1
|
Velasco S, Gallego I, Olivares-González L, Puras G, Castro MC, Salom D, Pedraz JL, Rodrigo R. Noninvasive ocular delivery of adalimumab-loaded nanostructured lipid carriers for targeted retinitis pigmentosa therapy. Biomed Pharmacother 2025; 185:117962. [PMID: 40073744 DOI: 10.1016/j.biopha.2025.117962] [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: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Retinitis pigmentosa is a genetically heterogeneous retinal degeneration process. There is hardly any treatment available. It is associated with extensive chronic inflammation and the release of proinflammatory cytokines such as TNFα. The blockade of TNFα through systemic or intraocular routes slows retinal degeneration. They are invasive routes with possible side effects. Herein, we propose a noninvasive approach to address the inflammatory component of retinitis pigmentosa. This approach is based on the development of eye drops of nanostructured lipid carriers (NLCs) loaded with the monoclonal antibody against TNFα, adalimumab (ADA). We physicochemically characterized NLC-ADA. We evaluated retinal and corneal toxicity; corneal permeation; diffusion to the retina; and effects on retinal dysfunction, degeneration and inflammation. These results prove that NLC-ADA eye drops exhibit excellent corneal permeation, no toxicity and high retinal distribution in mice. These compounds improve retinal function, reduce retinal degeneration and ameliorate the inflammatory process. In particular, NLC-ADA eye drops reduce M1 microglial activation, macrophage infiltration and the levels of some components of the NLRP3 inflammasome in rd10 mice, a model of retinitis pigmentosa. This strategy offers a noninvasive route that circumvents the bloodretinal barrier in a safe and efficient manner. Hence, this approach could offer a promising therapeutic option for treating retinitis pigmentosa regardless of genetic defects. This approach could be useful for other inflammation-related retinal diseases.
Collapse
Affiliation(s)
- Sheyla Velasco
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, Valencia 46012, Spain; Joint Research Unit on Rare Diseases CIPF-Health Research Institute Hospital La Fe (IIS-La Fe), Valencia 46026, Spain.
| | - Idoia Gallego
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Monforte de Lemos, 3-5. Pabellón 11, Madrid 28029, Spain; Bioaraba, NanoBioCel Research Group, Jose Atxotegi Kalea, s/n, Txagorritxu, Vitoria-Gasteiz 01009, Spain.
| | - Lorena Olivares-González
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, Valencia 46012, Spain; Joint Research Unit on Rare Diseases CIPF-Health Research Institute Hospital La Fe (IIS-La Fe), Valencia 46026, Spain.
| | - Gustavo Puras
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Monforte de Lemos, 3-5. Pabellón 11, Madrid 28029, Spain; Bioaraba, NanoBioCel Research Group, Jose Atxotegi Kalea, s/n, Txagorritxu, Vitoria-Gasteiz 01009, Spain.
| | - Ma Carmen Castro
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, Valencia 46012, Spain; Joint Research Unit on Rare Diseases CIPF-Health Research Institute Hospital La Fe (IIS-La Fe), Valencia 46026, Spain.
| | - David Salom
- Service of Ophthalmology, Manises Hospital, Generalitat Valenciana, 50, Manises, Valencia 46940, Spain; Biomedical Research Networking Center in Rare Diseases (CIBER-ER), Institute of Health Carlos III, Monforte de Lemos, 3-5. Pabellón 11, Madrid 28029, Spain; Catholic University of Valencia (UCV), Faculty of Health Sciences, Quevedo, 2, Valencia 46001, Spain.
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Monforte de Lemos, 3-5. Pabellón 11, Madrid 28029, Spain; Bioaraba, NanoBioCel Research Group, Jose Atxotegi Kalea, s/n, Txagorritxu, Vitoria-Gasteiz 01009, Spain; Joint Research Laboratory (JRL) on Bioprinting and Advanced Pharma Development, A Joined Venture of TECNALIA, Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, Vitoria-Gasteiz 01006, Spain.
| | - Regina Rodrigo
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, Valencia 46012, Spain; Joint Research Unit on Rare Diseases CIPF-Health Research Institute Hospital La Fe (IIS-La Fe), Valencia 46026, Spain; Biomedical Research Networking Center in Rare Diseases (CIBER-ER), Institute of Health Carlos III, Monforte de Lemos, 3-5. Pabellón 11, Madrid 28029, Spain; Catholic University of Valencia (UCV), Faculty of Health Sciences, Quevedo, 2, Valencia 46001, Spain.
| |
Collapse
|
2
|
Hung JH, Jain T, Khatri A, Nguyen BT, Nguyen CDT, Yavari N, Mobasserian A, Karaca I, Saeed Mohammadi S, Gupta AS, Or CMC, Akhavanrezayat A, Yasar C, Saengsirinavin AO, Than NTT, Anover FA, Elaraby O, El Feky D, Yoo WS, Zhang X, Thng ZX, Do DV, Nguyen QD. Inherited retinal disease-associated uveitis. Surv Ophthalmol 2025:S0039-6257(25)00057-8. [PMID: 40157547 DOI: 10.1016/j.survophthal.2025.03.011] [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: 11/01/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Inherited retinal diseases (IRDs) are genetic disorders characterized by progressive photoreceptor function loss, often leading to significant visual impairment. Uveitis has been increasingly recognized in the clinical course of some IRDs. Despite advances in understanding the genetic causes and pathophysiology of IRDs, gaps remain in understanding the roles of inflammation and autoimmunity in IRD and IRD-associated uveitis. This review discusses IRD-associated uveitis, including anterior, intermediate, posterior, and panuveitis, as well as complications such as cystoid macular edema and retinal vasculitis. In patients with IRD-associated uveitis, mutations affecting protein function in cilia or photoreceptor outer segments suggest a universal autoimmune mechanism triggered by the immunogenicity of shedding photoreceptor discs. Notably, in patients where uveitis is the initial sign, CRB1 mutations are often implicated, likely due to the compromised blood-retina barrier function or alterations in the external limiting membrane. Other mechanisms leading to uveitis preceding IRD diagnosis include ALPK1 mutations, which activate the proinflammatory NF-κB pathway, CAPN5 mutations, which lead to dysfunction of the innate and adaptive immune systems, and VCAN1 mutations, which elicit immunogenicity due to irregularities in vitreous modeling. Understanding these mechanisms could enhance the development of innovative treatments that target personalized inflammation pathways in IRDs.
Collapse
Affiliation(s)
- Jia-Horung Hung
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Genomic Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tanya Jain
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Dr. Shroff's Charity Eye Hospital, New Delhi, India
| | - Anadi Khatri
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Birat Eye Hospital, Biratnagar, Nepal; Gautam Buddha Eye care centre, Lumbini, Nepal
| | - Ba Trung Nguyen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Ophthalmology, Viet Nam National Children's Hospital, Ha Noi, Viet Nam
| | | | - Negin Yavari
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Azadeh Mobasserian
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Irmak Karaca
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; John A. Moran Eye Center, University of Utah, Salt Lake City, UT, US
| | - S Saeed Mohammadi
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ankur Sudhir Gupta
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Chi Mong Christopher Or
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Amir Akhavanrezayat
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Cigdem Yasar
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aim-On Saengsirinavin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Police General Hospital, Bangkok, Thailand
| | - Ngoc Trong Tuong Than
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Frances Andrea Anover
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Batangas Medical Center, Batangas, Philippines
| | - Osama Elaraby
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Dalia El Feky
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Woong-Sun Yoo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Ophthalmology, Gyeongsang National University College of Medicine, and Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Xiaoyan Zhang
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Zheng Xian Thng
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - Diana V Do
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Quan Dong Nguyen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
3
|
Chenjin Z. Exposure assessments of cadmium and lead with age-related eye disease: A systematic review and meta-analysis. J Trace Elem Med Biol 2025; 89:127631. [PMID: 40122018 DOI: 10.1016/j.jtemb.2025.127631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Age-related eye diseases, such as macular degeneration, cataracts, and glaucoma, are significant causes of vision loss in older adults. Emerging evidence suggests that environmental exposure to heavy metals, particularly Cadmium and lead, may play an essential role in the onset and progression of these conditions. METHODS This systematic review and meta-analysis aimed to evaluate the association between Cadmium and lead exposure and the risk of age-related eye diseases. A comprehensive literature search was conducted across multiple databases, including PubMed, Embase, Scopus, and Web of Science, covering studies published up to July 2024. The review included observational studies on the relationship between Cadmium or lead exposure and specific eye conditions. Data extraction and quality assessment were performed independently by two reviewers. The meta-analysis utilized a random effects model to analyze cadmium and lead levels in patient and control groups, with subgroup analyses based on the type of eye disease and study design. RESULTS The results revealed a significant association between cadmium exposure and an increased risk of eye diseases, particularly cataracts and glaucoma. Lead exposure was also linked to a higher risk of cataracts and macular degeneration. The study found substantial heterogeneity among the included studies, highlighting the variability in exposure assessment and population characteristics. CONCLUSIONS Despite these variations, the findings underscore the importance of addressing environmental exposures to toxic metals as potential risk factors for age-related eye diseases. Further research is needed to clarify the underlying mechanisms and to develop targeted interventions for reducing these risks.
