1
|
Jo DH, Jang H, Cho CS, Lee SJ, Heo JH, Kim JA, Kim SJ, Ryu W, Park CW, Kang BC, Gee HY, Sung YH, Kim HH, Kim JH. Intravitreal adenine base editing of RS1 improves vision in a preclinical mouse model of retinoschisis. Mol Ther 2025:S1525-0016(25)00295-3. [PMID: 40253584 DOI: 10.1016/j.ymthe.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/22/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Base editing offers high potential for treating genetic diseases, particularly those with limited treatment options. Retinoschisis, an X-linked retinal disease causing progressive vision loss, currently lacks effective therapies. We identified the c.422G>A (p.Arg141His) variant of the RS1 gene in six male patients with retinoschisis and generated a humanized mouse model harboring this variant, which mimicked the disease phenotype. By testing adenine base editors and single-guide RNAs, we identified an optimal combination of high editing efficiency and low bystander editing. Intravitreal injection of adeno-associated viral vectors encoding this adenine base editor achieved ∼40% editing efficiency in all retinal cells, restored retinal layer integrity, and preserved visual functions in 2-week-old male hemizygous mice. These mice exhibited retinal layer splitting at baseline, further validating the model. This study demonstrates a strategy for identifying effective base editing tools for clinical use through the preclinical evaluation of humanized mouse lines with patient-derived mutations and highlights their applicability in treating genetic diseases.
Collapse
Affiliation(s)
- Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Hyewon Jang
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chang Sik Cho
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Seok Jae Lee
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Ji Hwa Heo
- Department of Cell and Genetic Engineering and Convergence Medicine Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 03080, Republic of Korea
| | - Jung Ah Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Woo Choo Lee Institute for Precision Drug Development, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Se Jin Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Woo Choo Lee Institute for Precision Drug Development, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - WonHyoung Ryu
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Chan-Wook Park
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Byeong-Cheol Kang
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea; Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Woo Choo Lee Institute for Precision Drug Development, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young Hoon Sung
- Department of Cell and Genetic Engineering and Convergence Medicine Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 03080, Republic of Korea.
| | - Hyongbum Henry Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Woo Choo Lee Institute for Precision Drug Development, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Won-Sang Lee Institute for Hearing Loss, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jeong Hun Kim
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea; Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences & Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
2
|
Wei X, Ma S, Zhou Y, Cui H, Zhang S, Wang D, Fu Y, Li W, Han H, Li Y, Li W, Liu H, Sun Z, Han X, Zou X, Li H, Wang C, Sui R. A Novel AAV Capsid-Mediated RS1 Gene Therapy Restored Retinal Function to Wild-Type Levels in Rs1R213W Mouse Model. Invest Ophthalmol Vis Sci 2025; 66:37. [PMID: 40232710 PMCID: PMC12007669 DOI: 10.1167/iovs.66.4.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Purpose X-linked juvenile retinoschisis (XLRS) is a retinal disease caused by retinoschisin 1 (RS1) gene variants, potentially leading to severe visual impairment and blindness. This study aimed to develop a novel adeno-associated virus (AAV) serotype and evaluate its therapeutic potential in an XLRS mouse model. Methods A novel RS1 mouse model was established using the CRISPR-Cas9 gene editing system and underwent phenotypic characterization. AAV.IVT18-scRS/CMV-RS1, comprising a rationally designed novel capsid (AAV.IVT18) and an optimized RS1 gene expression cassette, was then constructed and delivered via intravitreal injection into mutant and wild-type (WT) mice at 3 to 4 weeks of age. Retinal structure, function, and inflammation levels were evaluated after treatment. Results A novel mouse model harboring the patient-derived RS1 missense variant R213W was generated, accurately recapitulating the human phenotype. We developed a novel AAV serotype, AAV.IVT18, which efficiently transfected photoreceptor and bipolar cells by intravitreal injection, and optimized the expression cassettes of RS1. AAV.IVT18-mediated expression of RS1 ameliorated retinoschisis in the mouse model and normalized the b/a ratio of the mouse electroretinogram (ERG). Remarkably, the ERG b-wave amplitudes of the treated groups began to increase at 8 weeks post-injection and recovered to the WT mouse level by 16 weeks of injection. Inflammatory activation in Rs1R213W mice was alleviated by treatment. Conclusions The novel AAV capsid-mediated RS1 gene therapy effectively improved retinal structure and function while downregulating inflammation in Rs1R213W mice. These results provide a robust foundation for future clinical trials on XLRS gene therapy, offering the potential to improve the vision and quality of life of patients.
