ijms-logo

Journal Browser

Journal Browser

Retinal Degeneration—From Genetics to Therapy

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Biology".

Deadline for manuscript submissions: closed (15 April 2023) | Viewed by 19002

Special Issue Editors


E-Mail Website
Guest Editor
Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY 10032, USA
Interests: genetics; mitochondrial function; inherited retinal dystrophies; optic neuropathy; electrophysiology; mouse models; myopic maculopathy; diabetes
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY 10032, USA
Interests: organoids; organ chips; induced pluripotent stem cells; neurodegeneration; neuroinflammation; retina, RPE, mitochondria; microglia; Crumbs homologue-1; CRB1; metabolome reprogramming, gene augmentation; gene editing; cell therapy
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

We are pleased to announce a new Special Issue of the International Journal of Molecular Sciences entitled “Retinal Degeneration—from Genetics to Therapy”, in which we invite you to submit your latest research findings in the fields of genetic and translational ophthalmology.

Loss of vision is one of the most feared health conditions in society and severely impacts the quality of life. The burden and prevalence of monogenic and multifactorial ophthalmic diseases in the human population highlights the need to better understand the phenotypic, histopathological, and molecular changes underlying these diseases. As such, there is an urgent need for the development, testing, and refinement of potential treatments to halt, slow, or hopefully cure progressive and disabling retinal degenerative diseases.

In this issue, we aim to gather a broad scope of contributions covering but not limited to animal models and induced pluripotent stem cell (iPSC)-derived retina or retinal pigment epithelium (RPE) models associated with retinal degeneration, prospective and retrospective natural history studies, hereditary disease, retinal imaging and therapeutics toward the amelioration of retinal degeneration including gene editing, gene augmentation, metabolic reprogramming, and splice modulation therapy.

The published papers in this Special Issue of IJMS will describe new developments in these areas. We will accept high-quality articles including original research results, case reports, and reviews.

We look forward to your contributions.

Dr. Nan-Kai Wang
Dr. Peter M.J. Quinn
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • ophthalmology
  • inherited retinal dystrophies
  • retinal degeneration
  • genetics
  • natural history studies
  • gene augmentation
  • gene editing
  • reprogramming
  • optogenetics and splice modulation therapy
  • animals models
  • human iPSC-derived retina and retinal pigment epithelium

Related Special Issue

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

14 pages, 4248 KiB  
Article
Long-Term Evaluation of Retinal Morphology and Function in Rosa26-Cas9 Knock-In Mice
by Kabhilan Mohan, Sushil Kumar Dubey, Kyungsik Jung, Rashmi Dubey, Qing Jun Wang, Subhash Prajapati, Jacob Roney, Jennifer Abney and Mark Ellsworth Kleinman
Int. J. Mol. Sci. 2023, 24(6), 5186; https://doi.org/10.3390/ijms24065186 - 8 Mar 2023
Viewed by 1753
Abstract
The CRISPR/Cas9 system is a robust, efficient, and cost-effective gene editing tool widely adopted in translational studies of ocular diseases. However, in vivo CRISPR-based editing in animal models poses challenges such as the efficient delivery of the CRISPR components in viral vectors with [...] Read more.
The CRISPR/Cas9 system is a robust, efficient, and cost-effective gene editing tool widely adopted in translational studies of ocular diseases. However, in vivo CRISPR-based editing in animal models poses challenges such as the efficient delivery of the CRISPR components in viral vectors with limited packaging capacity and a Cas9-associated immune response. Using a germline Cas9-expressing mouse model would help to overcome these limitations. Here, we evaluated the long-term effects of SpCas9 expression on retinal morphology and function using Rosa26-Cas9 knock-in mice. We observed abundant SpCas9 expression in the RPE and retina of Rosa26-Cas9 mice using the real-time polymerase chain reaction (RT-PCR), Western blotting, and immunostaining. SD-OCT imaging and histological analysis of the RPE, retinal layers, and vasculature showed no apparent structural abnormalities in adult and aged Cas9 mice. Full-field electroretinogram of adult and aged Cas9 mice showed no long-term functional changes in the retinal tissues because of constitutive Cas9 expression. The current study showed that both the retina and RPE maintain their phenotypic and functional features in Cas9 knock-in mice, establishing this as an ideal animal model for developing therapeutics for retinal diseases. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Graphical abstract