Collapse
Affiliation(s)
- Zhao Chenjin
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610000, China.
| |
Collapse
|
4
|
Purdy R, John M, Bray A, Clare AJ, Copland DA, Chan YK, Henderson RH, Nerinckx F, Leroy BP, Yang P, Pennesi ME, MacLaren RE, Fischer MD, Dick AD, Xue K. Gene Therapy-Associated Uveitis (GTAU): Understanding and mitigating the adverse immune response in retinal gene therapy. Prog Retin Eye Res 2025; 106:101354. [PMID: 40090458 DOI: 10.1016/j.preteyeres.2025.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
Retinal gene therapy using adeno-associated viral (AAV) vectors has been a groundbreaking step-change in the treatment of inherited retinal diseases (IRDs) and could also be used to treat more common retinal diseases such as age-related macular degeneration and diabetic retinopathy. The delivery and expression of therapeutic transgenes in the eye is limited by innate and adaptive immune responses against components of the vector product, which has been termed gene therapy-associated uveitis (GTAU). This is clinically important as intraocular inflammation could lead to irreversible loss of retinal cells, deterioration of visual function and reduced durability of treatment effect associated with a costly one-off treatment. For retinal gene therapy to achieve an improved efficacy and safety profile for treating additional IRDs and more common diseases, the risk of GTAU must be minimised. We have collated insights from pre-clinical research, clinical trials, and the real-world implementation of AAV-mediated retinal gene therapy to help understand the risk factors for GTAU. We draw attention to an emerging framework, which includes patient demographics, vector construct, vector dose, route of administration, and choice of immunosuppression regime. Importantly, we consider efforts to date and potential future strategies to mitigate the adverse immune response across each of these domains. We advocate for more targeted immunomodulatory approaches to the prevention and treatment of GTAU based on better understanding of the underlying immune response.
Collapse
Affiliation(s)
- Ryan Purdy
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Molly John
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Alison J Clare
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; University College London (UCL) Institute of Ophthalmology, London, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - David A Copland
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; University College London (UCL) Institute of Ophthalmology, London, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - Ying Kai Chan
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA; Cirrus Therapeutics, Cambridge, MA, USA
| | - Robert H Henderson
- University College London (UCL) Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fanny Nerinckx
- Chirec Delta Hospital, Brussels, Belgium; Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Bart P Leroy
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium; Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium; Department of Head & Skin, Ghent University, Ghent, Belgium
| | - Paul Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, USA; Retina Foundation of the Southwest, Dallas, TX, USA
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - M Dominik Fischer
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; University College London (UCL) Institute of Ophthalmology, London, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
5
|
Lara-López A, Gonzalez-Imaz K, Rodríguez-Hidalgo M, Sarasola-Gastesi M, Escudero-Arrarás L, Milla-Navarro S, de la Villa P, Sagartzazu-Aizpurua M, Miranda JI, Aizpurua JM, de Munain AL, Vallejo-Illarramendi A, Ruiz-Ederra J. Topical Administration of Novel FKBP12 Ligand MP-004 Improves Retinal Function and Structure in Retinitis Pigmentosa Models. Invest Ophthalmol Vis Sci 2025; 66:56. [PMID: 40136284 PMCID: PMC11951062 DOI: 10.1167/iovs.66.3.56] [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: 10/15/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose This study evaluates the therapeutic potential of MP-004, a novel FKBP12 ligand, in the treatment of inherited retinal dystrophies (IRDs). MP-004 targets the FKBP12/RyR interaction, which is disrupted in several neurologic disorders with underlying oxidative stress. Methods The toxicity and efficacy of MP-004 were examined in vitro in 661W cells. Efficacy was evaluated in phototoxic and H2O2-induced damage using impedance assays, calcium imaging, and in situ PLA. In vivo, MP-004 efficacy was evaluated in the rd10 mouse model of retinitis pigmentosa (RP) by topical ocular instillation. Retinal function was assessed by electroretinography (ERG), visual acuity was measured using a water maze test, and retinal structure was analyzed morphometrically. Results MP-004 exhibited low toxicity (LD50: 1.22 mM) and effectively protected 661W cells from phototoxicity (EC50: 30.6 nM). Under oxidative stress conditions, MP-004 preserved the FKBP12.6/RyR2 interaction, restored cytosolic and endoplasmic reticulum calcium levels, and prevented cell death. In vivo, MP-004 significantly preserved retinal function in rd10 mice, with ERG wave amplitude increases of up to 50% in scotopic and 71% in photopic conditions, corresponding to rod and cone functions, respectively. Additionally, MP-004 improved visual acuity for low spatial frequency patterns and preserved retinal structure, with a 23% increase in outer nuclear layer thickness and preservation in the number of rods and cones and their segment length. Conclusions MP-004 shows promise as a therapeutic agent for RP, preserving retinal structure and function, likely through modulation of the FKBP12.6/RyR2 interaction. Further studies are needed to explore its pharmacokinetics and efficacy in other IRD models.
Collapse
Affiliation(s)
- Araceli Lara-López
- Miramoon Pharma, S.L., Donostia-San Sebastian, Spain
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Klaudia Gonzalez-Imaz
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | - María Rodríguez-Hidalgo
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
| | - Miren Sarasola-Gastesi
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- Department of Dermatology, Ophthalmology and ORL, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Leire Escudero-Arrarás
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
| | - Santiago Milla-Navarro
- Department of System Biology, University of Alcalá, Alcalá de Henares, Spain
- Visual Neurophysiology Group, Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pedro de la Villa
- Department of System Biology, University of Alcalá, Alcalá de Henares, Spain
- Visual Neurophysiology Group, Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Maialen Sagartzazu-Aizpurua
- Department of Organic Chemistry-I, Korta Research Center, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - José Ignacio Miranda
- Department of Organic Chemistry-I, Korta Research Center, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Jesús María Aizpurua
- Department of Organic Chemistry-I, Korta Research Center, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Adolfo López de Munain
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, OSAKIDETZA, Donostia-San Sebastián, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | - Javier Ruiz-Ederra
- Miramoon Pharma, S.L., Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- Department of Dermatology, Ophthalmology and ORL, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| |
Collapse
|
6
|
Scavuzzi BM, Shanmugam S, Yang M, Yao J, Hager H, Kaur B, Jia L, Abcouwer SF, Zacks DN. Remote Preconditioning Provides Protection Against Retinal Cell Death From Retinal Detachment. Invest Ophthalmol Vis Sci 2025; 66:34. [PMID: 39937497 PMCID: PMC11827864 DOI: 10.1167/iovs.66.2.34] [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: 08/23/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Purpose Remote preconditioning involves injury to a tissue that results in protection to a subsequent injury in a distal tissue. Here, we investigated the impact of remote preconditioning on retinal detachment (RD) injury, hypothesizing that a previous contralateral RD would protect the fellow retina against inflammation and cell death following its detachment. Methods RD was created in adult C57BL/6J mice with subretinal sodium hyaluronate injection. Preconditioning involved RD in the right eye at 1, 3, 7, or 28 days before left eye detachment, whereas the control group only received RD to the left eye. Retinas were harvested 24 hours post-left eye detachment in both groups. Cell death was assessed using Cell Death Detection ELISA and mRNA expression was evaluated via qRT-PCR. Results Contralateral RD promoted a transient protection against retinal cell death from 1 to 3 days and waned by 7 days compared with control RD retinas with intact fellow retinas. Contralateral RD significantly protected against post-RD cell death (P = 0.0002) and caspase 3 cleavage (P = 0.0449), compared with control RD retinas with intact fellow retinas 1-day post-RD. Detached fellow retinas from the preconditioning group expressed significantly less Tnfa (P = 0.0066), Cxcl10 (P = 0.0099), and Fas (P = 0.0223) mRNAs, compared with the detached retinas of the control group. In contrast, upregulation of type-I-IFN pathway genes, including Irf7 (P = 0.0106) and Ifit1 (P = 0.0740), following RD was higher in the preconditioning group. Conclusions RD in one eye produces a transient remote preconditioning effect that protects the fellow retina against retinal cell death following subsequent RD.
Collapse
Affiliation(s)
- Bruna Miglioranza Scavuzzi
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Mengling Yang
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Jingyu Yao
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Bhavneet Kaur
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Lin Jia
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - Steven F. Abcouwer
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | - David N. Zacks
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
7
|
Jin F, Yan Y, Ye Z, Wang L, Deng C, Jiang J, Dong K. CDR1as Deficiency Prevents Photoreceptor Degeneration by Regulating miR-7a-5p/α-syn/Parthanatos Pathway in Retinal Detachment. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:293-305. [PMID: 39566824 DOI: 10.1016/j.ajpath.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024]
Abstract
Retinal detachment (RD) is the separation of the neural retina from the retinal pigment epithelium, with photoreceptor degeneration being a major cause of irreversible vision loss. Herein, ischemia and hypoxia after RD decreased the level of miR-7a-5p (miR-7) and promoted the expression of its main target, α-synuclein (α-syn), which activated the parthanatos pathway and led to photoreceptor damage. Circular RNA CDR1as is an antisense transcript of cerebellar degeneration-associated protein 1, which functions as a "sponge" for miR-7, thereby regulating the abundance and activity of miR-7. In this study, CDR1as expression was elevated after RD. Adeno-associated virus serotype 9 vector containing the shRNA-CDR1as sequence was used to inhibit CDR1as expression via subretinal injection. Hematoxylin and eosin staining and transmission electron microscopy revealed that the morphology and outer nuclear layer thickness of the retina were preserved and photoreceptor cell death was decreased after experimental RD in mice. Mechanistically, CDR1as deficiency significantly increased the expression of miR-7, then decreased the expression of α-syn, poly (ADP-ribose) polymerase 1, apoptosis-inducing factor, and migration inhibitory factor. Furthermore, visual function was improved as shown by Morris water maze experiments in the mouse model of RD. These findings suggest a surprisingly neuroprotective role for CDR1as deficiency, which is probably mediated by enhancing miR-7 activity and inhibiting α-syn/poly (ADP-ribose) polymerase 1/apoptosis-inducing factor pathway, thereby preventing photoreceptor degeneration.