Collapse
Affiliation(s)
- Xing Wei
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Sisi Ma
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Yunyu Zhou
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Haoliang Cui
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Shan Zhang
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Duanyang Wang
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Yingying Fu
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Wei Li
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Huihui Han
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Yamei Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wuyi Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Huixin Liu
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zixi Sun
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxu Han
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zou
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Cheng Wang
- Beijing InnoVec Biotherapeutics Inc., Beijing, China
| | - Ruifang Sui
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Lee SJ, Jiang H, Jeong HC, Jo DH, Song HB, Gee HY, Lee KH, Kim JH. Genotype-Phenotype Correlations in 83 Korean X-linked Retinoschisis Patients: Impact of Retinoschisin 1 Secretion Profiles on Clinical Phenotypes. Ophthalmol Retina 2025; 9:288-298. [PMID: 39293640 DOI: 10.1016/j.oret.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
PURPOSE To assess the correlation between genotype and phenotype severity in X-linked juvenile retinoschisis (XLRS) by examining clinical and genetic features of a cohort of Korean XLRS patients. DESIGN Retrospective, observational study. PARTICIPANTS Data from 83 consecutive male patients with molecularly confirmed XLRS were collected retrospectively. METHODS Clinical evaluation included best-corrected visual acuity (BCVA), fundus photography, spectral domain OCT (SD-OCT), and full-field electroretinography (ERG). MAIN OUTCOME MEASURES The phenotypic characteristics of a cohort of pediatric Korean patients with XLRS, based on mutation types (truncating vs. missense) and secretory profile (secretion vs. nonsecretion), were assessed. RESULTS A total of 166 eyes of 83 patients were included. The mean age at diagnosis was 6.1 ± 8.8 years (range, 0.5-20.7 years), with a mean follow-up time of 9.2 ± 7.0 years (range, 0.6-24.3 years). The BCVA at first and last examination ranged from light perception to 0.1 logarithm of the minimum angle of resolution (mean ± standard deviation, 0.75 ± 0.59 and 0.82 ± 0.65, respectively). No significant differences were observed between the truncating (0.71 ± 0.51 and 0.75 ± 0.44) and missense (0.77 ± 0.59 and 0.84 ± 0.66) variants (P = 0.678 and 0.551). Clinical parameters from fundus photography, SD-OCT, and ERG showed no differences. However, BCVA was better for the secretion group (0.51 ± 0.24 and 0.61 ± 0.30) than for the nonsecretion group (0.65 ± 0.71 and 0.87 ± 0.81), with a significant difference in the last BCVA (P = 0.021). OCT revealed a higher frequency of ellipsoid zone disruption in the nonsecretion group (P = 0.030), with no significant differences in other parameters. CONCLUSIONS The secretion profile of Retinoschisin 1 (RS1) could influence the severity of XLRS phenotypes. Patients with RS1-secreted mutants, particularly with intact octamerization, exhibit more homogeneous phenotypes and better visual acuity than the RS1-nonsecreted group. This data provide insights for studying genotype and phenotype correlations in both clinical and research fields. FINANCIAL DISCLOSURE(S) The authors have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Seok Jae Lee
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea; Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hui Jiang
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Chul Jeong
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea; Department of Anatomy & Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Beom Song
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ki Hwang Lee
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jeong Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea; Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Chien Y, Wu Y, Chen C, Yang Y, Ching L, Wang B, Chang W, Chiang I, Su P, Chen S, Lin W, Wang I, Lin T, Chen S, Chiou S. Identifying Multiomic Signatures of X-Linked Retinoschisis-Derived Retinal Organoids and Mice Harboring Patient-Specific Mutation Using Spatiotemporal Single-Cell Transcriptomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405818. [PMID: 39503290 PMCID: PMC11714187 DOI: 10.1002/advs.202405818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/26/2024] [Indexed: 11/08/2024]
Abstract
X-linked retinoschisis (XLRS) is an inherited retinal disorder with severe retinoschisis and visual impairments. Multiomics approaches integrate single-cell RNA-sequencing (scRNA-seq) and spatiotemporal transcriptomics (ST) offering potential for dissecting transcriptional networks and revealing cell-cell interactions involved in biomolecular pathomechanisms. Herein, a multimodal approach is demonstrated combining high-throughput scRNA-seq and ST to elucidate XLRS-specific transcriptomic signatures in two XLRS-like models with retinal splitting phenotypes, including genetically engineered (Rs1emR209C) mice and patient-derived retinal organoids harboring the same patient-specific p.R209C mutation. Through multiomics transcriptomic analysis, the endoplasmic reticulum (ER) stress/eukryotic initiation factor 2 (eIF2) signaling, mTOR pathway, and the regulation of eIF4 and p70S6K pathways are identified as chronically enriched and highly conserved disease pathways between two XLRS-like models. Western blots and proteomics analysis validate the occurrence of unfolded protein responses, chronic eIF2α signaling activation, and chronic ER stress-induced apoptosis. Furthermore, therapeutic targeting of the chronic ER stress/eIF2α pathway activation synergistically enhances the efficacy of AAV-mediated RS1 gene delivery, ultimately improving bipolar cell integrity, postsynaptic transmission, disorganized retinal architecture, and electrophysiological responses. Collectively, the complex transcriptomic signatures obtained from Rs1emR209C mice and patient-derived retinal organoids using the multiomics approach provide opportunities to unravel potential therapeutic targets for incurable retinal diseases, such as XLRS.
Collapse
Affiliation(s)
- Yueh Chien
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of PharmacologyCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - You‐Ren Wu
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of PharmacologyCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - Chih‐Ying Chen
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of PharmacologyCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - Yi‐Ping Yang
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of Food Safety and Health Risk AssessmentNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - Lo‐Jei Ching
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of Clinical MedicineSchool of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - Bo‐Xuan Wang
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of PharmacologyCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - Wei‐Chao Chang
- Center for Molecular MedicineChina Medical University HospitalTaichung40447Taiwan
| | - I‐Hsun Chiang
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
| | - Pong Su
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
| | - Shih‐Yu Chen
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
- Genome and Systems Biology Degree ProgramAcademia Sinica and National Taiwan UniversityTaipei10617Taiwan
| | - Wen‐Chang Lin
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
- Institute of Biomedical InformaticsNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - I‐Chieh Wang
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of PharmacologyCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
| | - Tai‐Chi Lin
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- School of MedicineCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
- Department of OphthalmologyTaipei Veterans General HospitalTaipei112201Taiwan
| | - Shih‐Jen Chen
- School of MedicineCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
- Department of OphthalmologyTaipei Veterans General HospitalTaipei112201Taiwan
| | - Shih‐Hwa Chiou
- Department of Medical ResearchTaipei Veterans General HospitalTaipei11217Taiwan
- Institute of PharmacologyCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipei11221Taiwan
- Department of OphthalmologyTaipei Veterans General HospitalTaipei112201Taiwan
- Genomics Research CenterAcademia SinicaTaipei11529Taiwan
| |
Collapse
|
5
|
Smit-McBride Z, Sun N, Thomas S, Cho IH, Stricklin RG, Sieving PA. Kir4.1 and Aqp4 Contribution to Schisis Cystic Water Accumulation and Clearance in the Rs1 Exon-1 Del XLRS Rat Model. Genes (Basel) 2024; 15:1583. [PMID: 39766850 PMCID: PMC11675908 DOI: 10.3390/genes15121583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVE The Rs1 exon-1-del rat (Rs1KO) XLRS model shows normal retinal development until postnatal day 12 (P12) when small cystic spaces start to form in the inner nuclear layer. These spaces enlarge rapidly, peak at P15, and then collapse by P19. METHODS We explored the possible involvement of Kir4.1 and Aqp4, the principal retina channels for water movement and homeostasis, along with Muller glia cells (MGCs), using semi-quantitative fluorescent immunohistochemistry at P7, P9, P12, and P30, in Rs1KO and WT littermates. RESULTS Kir4.1 expression was reduced in Rs1KO retinas at all the early time points-P7, P9, and P12-as the schisis cavities began to form; downregulation would reduce water egress from the retina. Aqp4 was upregulated at P30 in Rs1KO retinas during schisis cavity closure but not as cavities formed at P12. When examined by GFAP expression, MGCs were not activated at the preschisis P12 age but showed considerable GFAP expression at P30 following retinal cystic structural damage at P15, indicating that MGCs were activated during the period of retina water removal and cavity closure. CONCLUSIONS The study results implicate the downregulation of Kir4.1 in schisis formation and a role for both Kir4.1 and Aqp4 upregulation in subsequent schisis closure.