18 pages, 2305 KiB  
Article
Transcriptome Analysis of Retinal and Choroidal Pathologies in Aged BALB/c Mice Following Systemic Neonatal Murine Cytomegalovirus Infection
by Xinyan Zhang, Jinxian Xu, Brendan Marshall, Zheng Dong, Yutao Liu, Diego G. Espinosa-Heidmann and Ming Zhang
Int. J. Mol. Sci. 2023, 24(5), 4322; https://doi.org/10.3390/ijms24054322 - 21 Feb 2023
Viewed by 1373
Abstract
Our previous studies have shown that systemic neonatal murine cytomegalovirus (MCMV) infection of BALB/c mice spread to the eye with subsequent establishment of latency in choroid/RPE. In this study, RNA sequencing (RNA-Seq) analysis was used to determine the molecular genetic changes and pathways [...] Read more.
Our previous studies have shown that systemic neonatal murine cytomegalovirus (MCMV) infection of BALB/c mice spread to the eye with subsequent establishment of latency in choroid/RPE. In this study, RNA sequencing (RNA-Seq) analysis was used to determine the molecular genetic changes and pathways affected by ocular MCMV latency. MCMV (50 pfu per mouse) or medium as control were injected intra-peritoneally (i.p.) into BALB/c mice at <3 days after birth. At 18 months post injection, the mice were euthanized, and the eyes were collected and prepared for RNA-Seq. Compared to three uninfected control eyes, we identified 321 differentially expressed genes (DEGs) in six infected eyes. Using the QIAGEN Ingenuity Pathway Analysis (QIAGEN IPA), we identified 17 affected canonical pathways, 10 of which function in neuroretinal signaling, with the majority of DEGs being downregulated, while 7 pathways function in upregulated immune/inflammatory responses. Retinal and epithelial cell death pathways involving both apoptosis and necroptosis were also activated. MCMV ocular latency is associated with upregulation of immune and inflammatory responses and downregulation of multiple neuroretinal signaling pathways. Cell death signaling pathways are also activated and contribute to the degeneration of photoreceptors, RPE, and choroidal capillaries. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

19 pages, 5457 KiB  
Article
Analysis of Electric Field Stimulation in Blue Light Stressed 661W Cells
by Sharanya Bola, Pallavi Subramanian, Daniela Calzia, Andreas Dahl, Isabella Panfoli, Richard H. W. Funk and Cora Roehlecke
Int. J. Mol. Sci. 2023, 24(4), 3433; https://doi.org/10.3390/ijms24043433 - 8 Feb 2023
Viewed by 1465
Abstract
Though electrical stimulation is used as a therapeutic approach to treat retinal and spinal injuries, many protective mechanisms at cellular level have not been elucidated. We performed a detailed analysis of cellular events in blue light (Li) stressed 661W cells, which were subjected [...] Read more.
Though electrical stimulation is used as a therapeutic approach to treat retinal and spinal injuries, many protective mechanisms at cellular level have not been elucidated. We performed a detailed analysis of cellular events in blue light (Li) stressed 661W cells, which were subjected to direct current electric field (EF) stimulation. Our findings revealed that EF stimulation induced protective effects in 661W cells from Li-induced stress by multiple defense mechanisms, such as increase in mitochondrial activity, gain in mitochondrial potential, increase in superoxide levels, and the activation of unfolded protein response (UPR) pathways, all leading to an enhanced cell viability and decreased DNA damage. Here, our genetic screen results revealed the UPR pathway to be a promising target to ameliorate Li-induced stress by EF stimulation. Thus, our study is important for a knowledgeable transfer of EF stimulation into clinical application. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