Collapse
Affiliation(s)
- Feiyu Jin
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanye Yan
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ziyang Ye
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lisong Wang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Can Deng
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiazhen Jiang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kai Dong
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
8
|
Guo Y, Ye Z, Deng C, Wang L, Gu Q, Ji K, Li X, Dong K. Effect of Preoperative Glucocorticoid Application on Vitreous Parthanatos-Related Protein Expression in Patients with Rhegmatogenous Retinal Detachment Associated with Choroidal Detachment. Curr Eye Res 2025; 50:182-189. [PMID: 39373217 DOI: 10.1080/02713683.2024.2408755] [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: 02/18/2024] [Revised: 08/30/2024] [Accepted: 09/21/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE The protein concentrations of apoptosis inducing factor (AIF), macrophage migration inhibitory factor (MIF), interleukin-1β (IL-1β), poly ADP ribose polymerase-1 (PARP-1), poly (ADP-ribose) (PAR), α-synuclein (α-SYN), monocyte chemotactic protein‑1 (MCP-1) and tumor necrosis factor-α (TNF-α) in the vitreous of eyes with rhegmatogenous retinal detachment associated with choroidal detachment (RRDCD) were observed and analyzed. METHODS A total of 57 patients' samples were included. 30 patients with RRD were set as the control group, 27 patients with RRDCD were set as the experimental group (16 patients with preoperative glucocorticosteroid (GC+) and 11 patients without preoperative glucocorticosteroid (GC-)). The levels of AIF, MIF, IL-1β, PARP-1, PAR, α-SYN, MCP-1 and TNF-α in vitreous of patients in the control and experimental groups were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS The concentration of AIF in the vitreous was higher in the RRD group (9.96 ± 2.78 ng/ml) than in the RRDCD (GC+) group (7.65 ± 2.13 ng/ml, p = 0.006),the RRDCD (GC+) group was lower than the RRDCD (GC-) group (10.28 ± 2.81 ng/ml) (p = 0.013). The concentration of MIF in vitreous fluid was lower in the RRDCD (GC+) group (61.21 ± 17.56 ng/ml) than in the RRDCD (GC-) group (74.30 ± 9.66 ng/ml, p = 0.039). In the experimental group, the protein concentration of MCP-1 in the RRDCD (GC+) group was higher in the preoperative PVR grading C (284.93 ± 54.96 ng/ml) grade than in the D grade (225.94 ± 24.05 ng/ml) (p = 0.050); The protein concentration of MIF was lower in the RRDCD (GC+) group of patients with an ocular axis of <26 mm (56.19 ± 6.99 ng/ml) than in those with an ocular axis of ≥26 mm (76.26 ± 26.60 ng/ml, p = 0.043). CONCLUSION Low expression of Parthanatos-related proteins is present in the vitreous of patients with RRDCD (GC+), and preoperative treatment with glucocorticoids may reduce the expression of Parthanatos-related proteins.
Collapse
Affiliation(s)
- Yue Guo
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Ziyang Ye
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Can Deng
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Lin Wang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Qihong Gu
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Kangkang Ji
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Xiaomeng Li
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Kai Dong
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| |
Collapse
|
9
|
Azam M, Jastrzebska B. Mechanisms of Rhodopsin-Related Inherited Retinal Degeneration and Pharmacological Treatment Strategies. Cells 2025; 14:49. [PMID: 39791750 PMCID: PMC11720364 DOI: 10.3390/cells14010049] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary disease characterized by progressive vision loss ultimately leading to blindness. This condition is initiated by mutations in genes expressed in retinal cells, resulting in the degeneration of rod photoreceptors, which is subsequently followed by the loss of cone photoreceptors. Mutations in various genes expressed in the retina are associated with RP. Among them, mutations in the rhodopsin gene (RHO) are the most common cause of this condition. Due to the involvement of numerous genes and multiple mutations in a single gene, RP is a highly heterogeneous disease making the development of effective treatments particularly challenging. The progression of this disease involves complex cellular responses to restore cellular homeostasis, including the unfolded protein response (UPR) signaling, autophagy, and various cell death pathways. These mechanisms, however, often fail to prevent photoreceptor cell degradation and instead contribute to cell death under certain conditions. Current research focuses on the pharmacological modulation of the components of these pathways and the direct stabilization of mutated receptors as potential treatment strategies. Despite these efforts, the intricate interplay between these mechanisms and the diverse causative mutations involved has hindered the development of effective treatments. Advancing our understanding of the interactions between photoreceptor cell death mechanisms and the specific genetic mutations driving RP is critical to accelerate the discovery and development of therapeutic strategies for this currently incurable disease.
Collapse
Affiliation(s)
- Maria Azam
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
10
|
Bishayee K, Lee SH, Heo YJ, Cho ML, Park YS. The unanticipated contribution of Zap70 in retinal degeneration: Implications for microglial inflammatory activation. Prog Neurobiol 2025; 244:102706. [PMID: 39710334 DOI: 10.1016/j.pneurobio.2024.102706] [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: 06/27/2024] [Revised: 11/04/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Inflammation is a major mechanism of photoreceptor cell death in the retina during macular degeneration leading to the blindness. In this study, we investigated the role of the kinase molecule Zap70, which is an inflammatory regulator of the systemic immune system, to elucidate the control mechanism of inflammation in the retina. We observed activated microglial cells migrated and populated the retinal layer following blue LED-induced photoreceptor degeneration and activated microglial cells in the LED-injured retina expressed Zap70, unlike the inactive microglial cells in the normal retina. Visual function was considerably decreased in blue-LED light-exposed mice, and animals with Zap70 mutations were adversely affected. Furthermore, extensive photoreceptor cell death was observed in the SKG mice, bearing a Zap70 mutation that induces autoimmune disease. In the blue-LED light-exposed groups, SKG retinas had significantly higher levels of inflammatory cytokines than those in wild-type mice. Furthermore, regulating Zap70 activity has a significant influence on microglial inflammatory state. We discovered that active microglial cells expressing Zap70 could modify vascular endothelial growth factor A (Vegfa) signaling in primary retinal pigment epithelial (RPE) cells. Our novel study revealed that the production of Zap70 by retinal microglial cells is responsible for inflammatory signals that promote apoptosis in photoreceptor cells. Furthermore, Zap70-positive microglial cells were capable of regulating Vegfa signaling in RPE cells, which matches the hallmark of macular degeneration. Overall, we discovered Zap70's inflammatory activity in the retina, which is necessary for upregulating multiple inflammatory cytokines and cell death. Zap70 represents a novel therapeutic target for treating retinal degeneration.
Collapse
Affiliation(s)
- Kausik Bishayee
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Seung-Hee Lee
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Yeon-Jin Heo
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Mi-La Cho
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, South Korea
| | - Yong Soo Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
11
|
McDonald A, Gallego C, Andriessen C, Orlová M, Gonçalves MAFV, Wijnholds J. Conventional and Tropism-Modified High-Capacity Adenoviral Vectors Exhibit Similar Transduction Profiles in Human iPSC-Derived Retinal Organoids. Int J Mol Sci 2024; 26:55. [PMID: 39795914 PMCID: PMC11719574 DOI: 10.3390/ijms26010055] [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: 11/20/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Viral vector delivery of gene therapy represents a promising approach for the treatment of numerous retinal diseases. Adeno-associated viral vectors (AAV) constitute the primary gene delivery platform; however, their limited cargo capacity restricts the delivery of several clinically relevant retinal genes. In this study, we explore the feasibility of employing high-capacity adenoviral vectors (HC-AdVs) as alternative delivery vehicles, which, with a capacity of up to 36 kb, can potentially accommodate all known retinal gene coding sequences. We utilized HC-AdVs based on the classical adenoviral type 5 (AdV5) and on a fiber-modified AdV5.F50 version, both engineered to deliver a 29.6 kb vector genome encoding a fluorescent reporter construct. The tropism of these HC-AdVs was evaluated in an induced pluripotent stem cell (iPSC)-derived human retinal organoid model. Both vector types demonstrated robust transduction efficiency, with sustained transgene expression observed for up to 110 days post-transduction. Moreover, we found efficient transduction of photoreceptors and Müller glial cells, without evidence of reactive gliosis or loss of photoreceptor cell nuclei. However, an increase in the thickness of the photoreceptor outer nuclear layer was observed at 110 days post-transduction, suggesting potential unfavorable effects on Müller glial or photoreceptor cells associated with HC-AdV transduction and/or long-term reporter overexpression. These findings suggest that while HC-AdVs show promise for large retinal gene delivery, further investigations are required to assess their long-term safety and efficacy.
Collapse
Affiliation(s)
- Andrew McDonald
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.M.); (C.A.)
| | - Carmen Gallego
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.M.); (C.A.)
| | - Charlotte Andriessen
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.M.); (C.A.)
| | - Michaela Orlová
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.M.); (C.A.)
| | - Manuel A. F. V. Gonçalves
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (A.M.); (C.A.)