Collapse
Affiliation(s)
- Zeljka Smit-McBride
- Department of Ophthalmology, Eye Center, UC Davis School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Vitreoretinal Research Lab, UC Davis School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Ning Sun
- Department of Cell Biology & Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Serafina Thomas
- Department of Cell Biology & Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - In Hwan Cho
- Department of Ophthalmology, Eye Center, UC Davis School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea
| | - Robin G. Stricklin
- Department of Cell Biology & Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Paul A. Sieving
- Department of Ophthalmology, Eye Center, UC Davis School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Vitreoretinal Research Lab, UC Davis School of Medicine, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
6
|
Zhang L, Liu X, Sun L, Zhou X, Ke S, Ding X. Choroidal neovascularisation secondary toX-linked retinoschisis. Br J Ophthalmol 2024; 108:1564-1570. [PMID: 38811052 DOI: 10.1136/bjo-2023-324165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/27/2024] [Indexed: 05/31/2024]
Abstract
AIMS Choroidal neovascularisation (CNV) in patients with X-linked retinoschisis (XLRS) has been poorly documented. This study aims to investigate the prevalence and clinical characteristics of CNV in patients with XLRS, as well as analyse the preliminary genotype-phenotype correlation. METHODS A retrospective case series of patients with genetically confirmed XLRS was included. Demographic, clinical and genetic features were analysed, with a comparison between CNV and non-CNV eyes. RESULTS Among 185 eyes of 129 patients with XLRS, the prevalence of CNV was 8.1% (15/185). The mean diagnostic age of all patients with CNV is 5.1±2.56 years. CNV eyes exhibited a mean best-corrected visual acuity (BCVA) (logarithm of the minimal angle of resolution) of 1.37±0.74. All CNVs were classified as subretinal and active. Peripapillary CNVs accounted for 80.0% (12/15), while subfoveal CNVs accounted for 20.0% (3/15). In CNV eyes, the prevalence of macular atrophy (5/15, 33.3%, p=0.013) and bullous peripheral schisis (14/15, 93.3%, p=0.000) was higher compared with non-CNV eyes. Additionally, CNV eyes exhibited poorer integrity of the outer retina and BCVA (p=0.007) compared with non-CNV eyes. All 15 eyes with CNV underwent anti-vascular endothelial growth factor (anti-VEGF) therapy. Genotype analysis revealed that 7 of 10 patients (70.0%, 10 eyes) were predicted to have missense variants, while 3 of 10 patients (30.0%, 5 eyes) exhibited severe variants. CONCLUSIONS The prevalence of CNV in XLRS eyes was found to be 8.1%. All CNVs secondary to XLRS were active and classified as type 2. CNV eyes demonstrated poorer visual function and compromised retinal structures. Anti-VEGF therapy demonstrated effectiveness in treating XLRS-CNVs. No significant genotype-phenotype correlation was established.
Collapse
Affiliation(s)
- Linyan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Retina Division, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, People's Republic of China
| | - Xinyu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Retina Division, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, People's Republic of China
| | - Limei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Retina Division, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, People's Republic of China
| | - Xiaodi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Retina Division, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, People's Republic of China
| | - Shuya Ke
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Retina Division, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, People's Republic of China
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Retina Division, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, People's Republic of China
| |
Collapse
|
7
|
Alsalloum A, Gornostal E, Mingaleva N, Pavlov R, Kuznetsova E, Antonova E, Nadzhafova A, Kolotova D, Kadyshev V, Mityaeva O, Volchkov P. A Comparative Analysis of Models for AAV-Mediated Gene Therapy for Inherited Retinal Diseases. Cells 2024; 13:1706. [PMID: 39451224 PMCID: PMC11506034 DOI: 10.3390/cells13201706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Inherited retinal diseases (IRDs) represent a diverse group of genetic disorders leading to progressive degeneration of the retina due to mutations in over 280 genes. This review focuses on the various methodologies for the preclinical characterization and evaluation of adeno-associated virus (AAV)-mediated gene therapy as a potential treatment option for IRDs, particularly focusing on gene therapies targeting mutations, such as those in the RPE65 and FAM161A genes. AAV vectors, such as AAV2 and AAV5, have been utilized to deliver therapeutic genes, showing promise in preserving vision and enhancing photoreceptor function in animal models. Despite their advantages-including high production efficiency, low pathogenicity, and minimal immunogenicity-AAV-mediated therapies face limitations such as immune responses beyond the retina, vector size constraints, and challenges in large-scale manufacturing. This review systematically compares different experimental models used to investigate AAV-mediated therapies, such as mouse models, human retinal explants (HREs), and induced pluripotent stem cell (iPSC)-derived retinal organoids. Mouse models are advantageous for genetic manipulation and detailed investigations of disease mechanisms; however, anatomical differences between mice and humans may limit the translational applicability of results. HREs offer valuable insights into human retinal pathophysiology but face challenges such as tissue degradation and lack of systemic physiological effects. Retinal organoids, on the other hand, provide a robust platform that closely mimics human retinal development, thereby enabling more comprehensive studies on disease mechanisms and therapeutic strategies, including AAV-based interventions. Specific outcomes targeted in these studies include vision preservation and functional improvements of retinas damaged by genetic mutations. This review highlights the strengths and weaknesses of each experimental model and advocates for their combined use in developing targeted gene therapies for IRDs. As research advances, optimizing AAV vector design and delivery methods will be critical for enhancing therapeutic efficacy and improving clinical outcomes for patients with IRDs.