16 pages, 3074 KiB  
Article
Congenital Stationary Night Blindness: Clinical and Genetic Features
by Angela H. Kim, Pei-Kang Liu, Yin-Hsi Chang, Eugene Yu-Chuan Kang, Hung-Hsuan Wang, Nelson Chen, Yun-Ju Tseng, Go Hun Seo, Hane Lee, Laura Liu, An-Ning Chao, Kuan-Jen Chen, Yih-Shiou Hwang, Wei-Chi Wu, Chi-Chun Lai, Stephen H. Tsang, Meng-Chang Hsiao and Nan-Kai Wang
Int. J. Mol. Sci. 2022, 23(23), 14965; https://doi.org/10.3390/ijms232314965 - 29 Nov 2022
Cited by 7 | Viewed by 4213
Abstract
Congenital stationary night blindness (CSNB) is an inherited retinal disease (IRD) that causes night blindness in childhood with heterogeneous genetic, electrophysical, and clinical characteristics. The development of sequencing technologies and gene therapy have increased the ease and urgency of diagnosing IRDs. This study [...] Read more.
Congenital stationary night blindness (CSNB) is an inherited retinal disease (IRD) that causes night blindness in childhood with heterogeneous genetic, electrophysical, and clinical characteristics. The development of sequencing technologies and gene therapy have increased the ease and urgency of diagnosing IRDs. This study describes seven Taiwanese patients from six unrelated families examined at a tertiary referral center, diagnosed with CSNB, and confirmed by genetic testing. Complete ophthalmic exams included best corrected visual acuity, retinal imaging, and an electroretinogram. The effects of identified novel variants were predicted using clinical details, protein prediction tools, and conservation scores. One patient had an autosomal dominant CSNB with a RHO variant; five patients had complete CSNB with variants in GRM6, TRPM1, and NYX; and one patient had incomplete CSNB with variants in CACNA1F. The patients had Riggs and Schubert–Bornschein types of CSNB with autosomal dominant, autosomal recessive, and X-linked inheritance patterns. This is the first report of CSNB patients in Taiwan with confirmed genetic testing, providing novel perspectives on molecular etiology and genotype–phenotype correlation of CSNB. Particularly, variants in TRPM1, NYX, and CACNA1F in our patient cohort have not previously been described, although their clinical significance needs further study. Additional study is needed for the genotype–phenotype correlation of different mutations causing CSNB. In addition to genetic etiology, the future of gene therapy for CSNB patients is reviewed and discussed. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

24 pages, 5039 KiB  
Article
Depleted Calcium Stores and Increased Calcium Entry in Rod Photoreceptors of the Cacna2d4 Mouse Model of Cone-Rod Dystrophy RCD4
by Vittorio Vellani, Giovanna Mauro and Gian Carlo Demontis
Int. J. Mol. Sci. 2022, 23(21), 13080; https://doi.org/10.3390/ijms232113080 - 28 Oct 2022
Cited by 2 | Viewed by 1442
Abstract
Unidentified pathogenetic mechanisms and genetic and clinical heterogeneity represent critical factors hindering the development of treatments for inherited retinal dystrophies. Frameshift mutations in Cacna2d4, which codes for an accessory subunit of voltage-gated calcium channels (VGCC), cause cone-rod dystrophy RCD4 in patients, but [...] Read more.
Unidentified pathogenetic mechanisms and genetic and clinical heterogeneity represent critical factors hindering the development of treatments for inherited retinal dystrophies. Frameshift mutations in Cacna2d4, which codes for an accessory subunit of voltage-gated calcium channels (VGCC), cause cone-rod dystrophy RCD4 in patients, but the underlying mechanisms remain unknown. To define its pathogenetic mechanisms, we investigated the impact of a Cacna2d4 frameshift mutation on the electrophysiological profile and calcium handling of mouse rod photoreceptors by patch-clamp recordings and calcium imaging, respectively. In mutant (MUT) rods, the dysregulation of calcium handling extends beyond the reduction in calcium entry through VGCC and surprisingly involves internal calcium stores’ depletion and upregulation of calcium entry via non-selective cationic channels (CSC). The similar dependence of CSC on basal calcium levels in WT and MUT rods suggests that the primary defect in MUT rods lies in defective calcium stores. Calcium stores’ depletion, leading to upregulated calcium and sodium influx via CSC, represents a novel and, so far, unsuspected consequence of the Cacna2d4 mutation. Blocking CSC may provide a novel strategy to counteract the well-known pathogenetic mechanisms involved in rod demise, such as the reticulum stress response and calcium and sodium overload due to store depletion. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