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
12
|
Bacherini D, Maggi L, Faraldi F, Sodi A, Vannozzi L, Mazzoni A, Capone M, Virgili G, Vicini G, Falsini B, Cosmi L, Viggiano P, Rizzo S, Annunziato F, Giansanti F, Liotta F. CD3+CD4-CD8- Double-Negative Lymphocytes Are Increased in the Aqueous Humor of Patients with Retinitis Pigmentosa: Their Possible Role in Mediating Inflammation. Int J Mol Sci 2024; 25:13163. [PMID: 39684872 DOI: 10.3390/ijms252313163] [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: 10/20/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Recently, evidence has supported a significant role for immune and oxidative-mediated damage underlying the pathogenesis of different types of retinal diseases, including retinitis pigmentosa (RP). Our study aimed to evaluate the presence of immune cells and mediators in patients with RP using flow cytometric analysis of peripheral blood (PB) and aqueous humor (AH) samples. We recruited 12 patients with RP and nine controls undergoing cataract surgery. Flow cytometric analysis of PB and AH samples provided a membrane staining that targeted surface molecules (CD14, CD16, CD19, CD3, CD4, CD8, and CD161) identifying monocytes, natural killer (NK) cells, B cells, T cells, and T subpopulations, respectively. Moreover, lymphocytes were polyclonally stimulated to evaluate cytokine (CK) production at single-cell level. The circulating immune cell distribution was comparable between patients with RP and controls. Conversely, in the AH of controls we could detect no cells, while in the RP AH samples we found infiltrating leukocytes, consisting of T (CD3+), B (CD19+), NK (CD16+CD3-) cells, and monocytes (CD14+). In patients with RP, the frequency of most infiltrating immune cell populations was similar between the AH and PB. However, among T cell subpopulations, the frequency of CD3+CD4+ T cells was significantly lower in the RP AH compared to RP PB, whereas CD3+CD4-CD8- double-negative (DN) T cells were significantly higher in the RP AH compared to RP PB. Cytokine production analysis revealed a trend toward an increased frequency of CD3+CD8-CD161+IFN-ɣ-producing cells and a decreased frequency of CD3+CD8+IL-4-producing cells in the RP AH compared to RP PB. The detection of immune cells, particularly DN T cells, and a Th1-skewed phenotype in RP AH suggests immune-mediated and inflammatory mechanisms in the disease.
Collapse
Affiliation(s)
- Daniela Bacherini
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Francesco Faraldi
- Ophthalmology Unit, Surgical Department, A.O. Ordine Mauriziano, 10128 Turin, Italy
| | - Andrea Sodi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Lorenzo Vannozzi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Gianni Virgili
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Giulio Vicini
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Benedetto Falsini
- UOC Oculistica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Head and Neck and Sensory Organs, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Pasquale Viggiano
- Department of Translational Biomedicine Neuroscience, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Stanislao Rizzo
- UOC Oculistica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Head and Neck and Sensory Organs, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Consiglio Nazionale delle Ricerche, Istituto di Neuroscienze, 56124 Pisa, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Fabrizio Giansanti
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
13
|
Moseley J, Leest T, Larsson K, Magrelli A, Stoyanova-Beninska V. Inherited retinal dystrophies and orphan designations in the European Union. Eur J Ophthalmol 2024; 34:1631-1641. [PMID: 38500388 PMCID: PMC11542323 DOI: 10.1177/11206721241236214] [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: 10/02/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Inherited Retinal Dystrophies (IRD) are diverse rare diseases that affect the retina and lead to visual impairment or blindness. Research in this field is ongoing, with over 60 EU orphan medicinal products designated in this therapeutic area by the Committee for Orphan Medicinal Products (COMP) at the European Medicines Agency (EMA). Up to now, COMP has used traditional disease terms, like retinitis pigmentosa, for orphan designation regardless of the product's mechanism of action. The COMP reviewed the designation approach for IRDs taking into account all previous Orphan Designations (OD) experience in IRDs, the most relevant up to date scientific literature and input from patients and clinical experts. Following the review, the COMP decided that there should be three options available for orphan designation concerning the condition: i) an amended set of OD groups for therapies that might be used in a broad spectrum of conditions, ii) a gene-specific designation for targeted therapies, and iii) an occasional term for products that do not fit in the above two categories. The change in the approach to orphan designation in IRDs caters for different scenarios to allow an optimum approach for future OD applications including the option of a gene-specific designation. By applying this new approach, the COMP increases the regulatory clarity, efficiency, and predictability for sponsors, aligns EU regulatory tools with the latest scientific and medical developments in the field of IRDs, and ensures that all potentially treatable patients will be included in the scope of an OD.
Collapse
Affiliation(s)
- Jane Moseley
- European Medicines Agency, Amsterdam, The Netherlands
| | - Tim Leest
- Committee for Orphan Medicinal Products at the European Medicines Agency, Amsterdam, The Netherlands
- Federal Agency for Medicines and Health Products, Brussels, Belgium
| | | | - Armando Magrelli
- Committee for Orphan Medicinal Products at the European Medicines Agency, Amsterdam, The Netherlands
- National Center for Drug Research and Evaluation- Istituto Superiore di Sanità, Rome, Italy
| | - Violeta Stoyanova-Beninska
- Committee for Orphan Medicinal Products at the European Medicines Agency, Amsterdam, The Netherlands
- Medicines Evaluation Board (MEB), Utrecht, The Netherlands
| |
Collapse
|
14
|
Marques JP, Ferreira N, Moreno N, Marta A, Vaz-Pereira S, Estrela-Silva S, Costa J, Cardoso AR, Neves P, Duarte L, Meira D, Pires J, Menezes C, Rodrigues F, Arede P, Coutinho A, Cabral D, Coutinho I, Ribeiro M, Macedo M, Brito S, Isidro F, Rodrigues FG, Sousa JPC, Marques M, Martins R, Silva E. Current management of inherited retinal degenerations in Portugal (IRD-PT survey). Sci Rep 2024; 14:21473. [PMID: 39277603 PMCID: PMC11401845 DOI: 10.1038/s41598-024-72589-4] [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: 08/29/2023] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
Inherited retinal dystrophies/degenerations (IRDs) are the leading cause of visual impairment and incurable familial blindness in the Western world. Given the clinical and genetic heterogeneity, establishing a molecular diagnosis is especially relevant. The aim of this study was to perform the first nationwide survey to understand the prevalence and current management of IRDs in Portugal. A response was obtained from 26 healthcare providers (HCP) (76.5% response rate). Only 4 respondents reported not managing IRD patients. Most HCPs (68.1%) reported managing up to 100 patients, while three currently manage between 501 and 1000 patients. Based on the Portuguese population, an estimated IRD prevalence of 0.031%, i.e., about 1 in 3000 individuals, was calculated. In most HCPs (86.3%), most patients are adults, and non-syndromic retinitis pigmentosa is the most frequent diagnosis. Only 4 HCPs currently use the national, web-based IRD registry (IRD-PT). However, all but one respondent expressed interest in participating in such a registry. Genetic testing is available in 54.5%, with 58.3% HCPs reporting solved rates between 61-80%, but 4 to 9 months to get a genetic test result in 83.4% of cases. Based on this survey, the prevalence of biallelic RPE65-associated disease in Portugal is 0.00031%, i.e., approximately 1:300,000 individuals. Data from this study provide vital background information on national differences in the diagnosis and management of IRD patients. Nationwide implementation of the IRD-PT registry should be encouraged and supported to provide population-based reference data and to identify patients eligible for current and future therapies.
Collapse
Affiliation(s)
- João Pedro Marques
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Coimbra, EPE, Coimbra, Portugal.
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Coimbra, EPE, Praceta Prof. Mota Pinto, 3000-075, Coimbra, Portugal.
| | - Nuno Ferreira
- Ophthalmology Deparment, Unidade Local de Saúde (ULS) de Trás-os-Montes e Alto Douro, EPE, Vila Real, Portugal
| | - Natacha Moreno
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Barcelos/Esposende, EPE, Barcelos, Portugal
| | - Ana Marta
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Santo António, EPE, Porto, Portugal
| | - Sara Vaz-Pereira
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Santa Maria, EPE, Lisboa, Portugal
| | - Sérgio Estrela-Silva
- Ophthalmology Department, Unidade Local de Saúde (ULS) de São João, EPE, Porto, Portugal
| | - José Costa
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Braga, EPE, Braga, Portugal
| | - Ana Rocha Cardoso
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Baixo Alentejo, EPE, Beja, Portugal
| | - Pedro Neves
- Ophthalmology Department, Unidade Local de Saúde (ULS) da Arrábida, EPE, Setúbal, Portugal
| | - Lilianne Duarte
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Entre Douro e Vouga, EPE, Santa Maria da Feira, Portugal
| | - Dália Meira
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Gaia/Espinho, EPE, Gaia, Portugal
| | - Joana Pires
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Alto Ave, EPE, Guimarães, Portugal
| | - Carlos Menezes
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Alto Minho, EPE, Viana Do Castelo, Portugal
| | - Filipa Rodrigues
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Litoral Alentejano, EPE, Santiago Do Cacém, Portugal
| | - Pedro Arede
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Lisboa Ocidental, EPE, Lisboa, Portugal
| | - André Coutinho
- Ophthalmology Department, Unidade Local de Saúde (ULS) da Região de Aveiro, EPE, Aveiro, Portugal
| | - Diogo Cabral
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Almada-Seixal, EPE, Lisboa, Portugal
| | - Inês Coutinho
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Amadora/Sintra, EPE, Amadora, Portugal
| | - Miguel Ribeiro
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Viseu Dão-Lafões, EPE, Viseu, Portugal
| | - Marta Macedo
- Ophthalmology Department, Hospital Dr. Nélio Mendonça, EPE, Funchal, Portugal
| | - Sérgio Brito
- Ophthalmology Department, Unidade Local de Saúde (ULS) de Castelo Branco, EPE, Castelo Branco, Portugal
| | - Filipe Isidro
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Algarve, EPE, Faro, Portugal
| | - Filipa Gomes Rodrigues
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Estuário do Tejo, EPE, Vila Franca de Xira, Portugal
| | - João Paulo Castro Sousa
- Ophthalmology Department, Unidade Local de Saúde (ULS) da Região de Leiria, EPE, Leiria, Portugal
| | - Marco Marques
- Ophthalmology Department, Unidade Local de Saúde (ULS) do Baixo Mondego, EPE, Figueira da Foz, Portugal
| | | | - Eduardo Silva
- Ophthalmology Department, Unidade Local de Saúde (ULS) de São José, EPE, Lisboa, Portugal
| |
Collapse
|
15
|
Tao Y, Fukushima M, Shimokawa S, Zhao H, Okita A, Fujiwara K, Takeda A, Mukai S, Sonoda KH, Murakami Y. Ocular and Serum Profiles of Inflammatory Molecules Associated With Retinitis Pigmentosa. Transl Vis Sci Technol 2024; 13:18. [PMID: 39120884 PMCID: PMC11318359 DOI: 10.1167/tvst.13.8.18] [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: 09/24/2023] [Accepted: 06/30/2024] [Indexed: 08/10/2024] Open
Abstract
Purpose To investigate the profiles and correlations between local and systemic inflammatory molecules in patients with retinitis pigmentosa (RP). Methods The paired samples of aqueous humor and serum were collected from 36 eyes of 36 typical patients with RP and 25 eyes of age-matched patients with cataracts. The concentration of cytokines/chemokines was evaluated by a multiplexed immunoarray (Q-Plex). The correlations between ocular and serum inflammatory molecules and their association with visual function were analyzed. Results The aqueous levels of IL-6, Eotaxin, GROα, I-309, IL-8, IP-10, MCP-1, MCP-2, RANTES, and TARC were significantly elevated in patients with RP compared to controls (all P < 0.05). The detection rate of aqueous IL-23 was higher in patients with RP (27.8%) compared with controls (0%). In patients with RP, Spearman correlation test demonstrated positive correlations for IL-23, I-309, IL-8, and RANTES between aqueous and serum expression levels (IL-23: ⍴ = 0.8604, P < 0.0001; I-309: ρ = 0.4172, P = 0.0113; IL-8: ρ = 0.3325, P = 0.0476; RANTES: ρ = 0.6685, P < 0.0001). In addition, higher aqueous IL-23 was associated with faster visual acuity loss in 10 patients with RP with detected aqueous IL-23 (ρ = 0.4119 and P = 0.0264). Multiple factor analysis confirmed that aqueous and serum IL-23 were associated with visual acuity loss in patients with RP. Conclusions These findings suggest that ocular and systemic inflammatory responses have a close interaction in patients with RP. Further longitudinal studies with larger cohorts are needed to explore the correlation between specific inflammatory pathways and the progression of RP. Translational Relevance This study demonstrates the local-systemic interaction of immune responses in patients with RP.