Collapse
Affiliation(s)
- Almaqdad Alsalloum
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.V.)
| | | | - Natalia Mingaleva
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Roman Pavlov
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | | | - Ekaterina Antonova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Aygun Nadzhafova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Daria Kolotova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | | | - Olga Mityaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.V.)
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Pavel Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.V.)
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
- Moscow Clinical Scientific Center N.A. A.S. Loginov, 111123 Moscow, Russia
| |
Collapse
|
8
|
Duan C, Ding C, Sun X, Mao S, Liang Y, Liu X, Ding X, Chen J, Tang S. Retinal organoids with X-linked retinoschisis RS1 (E72K) mutation exhibit a photoreceptor developmental delay and are rescued by gene augmentation therapy. Stem Cell Res Ther 2024; 15:152. [PMID: 38816767 PMCID: PMC11140964 DOI: 10.1186/s13287-024-03767-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND X-linked juvenile retinoschisis (XLRS) is an inherited disease caused by RS1 gene mutation, which leads to retinal splitting and visual impairment. The mechanism of RS1-associated retinal degeneration is not fully understood. Besides, animal models of XLRS have limitations in the study of XLRS. Here, we used human induced pluripotent stem cell (hiPSC)-derived retinal organoids (ROs) to investigate the disease mechanisms and potential treatments for XLRS. METHODS hiPSCs reprogrammed from peripheral blood mononuclear cells of two RS1 mutant (E72K) XLRS patients were differentiated into ROs. Subsequently, we explored whether RS1 mutation could affect RO development and explore the effectiveness of RS1 gene augmentation therapy. RESULTS ROs derived from RS1 (E72K) mutation hiPSCs exhibited a developmental delay in the photoreceptor, retinoschisin (RS1) deficiency, and altered spontaneous activity compared with control ROs. Furthermore, the delays in development were associated with decreased expression of rod-specific precursor markers (NRL) and photoreceptor-specific markers (RCVRN). Adeno-associated virus (AAV)-mediated gene augmentation with RS1 at the photoreceptor immature stage rescued the rod photoreceptor developmental delay in ROs with the RS1 (E72K) mutation. CONCLUSIONS The RS1 (E72K) mutation results in the photoreceptor development delay in ROs and can be partially rescued by the RS1 gene augmentation therapy.
Collapse
Affiliation(s)
- Chunwen Duan
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan, China
| | | | - Xihao Sun
- Aier Eye Institute, Changsha, Hunan, China
| | - Shengru Mao
- Aier Eye Institute, Changsha, Hunan, China
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | - Xinyu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Jiansu Chen
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China.
- Aier Eye Institute, Changsha, Hunan, China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, Guangdong, China.
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China.
- Aier Eye Institute, Changsha, Hunan, China.
- Guangzhou Aier Eye Hospital, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Tarchick MJ, Beight C, Bonezzi PB, Peachey NS, Renna JM. Photoreceptor deficits appear at eye opening in Rs1 mutant mouse models of X-linked retinoschisis. Exp Eye Res 2024; 242:109872. [PMID: 38514024 DOI: 10.1016/j.exer.2024.109872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
X-linked retinoschisis (XLRS) is an early onset degenerative retinal disease characterized by cystic lesions in the middle layers of the retina. These structural changes are accompanied by a loss of visual acuity and decreased contrast sensitivity. XLRS is caused by mutations in the gene Rs1 which encodes the secreted protein Retinoschisin 1. Young Rs1-mutant mouse models develop key hallmarks of XLRS including intraretinal schisis and abnormal electroretinograms. The electroretinogram (ERG) comprises activity of multiple cellular generators, and it is not known how and when each of these is impacted in Rs1 mutant mice. Here we use an ex vivo ERG system and pharmacological blockade to determine how ERG components generated by photoreceptors, ON-bipolar, and Müller glial cells are impacted in Rs1 mutants and to determine the time course of these changes. We report that ERG abnormalities begin near eye-opening and that all ERG components are involved.
Collapse
Affiliation(s)
| | - Craig Beight
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Paul B Bonezzi
- Department of Biology, University of Akron, Akron, OH, USA
| | - Neal S Peachey
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, 44106, USA; Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jordan M Renna
- Department of Biology, University of Akron, Akron, OH, USA.
| |
Collapse
|
10
|
van der Veen I, Heredero Berzal A, Koster C, ten Asbroek ALMA, Bergen AA, Boon CJF. The Road towards Gene Therapy for X-Linked Juvenile Retinoschisis: A Systematic Review of Preclinical Gene Therapy in Cell-Based and Rodent Models of XLRS. Int J Mol Sci 2024; 25:1267. [PMID: 38279267 PMCID: PMC10816913 DOI: 10.3390/ijms25021267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is an early-onset progressive inherited retinopathy affecting males. It is characterized by abnormalities in the macula, with formation of cystoid retinal cavities, frequently accompanied by splitting of the retinal layers, impaired synaptic transmission of visual signals, and associated loss of visual acuity. XLRS is caused by loss-of-function mutations in the retinoschisin gene located on the X chromosome (RS1, MIM 30083). While proof-of-concept studies for gene augmentation therapy have been promising in in vitro and rodent models, clinical trials in XLRS patients have not been successful thus far. We performed a systematic literature investigation using search strings related to XLRS and gene therapy in in vivo and in vitro models. Three rounds of screening (title/abstract, full text and qualitative) were performed by two independent reviewers until consensus was reached. Characteristics related to study design and intervention were extracted from all studies. Results were divided into studies using (1) viral and (2) non-viral therapies. All in vivo rodent studies that used viral vectors were assessed for quality and risk of bias using the SYRCLE's risk-of-bias tool. Studies using alternative and non-viral delivery techniques, either in vivo or in vitro, were extracted and reviewed qualitatively, given the diverse and dispersed nature of the information. For in-depth analysis of in vivo studies using viral vectors, outcome data for optical coherence tomography (OCT), immunohistopathology and electroretinography (ERG) were extracted. Meta-analyses were performed on the effect of recombinant adeno-associated viral vector (AAV)-mediated gene augmentation therapies on a- and b-wave amplitude as well as the ratio between b- and a-wave amplitudes (b/a-ratio) extracted from ERG data. Subgroup analyses and meta-regression were performed for model, dose, age at injection, follow-up time point and delivery method. Between-study heterogeneity was assessed with a Chi-square test of homogeneity (I2). We identified 25 studies that target RS1 and met our search string. A total of 19 of these studies reported rodent viral methods in vivo. Six of the 25 studies used non-viral or alternative delivery methods, either in vitro or in vivo. Of these, five studies described non-viral methods and one study described an alternative delivery method. The 19 aforementioned in vivo studies were assessed for risk of bias and quality assessments and showed inconsistency in reporting. This resulted in an unclear risk of bias in most included studies. All 19 studies used AAVs to deliver intact human or murine RS1 in rodent models for XLRS. Meta-analyses of a-wave amplitude, b-wave amplitude, and b/a-ratio showed that, overall, AAV-mediated gene augmentation therapy significantly ameliorated the disease phenotype on these parameters. Subgroup analyses and meta-regression showed significant correlations between b-wave amplitude effect size and dose, although between-study heterogeneity was high. This systematic review reiterates the high potential for gene therapy in XLRS, while highlighting the importance of careful preclinical study design and reporting. The establishment of a systematic approach in these studies is essential to effectively translate this knowledge into novel and improved treatment alternatives.