10 pages, 4110 KiB  
Communication
Tropism of the Novel AAVBR1 Capsid Following Subretinal Delivery
by Lara Carroll, Hironori Uehara, Xiaohui Zhang and Balamurali Ambati
Int. J. Mol. Sci. 2022, 23(14), 7738; https://doi.org/10.3390/ijms23147738 - 13 Jul 2022
Viewed by 1594
Abstract
A serious limitation of current adeno-associated viral (AAV) capsids employed for subretinal delivery is achieving adequate lateral spread beyond the injection site, required for the efficient delivery of gene therapy to the outer retina and/or RPE. AAVBR1 is a unique AAV with exceptional [...] Read more.
A serious limitation of current adeno-associated viral (AAV) capsids employed for subretinal delivery is achieving adequate lateral spread beyond the injection site, required for the efficient delivery of gene therapy to the outer retina and/or RPE. AAVBR1 is a unique AAV with exceptional tropism for CNS microvasculature following systemic delivery. Here, we used in vivo and ex vivo analysis to show that subretinal delivery of AAVBR1.GFP in mice achieves superior tropism to RPE and outer retina than either AAV2.GFP or AAV8.GFP, two of the most common capsids used for subretinal delivery. At a low (5 × 108 vg) subretinal dose, the AAVBR1.GFP signal was visible by 48 h and significantly surpassed peak fluorescence of other AAVs in retina and RPE. The co-injection of AAVBR1.GFP with the AAVBR1-specific heptapeptide, NRGTEWD, significantly blocked the AAVBR1.GFP signal, but had no effect on AAV2.GFP fluorescence, confirming that AAVBR1’s enhanced tropism for RPE and outer retina derives from this 7AA modification within the capsid-binding motif. Enhanced dispersal and consequent transduction suggest that AAVBR1 can be employed at a lower dosage than the standard AAV2 capsid to achieve equivalent expression for gene therapy, warranting further evaluation of its utility as a therapeutic vehicle for subretinal delivery. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

Review

Jump to: Research, Other

16 pages, 8080 KiB  
Review
Primary versus Secondary Elevations in Fundus Autofluorescence
by Rait Parmann, Stephen H. Tsang and Janet R. Sparrow
Int. J. Mol. Sci. 2023, 24(15), 12327; https://doi.org/10.3390/ijms241512327 - 2 Aug 2023
Cited by 1 | Viewed by 1119
Abstract
The method of quantitative fundus autofluorescence (qAF) can be used to assess the levels of bisretinoids in retinal pigment epithelium (RPE) cells so as to aid the interpretation and management of a variety of retinal conditions. In this review, we focused on seven [...] Read more.
The method of quantitative fundus autofluorescence (qAF) can be used to assess the levels of bisretinoids in retinal pigment epithelium (RPE) cells so as to aid the interpretation and management of a variety of retinal conditions. In this review, we focused on seven retinal diseases to highlight the possible pathways to increased fundus autofluorescence. ABCA4- and RDH12-associated diseases benefit from known mechanisms whereby gene malfunctioning leads to elevated bisretinoid levels in RPE cells. On the other hand, peripherin2/RDS-associated disease (PRPH2/RDS), retinitis pigmentosa (RP), central serous chorioretinopathy (CSC), acute zonal occult outer retinopathy (AZOOR), and ceramide kinase like (CERKL)-associated retinal degeneration all express abnormally high fundus autofluorescence levels without a demonstrated pathophysiological pathway for bisretinoid elevation. We suggest that, while a known link from gene mutation to increased production of bisretinoids (as in ABCA4- and RDH12-associated diseases) causes primary elevation in fundus autofluorescence, a secondary autofluorescence elevation also exists, where an impairment and degeneration of photoreceptor cells by various causes leads to an increase in bisretinoid levels in RPE cells. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