Collapse
Affiliation(s)
- Yan Tao
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Fukushima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sakurako Shimokawa
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Huanyu Zhao
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ayako Okita
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohta Fujiwara
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsunobu Takeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Ophthalmology, Faculty of Medicine, Oita University, Oita, Japan
| | - Shizuo Mukai
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
16
|
Wang XL, Gao YX, Yuan QZ, Zhang M. Protective effects of CY-09 and astaxanthin on NaIO 3-induced photoreceptor inflammation via the NLRP3/autophagy pathway. Int J Ophthalmol 2024; 17:1217-1231. [PMID: 39026909 PMCID: PMC11246927 DOI: 10.18240/ijo.2024.07.05] [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: 11/29/2023] [Accepted: 04/11/2024] [Indexed: 07/20/2024] Open
Abstract
AIM To study the effect of the NLRP3/autophagy pathway on the photoreceptor inflammatory response and the protective mechanism of CY-09 and astaxanthin (AST). METHODS ICR mice were intraperitoneally injected NaIO3, CY-09, AST successively and divided into 5 groups, including the control, NaIO3, NaIO3+CY-09, NaIO3+AST, and NaIO3+CY-09+AST groups. Spectral domain optical coherence tomography and flash electroretinogram were examined and the retina tissues were harvested for immunohistochemistry, enzyme linked immunosorbent assay (ELISA), and Western blotting. Retinal pigment epithelium cell line (ARPE-19 cells) and mouse photoreceptor cells line (661W cells) were also treated with NaIO3, CY-09, and AST successively. Cell proliferation was assessed by cell counting kit-8 (CCK-8) assay. Apoptosis was analyzed by flow cytometry. Changes in autophagosome morphology were observed by transmission electron microscopy. Quantitative polymerase chain reaction (qPCR) was used to detect NLRP3 and caspase-1. NLRP3, caspase-1, cleaved caspase-1, p62, Beclin-1, and LC3 protein levels were measured by Western blotting. IL-1β and IL-18 were measured by ELISA. RESULTS Compared with the control group, the activity of NaIO3-treated 661W cells decreased within 24 and 48h, apoptosis increased, NLRP3, caspase-1, IL-1β and IL-18 levels increased, and autophagy-related protein levels increased (P<0.05). Compared with NaIO3 group, CY-09 and AST inhibited apoptosis (P<0.05), reduced NLRP3, caspase-1, IL-1β and IL-18 expression (P<0.05), and inhibited autophagy. Compared with the other groups, CY-09 combined with AST significantly decreased NLRP3 expression and inhibited the expression of the autophagy-related proteins p62, Beclin-1, and LC3 in vitro and in vivo (P<0.05). CONCLUSION CY-09 and AST inhibit NaIO3-induced inflammatory damage through the NLRP3/autophagy pathway in vitro and in vivo. CY-09 and AST may protect retina from inflammatory injury.
Collapse
Affiliation(s)
- Xiao-Li Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yun-Xia Gao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiong-Zhen Yuan
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
17
|
Mead AJ, Ahluwalia K, Ebright B, Zhang Z, Dave P, Li Z, Zhou E, Naik AA, Ngu R, Chester C, Lu A, Asante I, Pollalis D, Martinez JC, Humayun M, Louie S. Loss of 15-Lipoxygenase in Retinodegenerative RCS Rats. Int J Mol Sci 2024; 25:2309. [PMID: 38396985 PMCID: PMC10889776 DOI: 10.3390/ijms25042309] [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: 01/10/2024] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Retinitis pigmentosa (RP) is a retinal degenerative disease associated with a diversity of genetic mutations. In a natural progression study (NPS) evaluating the molecular changes in Royal College of Surgeons (RCS) rats using lipidomic profiling, RNA sequencing, and gene expression analyses, changes associated with retinal degeneration from p21 to p60 were evaluated, where reductions in retinal ALOX15 expression corresponded with disease progression. This important enzyme catalyzes the formation of specialized pro-resolving mediators (SPMs) such as lipoxins (LXs), resolvins (RvDs), and docosapentaenoic acid resolvins (DPA RvDs), where reduced ALOX15 corresponded with reduced SPMs. Retinal DPA RvD2 levels were found to correlate with retinal structural and functional decline. Retinal RNA sequencing comparing p21 with p60 showed an upregulation of microglial inflammatory pathways accompanied by impaired damage-associated molecular pattern (DAMP) clearance pathways. This analysis suggests that ALXR/FPR2 activation can ameliorate disease progression, which was supported by treatment with an LXA4 analog, NAP1051, which was able to promote the upregulation of ALOX12 and ALOX15. This study showed that retinal inflammation from activated microglia and dysregulation of lipid metabolism were central to the pathogenesis of retinal degeneration in RP, where ALXR/FPR2 activation was able to preserve retinal structure and function.
Collapse
Affiliation(s)
- Andrew James Mead
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Kabir Ahluwalia
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Brandon Ebright
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Zeyu Zhang
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Priyal Dave
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Zeyang Li
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Eugene Zhou
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Aditya Anil Naik
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Rachael Ngu
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Catherine Chester
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Angela Lu
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
| | - Isaac Asante
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
- University of Southern California Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (D.P.); (J.C.M.); (M.H.)
- University of Southern California Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dimitrios Pollalis
- University of Southern California Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (D.P.); (J.C.M.); (M.H.)
- University of Southern California Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Juan Carlos Martinez
- University of Southern California Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (D.P.); (J.C.M.); (M.H.)
- University of Southern California Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mark Humayun
- University of Southern California Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (D.P.); (J.C.M.); (M.H.)
- University of Southern California Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Stan Louie
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (A.J.M.); (K.A.); (B.E.); (Z.Z.); (P.D.); (Z.L.); (E.Z.); (A.A.N.); (R.N.); (C.C.); (A.L.); (I.A.)
- University of Southern California Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (D.P.); (J.C.M.); (M.H.)
- University of Southern California Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
18
|
Yi W, Xue Y, Qing W, Cao Y, Zhou L, Xu M, Sun Z, Li Y, Mai X, Shi L, He C, Zhang F, Duh EJ, Cao Y, Liu X. Effective treatment of optic neuropathies by intraocular delivery of MSC-sEVs through augmenting the G-CSF-macrophage pathway. Proc Natl Acad Sci U S A 2024; 121:e2305947121. [PMID: 38289952 PMCID: PMC10861878 DOI: 10.1073/pnas.2305947121] [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: 04/13/2023] [Accepted: 12/21/2023] [Indexed: 02/01/2024] Open
Abstract
Optic neuropathies, characterized by injury of retinal ganglion cell (RGC) axons of the optic nerve, cause incurable blindness worldwide. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) represent a promising "cell-free" therapy for regenerative medicine; however, the therapeutic effect on neural restoration fluctuates, and the underlying mechanism is poorly understood. Here, we illustrated that intraocular administration of MSC-sEVs promoted both RGC survival and axon regeneration in an optic nerve crush mouse model. Mechanistically, MSC-sEVs primarily targeted retinal mural cells to release high levels of colony-stimulating factor 3 (G-CSF) that recruited a neural restorative population of Ly6Clow monocytes/monocyte-derived macrophages (Mo/MΦ). Intravitreal administration of G-CSF, a clinically proven agent for treating neutropenia, or donor Ly6Clow Mo/MΦ markedly improved neurological outcomes in vivo. Together, our data define a unique mechanism of MSC-sEV-induced G-CSF-to-Ly6Clow Mo/MΦ signaling in repairing optic nerve injury and highlight local delivery of MSC-sEVs, G-CSF, and Ly6Clow Mo/MΦ as therapeutic paradigms for the treatment of optic neuropathies.