Collapse
Affiliation(s)
- Isa van der Veen
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Céline Koster
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Anneloor L. M. A. ten Asbroek
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Arthur A. Bergen
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Ophthalmology, Leiden University Medical Center, Leiden University, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
11
|
Ku CA, Wei LW, Sieving PA. X-Linked Retinoschisis. Cold Spring Harb Perspect Med 2023; 13:a041288. [PMID: 36690462 PMCID: PMC10513161 DOI: 10.1101/cshperspect.a041288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
X-linked retinoschisis (XLRS) is an inherited vitreoretinal dystrophy causing visual impairment in males starting at a young age with an estimated prevalence of 1:5000 to 1:25,000. The condition was first observed in two affected brothers by Josef Haas in 1898 and is clinically diagnosed by characteristic intraretinal cysts arranged in a petaloid "spoke-wheel" pattern centered in the macula. When clinical electroretinogram (ERG) testing began in the 1960s, XLRS was noted to have a characteristic reduction of the dark-adapted b-wave amplitude despite normal or usually nearly normal a-wave amplitudes, which became known as the "electronegative ERG response" of XLRS disease. The causative gene, RS1, was identified on the X-chromosome in 1997 and led to understanding the molecular and cellular basis of the condition, discerning the structure and function of the retinoschisin protein, and generating XLRS murine models. Along with parallel development of gene delivery vectors suitable for targeting retinal diseases, successful gene augmentation therapy was demonstrated by rescuing the XLRS phenotype in mouse. Two human phase I/II therapeutic XLRS gene augmentation studies were initiated; and although these did not yield definitive improvement in visual function, they gave significant new knowledge and experience, which positions the field for further near-term clinical testing with enhanced, next-generation gene therapy for XLRS patients.
Collapse
Affiliation(s)
- Cristy A Ku
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, California 95817, USA
| | - Lisa W Wei
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, NIH Office of Biodefense, Research Resources and Translational Research/Vaccine Section, Bethesda, Maryland 20892, USA
| | - Paul A Sieving
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, California 95817, USA
| |
Collapse
|
12
|
Sin TN, Kim S, Li Y, Wang J, Chen R, Chung SH, Kim S, Casanova MI, Park S, Smit-McBride Z, Sun N, Pomerantz O, Roberts JA, Guan B, Hufnagel RB, Moshiri A, Thomasy SM, Sieving PA, Yiu G. A Spontaneous Nonhuman Primate Model of Myopic Foveoschisis. Invest Ophthalmol Vis Sci 2023; 64:18. [PMID: 36689233 PMCID: PMC9896856 DOI: 10.1167/iovs.64.1.18] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023] Open
Abstract
Purpose Foveoschisis involves the pathologic splitting of retinal layers at the fovea, which may occur congenitally in X-linked retinoschisis (XLRS) or as an acquired complication of myopia. XLRS is attributed to functional loss of the retinal adhesion protein retinoschisin 1 (RS1), but the pathophysiology of myopic foveoschisis is unclear due to the lack of animal models. Here, we characterized a novel nonhuman primate model of myopic foveoschisis through clinical examination and multimodal imaging followed by morphologic, cellular, and transcriptional profiling of retinal tissues and genetic analysis. Methods We identified a rhesus macaque with behavioral and anatomic features of myopic foveoschisis, and monitored disease progression over 14 months by fundus photography, fluorescein angiography, and optical coherence tomography (OCT). After necropsy, we evaluated anatomic and cellular changes by immunohistochemistry and transcriptomic changes using single-nuclei RNA-sequencing (snRNA-seq). Finally, we performed Sanger and whole exome sequencing with focus on the RS1 gene. Results Affected eyes demonstrated posterior hyaloid traction and progressive splitting of the outer plexiform layer on OCT. Immunohistochemistry showed increased GFAP expression in Müller glia and loss of ramified Iba-1+ microglia, suggesting macro- and microglial activation with minimal photoreceptor alterations. SnRNA-seq revealed gene expression changes predominantly in cones and retinal ganglion cells involving chromatin modification, suggestive of cellular stress at the fovea. No defects in the RS1 gene or its expression were detected. Conclusions This nonhuman primate model of foveoschisis reveals insights into how acquired myopic traction leads to phenotypically similar morphologic and cellular changes as congenital XLRS without alterations in RS1.
