [CrossRef] [PubMed]
  231. Wang, Y.; Zhang, Q.; Yang, G.; Wei, Y.; Li, M.; Du, E.; Li, H.; Song, Z.; Tao, Y. RPE-Derived Exosomes Rescue the Photoreceptors during Retina Degeneration: An Intraocular Approach to Deliver Exosomes into the Subretinal Space. Drug Deliv. 2021, 28, 218–228. [Google Scholar] [CrossRef] [PubMed]
  232. Deng, C.-L.; Hu, C.-B.; Ling, S.-T.; Zhao, N.; Bao, L.-H.; Zhou, F.; Xiong, Y.-C.; Chen, T.; Sui, B.-D.; Yu, X.-R.; et al. Photoreceptor Protection by Mesenchymal Stem Cell Transplantation Identifies Exosomal MiR-21 as a Therapeutic for Retinal Degeneration. Cell Death Differ. 2021, 28, 1041–1061. [Google Scholar] [CrossRef] [PubMed]
  233. Jenike, A.E.; Halushka, M.K. MiR-21: A Non-specific Biomarker of All Maladies. Biomark. Res. 2021, 9, 18. [Google Scholar] [CrossRef]
  234. Bian, B.; Zhao, C.; He, X.; Gong, Y.; Ren, C.; Ge, L.; Zeng, Y.; Li, Q.; Chen, M.; Weng, C.; et al. Exosomes Derived from Neural Progenitor Cells Preserve Photoreceptors during Retinal Degeneration by Inactivating Microglia. J. Extracell. Vesicles 2020, 9, 1748931. [Google Scholar] [CrossRef][Green Version]
  235. Zhang, Z.; Mugisha, A.; Fransisca, S.; Liu, Q.; Xie, P.; Hu, Z. Emerging Role of Exosomes in Retinal Diseases. Front. Cell Dev. Biol. 2021, 9, 643680. [Google Scholar] [CrossRef]
  236. Elliott, R.O.; He, M. Unlocking the Power of Exosomes for Crossing Biological Barriers in Drug Delivery. Pharmaceutics 2021, 13, 122. [Google Scholar] [CrossRef]
  237. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering Exosomes for Targeted Drug Delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
  238. Hung, M.E.; Leonard, J.N. Stabilization of Exosome-Targeting Peptides via Engineered Glycosylation. J. Biol. Chem. 2015, 290, 8166–8172. [Google Scholar] [CrossRef][Green Version]
  239. Boukouris, S.; Mathivanan, S. Exosomes in Bodily Fluids Are a Highly Stable Resource of Disease Biomarkers. Proteom. Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef][Green Version]
  240. Wassmer, S.J.; Carvalho, L.S.; György, B.; Vandenberghe, L.H.; Maguire, C.A. Exosome-Associated AAV2 Vector Mediates Robust Gene Delivery into the Murine Retina upon Intravitreal Injection. Sci. Rep. 2017, 7, 45329. [Google Scholar] [CrossRef] [PubMed][Green Version]
More
This entry is offline, you can click here to edit this entry!
Video Production Service