Collapse
Affiliation(s)
- Wei Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Ying Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Wenjie Qing
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Yingxue Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Lingli Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Mingming Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Zehui Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Yuying Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Xiaomei Mai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Le Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Chang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| | - Elia J. Duh
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm17165, Stockholm, Sweden
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, People’s Republic of China
| |
Collapse
|
19
|
Sarkar H, Tracey-White D, Hagag AM, Burgoyne T, Nair N, Jensen LD, Edwards MM, Moosajee M. Loss of REP1 impacts choroidal melanogenesis and vasculogenesis in choroideremia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166963. [PMID: 37989423 PMCID: PMC11157692 DOI: 10.1016/j.bbadis.2023.166963] [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: 06/15/2023] [Revised: 10/13/2023] [Accepted: 11/14/2023] [Indexed: 11/23/2023]
Abstract
Choroideremia (CHM) is a rare X-linked chorioretinal dystrophy affecting the photoreceptors, retinal pigment epithelium (RPE) and choroid, however, the involvement of the choroid in disease progression is not fully understood. CHM is caused by mutations in the CHM gene, encoding the ubiquitously expressed Rab escort protein 1 (REP1). REP1 plays an important role in intracellular trafficking of vesicles, including melanosomes. In this study, we examined the ultrastructure of the choroid in chmru848 fish and Chmnull/WT mouse models using transmission electron and confocal microscopy. Significant pigmentary disruptions were observed, with lack of melanosomes in the choroid of chmru848 fish from 4 days post fertilisation (4dpf), and a reduction in choroidal blood vessel diameter and interstitial pillars suggesting a defect in vasculogenesis. Total melanin and expression of melanogenesis genes tyr, tryp1a, mitf, dct and pmel were also reduced from 4dpf. In Chmnull/WT mice, choroidal melanosomes were significantly smaller at 1 month, with reduced eumelanin at 1 year. The choroid in CHM patients were also examined using spectral domain optical coherence tomography (SD-OCT) and OCT-angiography (OCT-A) and the area of preserved choriocapillaris (CC) was found to be smaller than that of overlying photoreceptors, suggesting that the choroid is degenerating at a faster rate. Histopathology of an enucleated eye from a 74-year-old CHM male patient revealed isolated areas of RPE but no associated underlying CC. Pigmentary disruptions in CHM animal models reveal an important role for REP1 in melanogenesis, and drugs that improve melanin production represent a potential novel therapeutic avenue.
Collapse
Affiliation(s)
- Hajrah Sarkar
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, UK; The Francis Crick Institute, London, UK
| | - Dhani Tracey-White
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, UK
| | - Ahmed M Hagag
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, UK; Department of Genetics, Moorfields Eye Hospital NHS Foundation Trust, London, UK; Boehringer Ingelheim Limited, Bracknell, UK
| | - Thomas Burgoyne
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, UK
| | - Neelima Nair
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, UK; The Francis Crick Institute, London, UK
| | - Lasse D Jensen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Malia M Edwards
- The Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mariya Moosajee
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, UK; Department of Genetics, Moorfields Eye Hospital NHS Foundation Trust, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
20
|
Fischer MD, Simonelli F, Sahni J, Holz FG, Maier R, Fasser C, Suhner A, Stiehl DP, Chen B, Audo I, Leroy BP. Real-World Safety and Effectiveness of Voretigene Neparvovec: Results up to 2 Years from the Prospective, Registry-Based PERCEIVE Study. Biomolecules 2024; 14:122. [PMID: 38254722 PMCID: PMC10813228 DOI: 10.3390/biom14010122] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Voretigene neparvovec (VN) is the first available gene therapy for patients with biallelic RPE65-mediated inherited retinal dystrophy who have sufficient viable retinal cells. PERCEIVE is an ongoing, post-authorization, prospective, multicenter, registry-based observational study and is the largest study assessing the real-world, long-term safety and effectiveness of VN. Here, we present the outcomes of 103 patients treated with VN according to local prescribing information. The mean (SD) age was 19.5 (10.85) years, 52 (50.5%) were female, and the mean (SD) duration of the follow up was 0.8 (0.64) years (maximum: 2.3 years). Thirty-five patients (34%) experienced ocular treatment-emergent adverse events (TEAEs), most frequently related to chorioretinal atrophy (n = 13 [12.6%]). Eighteen patients (17.5%; 24 eyes [13.1%]) experienced ocular TEAEs of special interest, including intraocular inflammation and/or infection related to the procedure (n = 7). The mean (SD) changes from baseline in full-field light-sensitivity threshold testing (white light) at month 1, month 6, year 1, and year 2 were -16.59 (13.48) dB (51 eyes), -18.24 (14.62) dB (42 eyes), -15.84 (14.10) dB (10 eyes), and -13.67 (22.62) dB (13 eyes), respectively. The change in visual acuity from baseline was not clinically significant. Overall, the outcomes of the PERCEIVE study are consistent with the findings of VN pivotal clinical trials.
Collapse
Affiliation(s)
- M. Dominik Fischer
- Centre for Ophthalmology, University of Tübingen, 72076 Tübingen, Germany
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Francesca Simonelli
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania Luigi Vanvitelli, Via S. Pansini, 5, 80131 Napoli, Italy;
| | - Jayashree Sahni
- Novartis Pharma AG, 4056 Basel, Switzerland; (J.S.); (A.S.); (D.P.S.)
| | - Frank G. Holz
- Department of Ophthalmology, University of Bonn, Ernst-Abbe-Straße 2, 53127 Bonn, Germany;
| | - Rainer Maier
- Novartis Pharma AG, 4056 Basel, Switzerland; (J.S.); (A.S.); (D.P.S.)
| | - Christina Fasser
- Retina International, D02 TW98 Dublin, Ireland; Retina Suisse, 8005 Zürich, Switzerland;
| | - Andrea Suhner
- Novartis Pharma AG, 4056 Basel, Switzerland; (J.S.); (A.S.); (D.P.S.)
| | - Daniel P. Stiehl
- Novartis Pharma AG, 4056 Basel, Switzerland; (J.S.); (A.S.); (D.P.S.)
| | - Bee Chen
- Novartis Pharmaceutical Corporation, East Hanover, NJ 7936, USA;
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France;
- 15–20 Hôpital National de la Vision, National Rare Disease Center REFERET, INSERM-DGOS CIC1423, 75012 Paris, France
| | - Bart P. Leroy
- Department of Ophthalmology & Center for Medical Genetics Ghent, Ghent University & Ghent University Hospital, 9000 Ghent, Belgium;
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Jahnke L, Perrenoud V, Zandi S, Li Y, Conedera FM, Enzmann V. Modulation of Extracellular Matrix Composition and Chronic Inflammation with Pirfenidone Promotes Scar Reduction in Retinal Wound Repair. Cells 2024; 13:164. [PMID: 38247855 PMCID: PMC10814251 DOI: 10.3390/cells13020164] [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: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Wound repair in the retina is a complex mechanism, and a deeper understanding of it is necessary for the development of effective treatments to slow down or even prevent degenerative processes leading to photoreceptor loss. In this study, we harnessed a laser-induced retinal degeneration model (532-nm laser photocoagulation with 300 μm spot size, 60 ms duration and 60 mV pulse), enabling a profound molecular elucidation and a comprehensive, prolonged observation of the wound healing sequence in a murine laser-induced degeneration model (C57BL/6J mice, 6-12 weeks) until day 49 post-laser. Our observations included the expression of specific extracellular matrix proteins and myofibroblast activity, along with an analysis of gene expression related to extracellular matrix and adhesion molecules through RNA measurements. Furthermore, the administration of pirfenidone (10 mg/kg via drinking water), an anti-inflammatory and anti-fibrotic compound, was used to modulate scar formation after laser treatment. Our data revealed upregulated collagen expression in late regenerative phases and sustained inflammation in the damaged tissue. Notably, treatment with pirfenidone was found to mitigate scar tissue formation, effectively downregulating collagen production and diminishing the presence of inflammatory markers. However, it did not lead to the regeneration of the photoreceptor layer.
Collapse
Affiliation(s)
- Laura Jahnke
- Department of Ophthalmology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Virginie Perrenoud
- Department of Ophthalmology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Souska Zandi
- Department of Ophthalmology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Yuebing Li
- Department of Ophthalmology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Federica Maria Conedera
- Department of Ophthalmology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
22
|
Cioanca AV, Wooff Y, Aggio‐Bruce R, Sekar R, Dietrich C, Natoli R. Multiomic integration reveals neuronal-extracellular vesicle coordination of gliotic responses in degeneration. J Extracell Vesicles 2023; 12:e12393. [PMID: 38082562 PMCID: PMC10714032 DOI: 10.1002/jev2.12393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
In the central nervous system (CNS), including in the retina, neuronal-to-glial communication is critical for maintaining tissue homeostasis including signal transmission, transfer of trophic factors, and in the modulation of inflammation. Extracellular vesicle (EV)-mediated transport of molecular messages to regulate these processes has been suggested as a mechanism by which bidirectional communication between neuronal and glial cells can occur. In this work we employed multiomics integration to investigate the role of EV communication pathways from neurons to glial cells within the CNS, using the mouse retina as a readily accessible representative CNS tissue. Further, using a well-established model of degeneration, we aimed to uncover how dysregulation of homeostatic messaging between neurons and glia via EV can result in retinal and neurodegenerative diseases. EV proteomics, glia microRNA (miRNA) Open Array and small RNA sequencing, and retinal single cell sequencing were performed, with datasets integrated and analysed computationally. Results demonstrated that exogenous transfer of neuronal miRNA to glial cells was mediated by EV and occurred as a targeted response during degeneration to modulate gliotic inflammation. Taken together, our results support a model of neuronal-to-glial communication via EV, which could be harnessed for therapeutic targeting to slow the progression of retinal-, and neuro-degenerations of the CNS.