Collapse
Affiliation(s)
- Tzu-Ni Sin
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| | - Sangbae Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Jun Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Sook Hyun Chung
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| | - Soohyun Kim
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
- Department of Surgical & Radiological Sciences, University of California Davis, Davis, California, United States
| | - M. Isabel Casanova
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
- Department of Surgical & Radiological Sciences, University of California Davis, Davis, California, United States
| | - Sangwan Park
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
- Department of Surgical & Radiological Sciences, University of California Davis, Davis, California, United States
| | - Zeljka Smit-McBride
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| | - Ning Sun
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| | - Ori Pomerantz
- California National Primate Research Center, Davis, California, United States
| | - Jeffrey A. Roberts
- California National Primate Research Center, Davis, California, United States
| | - Bin Guan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robert B. Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| | - Sara M. Thomasy
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
- Department of Surgical & Radiological Sciences, University of California Davis, Davis, California, United States
| | - Paul A. Sieving
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, California, United States
| |
Collapse
|
13
|
Lorenz B, Tavares J, van den Born LI, Marques JP, Pilotto E, Stingl K, Charbel Issa P, Leroux D, Dollfus H, Scholl HPN. Current Management of Inherited Retinal Degeneration Patients in Europe: Results of a 2-Year Follow-Up Multinational Survey by the European Vision Institute Clinical Research Network - EVICR.net. Ophthalmic Res 2023; 66:550-568. [PMID: 36592621 DOI: 10.1159/000528716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/05/2022] [Indexed: 01/03/2023]
Abstract
INTRODUCTION An increasing number of gene-specific therapies are being developed for inherited retinal degenerations (IRDs). Identification of well-characterized patients is an emerging need. We conducted the second multinational survey among the www.evicr.net and ERN-EYE members to understand the management and treatment of IRDs in Europe and compared it to the 2019 survey. METHODS An electronic survey questionnaire was developed and sent to 124 clinical centers (25 countries) by June/July 2021. Statistical analysis was performed with Excel and R. RESULTS The overall response rate was 44% but varied among countries. Only 9% of responding centers do not see IRD patients (2019 survey 14%), 42% follow at least 200 patients per year, 18% follow 500-999, and 2% more than 1,000. Databases exist in 86% of the centers (local 86%; national web based 12%). IRD patients are referred to www.evicr.net and ERN-EYE centers mainly by general ophthalmologists, patient self-referral, or medical retina specialists. Most IRD patients are first seen as adults. Signs and symptoms depend on age of onset: in infancy, nystagmus; at older age, night blindness and reduced visual field; reduced visual acuity is described at any age. Comprehensive ophthalmic examination always includes visual acuity and almost always visual field multimodal retinal imaging, electrophysiology, color vision testing, and refraction. Identification of genotypes is successful in 72% of centers in 40-80% of cases (2019 survey 69% of centers). The time for confirmation of the genetic diagnosis varies from 2-4 weeks to 24 months (2019 survey >4 weeks ≤10 years). Genetic testing is covered by public health service in 83%, private health insurance in 29%, research funds in 24%; 5% do not have access to genetic testing (2019 survey 15%). The most striking result is the high increase in the involvement of centers in natural history and gene therapy trials that more than doubled for the latter. DISCUSSION This second multinational survey on management of IRDs in Europe highlights persistent important differences in the number of IRD patients managed per center, comparable diagnostic work-up, and increasing genotyping in diagnostic laboratories. The important increase in involvement of centers in natural history and gene therapy trials reflects the rapidly evolving field of gene therapy development. The survey provides important follow-up data for researchers, clinicians, caregivers, patient advocate groups, pharmaceutical companies, and investors.
Collapse
Affiliation(s)
- Birgit Lorenz
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Department of Ophthalmology, Justus-Liebig-University, Giessen, Germany
| | - Joana Tavares
- AIBILI - Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal,
| | - L Ingeborgh van den Born
- Rotterdam Eye Hospital, Rotterdam, The Netherlands
- Rotterdam Ophthalmic Institute, Rotterdam, The Netherlands
| | - João Pedro Marques
- AIBILI - Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Department of Ophthalmology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Katarina Stingl
- Center for Ophthalmology, University Eye Hospital, University of Tuebingen, Tuebingen, Germany
| | - Peter Charbel Issa
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Dorothée Leroux
- CARGO & ERN-EYE management, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Hélène Dollfus
- CARGO & ERN-EYE management, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Hendrik P N Scholl
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| |
Collapse
|
14
|
Ye EA, Zeng Y, Thomas S, Sun N, Smit-McBride Z, Sieving PA. XLRS Rat with Rs1 -/Y Exon-1-Del Shows Failure of Early Postnatal Outer Retina Development. Genes (Basel) 2022; 13:1995. [PMID: 36360232 PMCID: PMC9690472 DOI: 10.3390/genes13111995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 05/19/2024] Open
Abstract
We generated a Long Evans transgenic rat with targeted deletion of the whole Rs1 exon-1 and evaluated the pathological retinal phenotype of this Rs1-/Y rat model of X-linked retinoschisis (XLRS). The Rs1-/Y rat exhibited very early onset and rapidly progressive photoreceptor degeneration. The outer limiting membrane (OLM) was disrupted and discontinuous by post-natal day (P15) and allowed photoreceptor nuclei to dislocate from the outer nuclear layers (ONL) into the sub-retinal side of the OLM. Dark-adapted electroretinogram (ERG) a-wave and b-wave amplitudes were considerably reduced to only 20-25% of WT by P17. Microglia and Müller glial showed cell marker activation by P7. Intravitreal application of AAV8-RS1 at P5-6 induced RS1 expression by P15 and rescued the inner nuclear layer (INL) and outer plexiform layer (OPL) cavity formation otherwise present at P15, and the outer-retinal structure was less disrupted. This Rs1-/Y exon-1-del rat model displays substantially faster rod cell loss compared to the exon-1-del Rs1-KO mouse. Most unexpected was the rapid appearance of schisis cavities between P7 and P15, and then cavities rapidly disappeared by P21/P30. The rat model provides clues on the molecular and cellular mechanisms underlying XLRS pathology in this model and points to a substantial and early changes to normal retinal development.