12 pages, 1367 KiB  
Review
Base and Prime Editing in the Retina—From Preclinical Research toward Human Clinical Trials
by Tiffany Yee and Katherine J. Wert
Int. J. Mol. Sci. 2022, 23(20), 12375; https://doi.org/10.3390/ijms232012375 - 16 Oct 2022
Cited by 5 | Viewed by 3146
Abstract
Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous group of diseases that are one of the leading causes of vision loss in young and aged individuals. IRDs are mainly caused by a loss of the post-mitotic photoreceptor neurons of the retina, [...] Read more.
Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous group of diseases that are one of the leading causes of vision loss in young and aged individuals. IRDs are mainly caused by a loss of the post-mitotic photoreceptor neurons of the retina, or by the degeneration of the retinal pigment epithelium. Unfortunately, once these cells are damaged, it is irreversible and leads to permanent vision impairment. Thought to be previously incurable, gene therapy has been rapidly evolving to be a potential treatment to prevent further degeneration of the retina and preserve visual function. The development of clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) base and prime editors have increased the capabilities of the genome editing toolbox in recent years. Both base and prime editors evade the creation of double-stranded breaks in deoxyribonucleic acid (DNA) and the requirement of donor template of DNA for repair, which make them advantageous methods in developing clinical therapies. In addition, establishing a permanent edit within the genome could be better suited for patients with progressive degeneration. In this review, we will summarize published uses of successful base and prime editing in treating IRDs. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

Other

Jump to: Research, Review

9 pages, 1306 KiB  
Case Report
Large Benefit from Simple Things: High-Dose Vitamin A Improves RBP4-Related Retinal Dystrophy
by Vasily M. Smirnov, Baptiste Wilmet, Marco Nassisi, Christel Condroyer, Aline Antonio, Camille Andrieu, Céline Devisme, Serge Sancho, José-Alain Sahel, Christina Zeitz and Isabelle Audo
Int. J. Mol. Sci. 2022, 23(12), 6590; https://doi.org/10.3390/ijms23126590 - 13 Jun 2022
Cited by 4 | Viewed by 1784
Abstract
Inherited retinal diseases (IRD) are a group of heterogeneous disorders, most of which lead to blindness with limited therapeutic options. Pathogenic variants in RBP4, coding for a major blood carrier of retinol, retinol-binding protein 4, are responsible for a peculiar form of [...] Read more.
Inherited retinal diseases (IRD) are a group of heterogeneous disorders, most of which lead to blindness with limited therapeutic options. Pathogenic variants in RBP4, coding for a major blood carrier of retinol, retinol-binding protein 4, are responsible for a peculiar form of IRD. The aim of this study was to investigate if retinal function of an RBP4-related IRD patient can be improved by retinol administration. Our patient presented a peculiar white-dot retinopathy, reminiscent of vitamin A deficient retinopathy. Using a customized next generation sequencing (NGS) IRD panel we discovered a novel loss-of-function homozygous pathogenic variant in RBP4: c.255G >A, p.(Trp85*). Western blotting revealed the absence of RBP4 protein in the patient’s serum. Blood retinol levels were undetectable. The patient was put on a high-dose oral retinol regimen (50,000 UI twice a week). Subjective symptoms and retinal function markedly and sustainably improved at 5-months and 1-year follow-up. Here we show that this novel IRD case can be treated by oral retinol administration. Full article
(This article belongs to the Special Issue Retinal Degeneration—From Genetics to Therapy)
Show Figures

Figure 1

Back to TopTop