Collapse
Affiliation(s)
- Adrian V. Cioanca
- Clear Vision Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityCanberraAustralia
- School of Medicine and Psychology, College of Health and MedicineThe Australian National UniversityCanberraAustralia
| | - Yvette Wooff
- Clear Vision Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityCanberraAustralia
- School of Medicine and Psychology, College of Health and MedicineThe Australian National UniversityCanberraAustralia
| | - Riemke Aggio‐Bruce
- Clear Vision Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityCanberraAustralia
- School of Medicine and Psychology, College of Health and MedicineThe Australian National UniversityCanberraAustralia
| | - Rakshanya Sekar
- Clear Vision Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityCanberraAustralia
- School of Medicine and Psychology, College of Health and MedicineThe Australian National UniversityCanberraAustralia
| | - Catherine Dietrich
- Clear Vision Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityCanberraAustralia
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Riccardo Natoli
- Clear Vision Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityCanberraAustralia
- School of Medicine and Psychology, College of Health and MedicineThe Australian National UniversityCanberraAustralia
| |
Collapse
|
23
|
Becherucci V, Bacci GM, Marziali E, Sodi A, Bambi F, Caputo R. The New Era of Therapeutic Strategies for the Treatment of Retinitis Pigmentosa: A Narrative Review of Pathomolecular Mechanisms for the Development of Cell-Based Therapies. Biomedicines 2023; 11:2656. [PMID: 37893030 PMCID: PMC10604477 DOI: 10.3390/biomedicines11102656] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Retinitis pigmentosa, defined more properly as cone-rod dystrophy, is a paradigm of inherited diffuse retinal dystrophies, one of the rare diseases with the highest prevalence in the worldwide population and one of the main causes of low vision in the pediatric and elderly age groups. Advancements in and the understanding of molecular biology and gene-editing technologies have raised interest in laying the foundation for new therapeutic strategies for rare diseases. As a consequence, new possibilities for clinicians and patients are arising due to the feasibility of treating such a devastating disorder, reducing its complications. The scope of this review focuses on the pathomolecular mechanisms underlying RP better to understand the prospects of its treatment using innovative approaches.
Collapse
Affiliation(s)
- Valentina Becherucci
- Cell Factory Meyer, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (V.B.); (F.B.)
| | - Giacomo Maria Bacci
- Pediatric Ophthalmology Unit, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (E.M.); (R.C.)
| | - Elisa Marziali
- Pediatric Ophthalmology Unit, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (E.M.); (R.C.)
| | - Andrea Sodi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50139 Florence, Italy;
| | - Franco Bambi
- Cell Factory Meyer, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (V.B.); (F.B.)
| | - Roberto Caputo
- Pediatric Ophthalmology Unit, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (E.M.); (R.C.)
| |
Collapse
|
24
|
Jiménez-Loygorri JI, Benítez-Fernández R, Viedma-Poyatos Á, Zapata-Muñoz J, Villarejo-Zori B, Gómez-Sintes R, Boya P. Mitophagy in the retina: Viewing mitochondrial homeostasis through a new lens. Prog Retin Eye Res 2023; 96:101205. [PMID: 37454969 DOI: 10.1016/j.preteyeres.2023.101205] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial function is key to support metabolism and homeostasis in the retina, an organ that has one of the highest metabolic rates body-wide and is constantly exposed to photooxidative damage and external stressors. Mitophagy is the selective autophagic degradation of mitochondria within lysosomes, and can be triggered by distinct stimuli such as mitochondrial damage or hypoxia. Here, we review the importance of mitophagy in retinal physiology and pathology. In the developing retina, mitophagy is essential for metabolic reprogramming and differentiation of retina ganglion cells (RGCs). In basal conditions, mitophagy acts as a quality control mechanism, maintaining a healthy mitochondrial pool to meet cellular demands. We summarize the different autophagy- and mitophagy-deficient mouse models described in the literature, and discuss the potential role of mitophagy dysregulation in retinal diseases such as glaucoma, diabetic retinopathy, retinitis pigmentosa, and age-related macular degeneration. Finally, we provide an overview of methods used to monitor mitophagy in vitro, ex vivo, and in vivo. This review highlights the important role of mitophagy in sustaining visual function, and its potential as a putative therapeutic target for retinal and other diseases.
Collapse
Affiliation(s)
- Juan Ignacio Jiménez-Loygorri
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Rocío Benítez-Fernández
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland
| | - Álvaro Viedma-Poyatos
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Juan Zapata-Muñoz
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
25
|
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
|
26
|
Martínez-González J, Fernández-Carbonell Á, Cantó A, Gimeno-Hernández R, Almansa I, Bosch-Morell F, Miranda M, Olivar T. Sequences of Alterations in Inflammation and Autophagy Processes in Rd1 Mice. Biomolecules 2023; 13:1277. [PMID: 37759678 PMCID: PMC10527025 DOI: 10.3390/biom13091277] [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: 07/15/2023] [Revised: 08/10/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: the aim of this work was to study microglia and autophagy alterations in a one retinitis pigmentosa (RP) model at different stages of the disease (when rods are dying and later, when there are almost no rods, and cones are the cells that die. (2) Methods: rd1 mice were used and retinas obtained at postnatal days (PN) 11, 17, 28, 35, and 42. Iba1 (ionized calcium-binding adapter molecule 1) was the protein selected to study microglial changes. The macroautophagy markers Beclin-1, Atg5, Atg7, microtubule-associated protein light chain 3 (LC3), and lysosomal-associated membrane protein 2 (LAMP2) (involved in chaperone-mediated autophagy (CMA)) were determined. (3) Results: the expression of Iba1 was increased in rd1 retinas compared to the control group at PN17 (after the period of maximum rod death), PN28 (at the beginning of the period of cone death), and PN42. The number of activated (ameboid) microglial cells increased in the early ages of the retinal degeneration and the deactivated forms (branched cells) in more advanced ages. The macroautophagy markers Atg5 at PN11, Atg7 and LC3II at PN17, and Atg7 again at PN28 were decreased in rd1 retinas. At PN35 and PN42, the results reveal alterations in LAMP2A, a marker of CMA in the retina of rd1 mice. (4) Conclusions: we can conclude that during the early phases of retinal degeneration in the rd1 mouse, there is an alteration in microglia and a decrease in the macroautophagy cycle. Subsequently, the CMA is decreased and later on appears activated as a compensatory mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Teresa Olivar
- Department of Biomedical Sciences, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, 46115 Valencia, Spain; (J.M.-G.); (Á.F.-C.); (A.C.); (R.G.-H.); (I.A.); (F.B.-M.); (M.M.)
| |
Collapse
|
27
|
Samelska K, Szaflik JP, Guszkowska M, Kurowska AK, Zaleska-Żmijewska A. Characteristics of Rare Inherited Retinal Dystrophies in Adaptive Optics-A Study on 53 Eyes. Diagnostics (Basel) 2023; 13:2472. [PMID: 37568834 PMCID: PMC10417470 DOI: 10.3390/diagnostics13152472] [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: 05/16/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are genetic disorders that lead to the bilateral degeneration of the retina, causing irreversible vision loss. These conditions often manifest during the first and second decades of life, and their primary symptoms can be non-specific. Diagnostic processes encompass assessments of best-corrected visual acuity, fundoscopy, optical coherence tomography, fundus autofluorescence, fluorescein angiography, electrophysiological tests, and genetic testing. This study focuses on the application of adaptive optics (AO), a non-invasive retinal examination, for the assessment of patients with IRDs. AO facilitates the high-quality, detailed observation of retinal photoreceptor structures (cones and rods) and enables the quantitative analysis of parameters such as cone density (DM), cone spacing (SM), cone regularity (REG), and Voronoi analysis (N%6). AO examinations were conducted on eyes diagnosed with Stargardt disease (STGD, N=36), cone dystrophy (CD, N=9), and cone-rod dystrophy (CRD, N=8), and on healthy eyes (N=14). There were significant differences in the DM, SM, REG, and N%6 parameters between the healthy and IRD-affected eyes (p<0.001 for DM, SM, and REG; p=0.008 for N%6). The mean DM in the CD, CRD, and STGD groups was 8900.39/mm2, 9296.32/mm2, and 16,209.66/mm2, respectively, with a significant inter-group difference (p=0.006). The mean SM in the CD, CRD, and STGD groups was 12.37 μm, 14.82 μm, and 9.65 μm, respectively, with a significant difference observed between groups (p=0.002). However, no significant difference was found in REG and N%6 among the CD, CRD, and STGD groups. Significant differences were found in SM and DM between CD and STGD (p=0.014 for SM; p=0.003 for DM) and between CRD and STGD (p=0.027 for SM; p=0.003 for DM). Our findings suggest that AO holds significant potential as an impactful diagnostic tool for IRDs.