Collapse
Affiliation(s)
- Eun-Ah Ye
- Department of Human Anatomy and Cell Biology, University of California Davis, Davis, CA 95616, USA
| | - Yong Zeng
- National Eye Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - Serafina Thomas
- Department of Human Anatomy and Cell Biology, University of California Davis, Davis, CA 95616, USA
| | - Ning Sun
- Department of Human Anatomy and Cell Biology, University of California Davis, Davis, CA 95616, USA
| | - Zeljka Smit-McBride
- Department of Ophthalmology, University of California Davis, Davis, CA 95616, USA
| | - Paul A. Sieving
- Department of Ophthalmology, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
15
|
Vijayasarathy C, Zeng Y, Marangoni D, Dong L, Pan ZH, Simpson EM, Fariss RN, Sieving PA. Targeted Expression of Retinoschisin by Retinal Bipolar Cells in XLRS Promotes Resolution of Retinoschisis Cysts Sans RS1 From Photoreceptors. Invest Ophthalmol Vis Sci 2022; 63:8. [PMID: 36227606 PMCID: PMC9583743 DOI: 10.1167/iovs.63.11.8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/18/2022] [Indexed: 01/14/2023] Open
Abstract
Purpose Loss of retinoschisin (RS1) function underlies X-linked retinoschisis (XLRS) pathology. In the retina, both photoreceptor inner segments and bipolar cells express RS1. However, the loss of RS1 function causes schisis primarily in the inner retina. To understand these cell type-specific phenotypes, we decoupled RS1 effects in bipolar cells from that in photoreceptors. Methods Bipolar cell transgene RS1 expression was achieved using two inner retina-specific promoters: (1) a minimal promoter engineered from glutamate receptor, metabotropic glutamate receptor 6 gene (mini-mGluR6/ Grm6) and (2) MiniPromoter (Ple155). Adeno-associated virus vectors encoding RS1 gene under either the mini-mGluR6 or Ple-155 promoter were delivered to the XLRS mouse retina through intravitreal or subretinal injection on postnatal day 14. Retinal structure and function were assessed 5 weeks later: immunohistochemistry for morphological characterization, optical coherence tomography and electroretinography (ERG) for structural and functional evaluation. Results Immunohistochemical analysis of RS1expression showed that expression with the MiniPromoter (Ple155) was heavily enriched in bipolar cells. Despite variations in vector penetrance and gene transfer efficiency across the injected retinas, those retinal areas with robust bipolar cell RS1 expression showed tightly packed bipolar cells with fewer cavities and marked improvement in inner retinal structure and synaptic function as judged by optical coherence tomography and electroretinography, respectively. Conclusions These results demonstrate that RS1 gene expression primarily in bipolar cells of the XLRS mouse retina, independent of photoreceptor expression, can ameliorate retinoschisis structural pathology and provide further evidence of RS1 role in cell adhesion.
Collapse
Affiliation(s)
- Camasamudram Vijayasarathy
- Section for Translational Research in Retinal and Macular Degeneration, National Institutes of Health, Bethesda, Maryland, United States
| | - Yong Zeng
- Section for Translational Research in Retinal and Macular Degeneration, National Institutes of Health, Bethesda, Maryland, United States
| | - Dario Marangoni
- Section for Translational Research in Retinal and Macular Degeneration, National Institutes of Health, Bethesda, Maryland, United States
| | - Lijin Dong
- Genetic Engineering Facility, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Zhuo-Hua Pan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Elizabeth M. Simpson
- Centre for Molecular Medicine and Therapeutics at BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert N. Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Paul A. Sieving
- Section for Translational Research in Retinal and Macular Degeneration, National Institutes of Health, Bethesda, Maryland, United States
- Center for Ocular Regenerative Therapy, Department of Ophthalmology, University of California Davis, United States
| |
Collapse
|
16
|
Retinal Proteomic Alterations and Combined Transcriptomic-Proteomic Analysis in the Early Stages of Progression of a Mouse Model of X-Linked Retinoschisis. Cells 2022; 11:cells11142150. [PMID: 35883593 PMCID: PMC9321393 DOI: 10.3390/cells11142150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
X-linked retinoschisis (XLRS) is among the most commonly inherited degenerative retinopathies. XLRS is caused by functional impairment of RS1. However, the molecular mechanisms underlying RS1 malfunction remain largely uncharacterized. Here, we performed a data-independent acquisition-mass spectrometry-based proteomic analysis in RS1-null mouse retina with different postal days (Ps), including the onset (P15) and early progression stage (P56). Gene set enrichment analysis showed that type I interferon-mediated signaling was upregulated and photoreceptor proteins responsible for detection of light stimuli were downregulated at P15. Positive regulation of Tor signaling was downregulated and nuclear transcribed mRNA catabolic process nonsense-mediated decay was upregulated at P56. Moreover, the differentially expressed proteins at P15 were enriched in metabolism of RNA and RNA destabilization. A broader subcellular localization distribution and enriched proteins in visual perception and phototransduction were evident at P56. Combined transcriptomic-proteomic analysis revealed that functional impairments, including detection of visible light, visual perception, and visual phototransduction, occurred at P21 and continued until P56. Our work provides insights into the molecular mechanisms underlying the onset and progression of an XLRS mouse model during the early stages, thus enhancing the understanding of the mechanism of XLRS.
Collapse
|
17
|
Liu M, Liu J, Wang W, Liu G, Jin X, Lei B. Longitudinal Photoreceptor Phenotype Observation and Therapeutic Evaluation of a Carbonic Anhydrase Inhibitor in a X-Linked Retinoschisis Mouse Model. Front Med (Lausanne) 2022; 9:886947. [PMID: 35836954 PMCID: PMC9273824 DOI: 10.3389/fmed.2022.886947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose To study the long-term photoreceptor changes and to evaluate the effects of topical application of a carbonic anhydrase inhibitor (CAI) in a mouse model of X-linked retinoschisis (XLRS). Methods Conventional electroretinograms (ERGs) and dark-adapted 10-Hz flicker ERGs were recorded in control and Rs1−/Y mice generated with CRISPR/Cas9. ON-pathway blocker 2-amino-4-phosphobutyric acid (APB) was injected intravitreally. Morphology was evaluated with histology and optical coherence tomography (OCT). Mice were treated with a CAI inhibitor brinzolamide eye drops (10 mg/ml) three times a day for 3 months. OCT and ERG findings at 1, 4, and 10 months were analyzed. Results Negative ERGs and retinal cavities were evident in Rs1−/Y mice. Both a-wave and b-wave amplitudes decreased with age when compared with age-matched controls. The APB-isolated a-wave (a′) amplitudes of Rs1−/Y mice were reduced in all age groups. In dark-adapted 10-Hz flicker ERG, the amplitude-intensity curve of Rs1−/Y mice shifted down. The thickness of ONL and IS/OS decreased in Rs1−/Y mice. CAI reduced the splitting retinal cavities but didn't affect the ERG. Conclusions In addition to post receptoral impairments, photoreceptor cells underwent progressive dysfunction since early age in Rs1−/Y mice. Long-term CAI treatment improved the shrinkage of the splitting retinal cavity, while no functional improvement was observed.
Collapse
Affiliation(s)
- Meng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingyang Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Weiping Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Guangming Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiuxiu Jin
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Bo Lei
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- *Correspondence: Bo Lei ; orcid.org/0000-0002-5497-0905
| |
Collapse
|
18
|
Xiang L, Zhang J, Rao FQ, Yang QL, Zeng HY, Huang SH, Xie ZX, Lv JN, Lin D, Chen XJ, Wu KC, Lu F, Huang XF, Chen Q. Depletion of miR-96 Delays, But Does Not Arrest, Photoreceptor Development in Mice. Invest Ophthalmol Vis Sci 2022; 63:24. [PMID: 35481839 PMCID: PMC9055555 DOI: 10.1167/iovs.63.4.24] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Abundant retinal microRNA-183 cluster (miR-183C) has been reported to be a key player in photoreceptor development and functionality in mice. However, whether there is a protagonist in this cluster remains unclear. Here, we used a mutant mouse model to study the role of miR-96, a member of miR-183C, in photoreceptor development and functionality. Methods The mature miR-96 sequence was removed using the CRISPR/Cas9 genome-editing system. Electroretinogram (ERG) and optical coherence tomography (OCT) investigated the changes in structure and function in mouse retinas. Immunostaining determined the localization and morphology of the retinal cells. RNA sequencing was conducted to observe retinal transcription alterations. Results The miR-96 mutant mice exhibited cone developmental delay, as occurs in miR-183/96 double knockout mice. Immunostaining of cone-specific marker genes revealed cone nucleus mislocalization and exiguous Opn1mw/Opn1sw in the mutant (MT) mouse outer segments at postnatal day 10. Interestingly, this phenomenon could be relieved in the adult stages. Transcriptome analysis revealed activation of microtubule-, actin filament–, and cilia-related pathways, further supporting the findings. Based on ERG and OCT results at different ages, the MT mice displayed developmental delay not only in cones but also in rods. In addition, a group of miR-96 potential direct and indirect target genes was summarized for interpretation and further studies of miR-96–related retinal developmental defects. Conclusions Depletion of miR-96 delayed but did not arrest photoreceptor development in mice. This miRNA is indispensable for mouse photoreceptor maturation, especially for cones.
Collapse
Affiliation(s)
- Lue Xiang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Juan Zhang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Feng-Qin Rao
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, China
| | - Qiao-Li Yang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui-Yi Zeng
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Sheng-Hai Huang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhen-Xiang Xie
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ji-Neng Lv
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Dan Lin
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xue-Jiao Chen
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Kun-Chao Wu
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fan Lu
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| | - Xiu-Feng Huang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Chen
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou, China
| |
Collapse
|
19
|
Guo Q, Li Y, Li J, You Y, Liu C, Chen K, Li S, Lei B. Phenotype Heterogeneity and the Association Between Visual Acuity and Outer Retinal Structure in a Cohort of Chinese X-Linked Juvenile Retinoschisis Patients. Front Genet 2022; 13:832814. [PMID: 35309139 PMCID: PMC8931683 DOI: 10.3389/fgene.2022.832814] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/03/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose: X-linked juvenile retinoschisis (XLRS), caused by mutations in the RS1 gene, is an X-linked recessive inherited disease that typically involves both eyes in the first 2 decades of life. Recently, the phenotype heterogeneity of this condition has drawn increasing attention. We reported various phenotypes caused by RS1 gene mutations in eleven patients from ten Chinese families. Methods: Data on the medical history of the patients from ten Han families of central China were collected. Ophthalmic examinations including best-corrected visual acuity (BCVA), fundus photography, ultra-wide-angle sweep source optical coherence tomography (SS-OCT), and electroretinography (ERG) were performed. Adaptive optics (AO) images were acquired to evaluate the cone photoreceptor mosaic when applicable. Venous blood of the probands and their family members was collected, and DNA was subjected to sequencing based on next-generation sequencing with a custom-designed targeted gene panel PS400 for inherited retinal diseases. Validation was performed by Sanger sequencing and cosegregation. Pathogenicity was determined in accordance with the American College of Medical Genetics and Genomics (ACMG) guidelines. Results: Ten RS1 mutations, including eight missense mutations and two terminator mutations, were identified in 10 XLRS families. c.657C > A (p.C219X) was a novel mutation in this cohort. These patients showed a variety of clinical phenotypes, including fovea schisis, bullous retinoschisis, and macular or peripheral atrophy. Fifteen eyes of eight patients exhibited macular retinoschisis, and twelve eyes of seven patients exhibited peripheral retinoschisis. In addition, three patients showed asymmetrical fundus manifestations. Of importance, three patients without macular retinoschisis were misdiagnosed until genetic testing results were obtained. AO showed a decrease in cone density and loss of regularity in the cystic schisis macular of XLRS. Furthermore, the BCVA was associated with the photoreceptor inner segment and outer segment (IS/OS) thickness. Conclusion: With complicated clinical manifestations, a considerable portion of XLRS patients may present various phenotypes. It should be noted that asymmetry in fundus appearance in both eyes could lead to misdiagnosis easily. Thus, genetic testing is crucial for making a final diagnosis in those patients who are suspected of having amblyopia, bilateral or unilateral macular atrophy, or conditions presenting an asymmetric fundus appearance. In addition, the residual cone photoreceptor structure was critical for the maintenance of useful vision.
Collapse
Affiliation(s)
- Qingge Guo
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya Li
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiarui Li
- School of Medicine, People’s Hospital of Henan University, Henan University, Zhengzhou, China
| | - Ya You
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Changgeng Liu
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Kang Chen
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuyin Li
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Lei
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Henan Clinical Research Center for Ocular Diseases, People’s Hospital of Zhengzhou University, Zhengzhou, China
- School of Medicine, People’s Hospital of Henan University, Henan University, Zhengzhou, China
- *Correspondence: Bo Lei,
| |
Collapse
|