Collapse
Affiliation(s)
- Katarzyna Samelska
- Department of Ophthalmology, Medical University of Warsaw, 02-091 Warsaw, Poland
- SPKSO Ophthalmic University Hospital, 00-576 Warsaw, Poland
| | - Jacek Paweł Szaflik
- Department of Ophthalmology, Medical University of Warsaw, 02-091 Warsaw, Poland
- SPKSO Ophthalmic University Hospital, 00-576 Warsaw, Poland
| | | | - Anna Katarzyna Kurowska
- Department of Ophthalmology, Medical University of Warsaw, 02-091 Warsaw, Poland
- SPKSO Ophthalmic University Hospital, 00-576 Warsaw, Poland
| | - Anna Zaleska-Żmijewska
- Department of Ophthalmology, Medical University of Warsaw, 02-091 Warsaw, Poland
- SPKSO Ophthalmic University Hospital, 00-576 Warsaw, Poland
| |
Collapse
|
28
|
Xu P, Jiang YY, Morgan JIW. Cone Photoreceptor Morphology in Choroideremia Assessed Using Non-Confocal Split-Detection Adaptive Optics Scanning Light Ophthalmoscopy. Invest Ophthalmol Vis Sci 2023; 64:36. [PMID: 37504961 PMCID: PMC10383007 DOI: 10.1167/iovs.64.10.36] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023] Open
Abstract
Purpose Choroideremia (CHM) is an X-linked inherited retinal degeneration causing loss of the photoreceptors, retinal pigment epithelium, and choriocapillaris, although patients typically retain a central island of relatively preserved, functioning retina until late-stage disease. Here, we investigate cone photoreceptor morphology within the retained retinal island by examining cone inner segment area, density, circularity, and intercone space. Methods Using a custom-built, multimodal adaptive optics scanning light ophthalmoscope, nonconfocal split-detection images of the photoreceptor mosaic were collected at 1°, 2°, and 4° temporal to the fovea from 13 CHM and 12 control subjects. Cone centers were manually identified, and cone borders were segmented. A custom MATLAB script was used to extract area and circularity for each cone and calculate the percentage of intercone space in each region of interest. Bound cone density was also calculated. An unbalanced two-way ANOVA and Bonferroni post hoc tests were used to assess statistical differences between the CHM and control groups and along retinal eccentricity. Results Cone density was lower in the CHM group than in the control group (P < 0.001) and decreased with eccentricity from the fovea (P < 0.001). CHM cone inner segments were larger in area (P < 0.001) and more circular (P = 0.042) than those of the controls. Intercone space in CHM was also higher than in the controls (P < 0.001). Conclusions Cone morphology is altered in CHM compared to control, even within the centrally retained, functioning retinal area. Further studies are required to determine whether such morphology is a precursor to cone degeneration.
Collapse
Affiliation(s)
- Peiluo Xu
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Yu You Jiang
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jessica I. W. Morgan
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
29
|
Shi Y, Liu Y, Wu C, Liu X, Hu W, Yang Z, Li Z, Li Y, Deng C, Wei K, Gu C, Chen X, Su W, Zhuo Y. N,N-Dimethyl-3β-hydroxycholenamide attenuates neuronal death and retinal inflammation in retinal ischemia/reperfusion injury by inhibiting Ninjurin 1. J Neuroinflammation 2023; 20:91. [PMID: 37029422 PMCID: PMC10082498 DOI: 10.1186/s12974-023-02754-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/01/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Retinal ischemia-reperfusion (RIR) injury refers to an obstruction in the retinal blood supply followed by reperfusion. Although the molecular mechanism underlying the ischemic pathological cascade is not fully understood, neuroinflammation plays a crucial part in the mortality of retinal ganglion cells. METHODS Single-cell RNA sequencing (scRNA-seq), molecular docking, and transfection assay were used to explore the effectiveness and pathogenesis of N,N-dimethyl-3β-hydroxycholenamide (DMHCA)-treated mice with RIR injury and DMHCA-treated microglia after oxygen and glucose deprivation/reoxygenation (OGD/R). RESULTS DMHCA could suppress inflammatory gene expression and attenuate neuronal lesions, restoring the retinal structure in vivo. Using scRNA-seq on the retina of DMHCA-treated mice, we provided novel insights into RIR immunity and demonstrated nerve injury-induced protein 1 (Ninjurin1/Ninj 1) as a promising treatment target for RIR. Moreover, the expression of Ninj1, which was increased in RIR injury and OGD/R-treated microglia, was downregulated in the DMHCA-treated group. DMHCA suppressed the activation of the nuclear factor kappa B (NF-κB) pathways induced by OGD/R, which was undermined by the NF-κB pathway agonist betulinic acid. Overexpressed Ninj1 reversed the anti-inflammatory and anti-apoptotic function of DMHCA. Molecular docking indicated that for Ninj1, DMHCA had a low binding energy of - 6.6 kcal/mol, suggesting highly stable binding. CONCLUSION Ninj1 may play a pivotal role in microglia-mediated inflammation, while DMHCA could be a potential treatment strategy against RIR injury.
Collapse
Affiliation(s)
- Yunhong Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Yidan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Caiqing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Wenfei Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Zhenlan Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Zhidong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Yangyang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Caibin Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Kun Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Chenyang Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Xuhao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
30
|
Song DJ, Bao XL, Fan B, Li GY. Mechanism of Cone Degeneration in Retinitis Pigmentosa. Cell Mol Neurobiol 2023; 43:1037-1048. [PMID: 35792991 PMCID: PMC11414453 DOI: 10.1007/s10571-022-01243-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/13/2022] [Indexed: 11/27/2022]
Abstract
Retinitis pigmentosa (RP) is a group of genetic disorders resulting in inherited blindness due to the degeneration of rod and cone photoreceptors. The various mechanisms underlying rod degeneration primarily rely on genetic mutations, leading to night blindness initially. Cones gradually degenerate after rods are almost eliminated, resulting in varying degrees of visual disability and blindness. The mechanism of cone degeneration remains unclear. An understanding of the mechanisms underlying cone degeneration in RP, a highly heterogeneous disease, is essential to develop novel treatments of RP. Herein, we review recent advancements in the five hypotheses of cone degeneration, including oxidative stress, trophic factors, metabolic stress, light damage, and inflammation activation. We also discuss the connection among these theories to provide a better understanding of secondary cone degeneration in RP. Five current mechanisms of cone degenerations in RP Interactions among different pathways are involved in RP.
Collapse
Affiliation(s)
- De-Juan Song
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Xiao-Li Bao
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Bin Fan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Guang-Yu Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
31
|
Abstract
Vision is an ability that depends on the precise structure and functioning of the retina. Any kind of stress or injury can disrupt the retinal architecture and leads to vision impairment, vision loss, and blindness. Immune system and immune response function maintain homeostasis in the microenvironment. Several genetic, metabolic, and environmental factors may alter retinal homeostasis, and these events may initiate various inflammatory cascades. The prolonged inflammatory state may contribute to the initiation and development of retinal disorders such as glaucoma, age-related macular degeneration, diabetic retinopathy, and retinitis pigmentosa, which pose a threat to vision. In the current review, we attempted to provide sufficient evidence on the role of inflammation in these retinal disorders. Moreover, this review paves the way to focus on therapeutic targets of the disease, which are found to be promising.
Collapse
Affiliation(s)
- Geetika Kaur
- Integrative Biosciences Center, Wayne State University; Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikhlesh K Singh
- Integrative Biosciences Center, Wayne State University; Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
32
|
Wu KY, Kulbay M, Toameh D, Xu AQ, Kalevar A, Tran SD. Retinitis Pigmentosa: Novel Therapeutic Targets and Drug Development. Pharmaceutics 2023; 15:685. [PMID: 36840007 PMCID: PMC9963330 DOI: 10.3390/pharmaceutics15020685] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of hereditary diseases characterized by progressive degeneration of retinal photoreceptors leading to progressive visual decline. It is the most common type of inherited retinal dystrophy and has a high burden on both patients and society. This condition causes gradual loss of vision, with its typical manifestations including nyctalopia, concentric visual field loss, and ultimately bilateral central vision loss. It is one of the leading causes of visual disability and blindness in people under 60 years old and affects over 1.5 million people worldwide. There is currently no curative treatment for people with RP, and only a small group of patients with confirmed RPE65 mutations are eligible to receive the only gene therapy on the market: voretigene neparvovec. The current therapeutic armamentarium is limited to retinoids, vitamin A supplements, protection from sunlight, visual aids, and medical and surgical interventions to treat ophthalmic comorbidities, which only aim to slow down the progression of the disease. Considering such a limited therapeutic landscape, there is an urgent need for developing new and individualized therapeutic modalities targeting retinal degeneration. Although the heterogeneity of gene mutations involved in RP makes its target treatment development difficult, recent fundamental studies showed promising progress in elucidation of the photoreceptor degeneration mechanism. The discovery of novel molecule therapeutics that can selectively target specific receptors or specific pathways will serve as a solid foundation for advanced drug development. This article is a review of recent progress in novel treatment of RP focusing on preclinical stage fundamental research on molecular targets, which will serve as a starting point for advanced drug development. We will review the alterations in the molecular pathways involved in the development of RP, mainly those regarding endoplasmic reticulum (ER) stress and apoptotic pathways, maintenance of the redox balance, and genomic stability. We will then discuss the therapeutic approaches under development, such as gene and cell therapy, as well as the recent literature identifying novel potential drug targets for RP.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Merve Kulbay
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Dana Toameh
- Faculty of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - An Qi Xu
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Ananda Kalevar
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
33
|
Paliwal H, Prajapati BG, Srichana T, Singh S, Patel RJ. Novel Approaches in the Drug Development and Delivery Systems for Age-Related Macular Degeneration. Life (Basel) 2023; 13:life13020568. [PMID: 36836923 PMCID: PMC9960288 DOI: 10.3390/life13020568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The number of patients with ocular disorders has increased due to contributing factors such as aging populations, environmental changes, smoking, genetic abnormalities, etc. Age-related macular degeneration (AMD) is one of the common ocular disorders which may advance to loss of vision in severe cases. The advanced form of AMD is classified into two types, dry (non-exudative) and wet (exudative) AMD. Although several therapeutic approaches are explored for the management of AMD, no approved therapy can substantially slow down the progression of dry AMD into the later stages. The focus of researchers in recent times has been engaged in developing targeted therapeutic products to halt the progression and maintain or improve vision in individuals diagnosed with AMD. The delivery of anti-VEGF agents using intravitreal therapy has found some success in managing AMD, and novel formulation approaches have been introduced in various studies to potentiate the efficacy. Some of the novel approaches, such as hydrogel, microspheres, polymeric nanoparticles, liposomes, implants, etc. have been discussed. Apart from this, subretinal, suprachoroidal, and port delivery systems have also been investigated for biologics and gene therapies. The unmet potential of approved therapeutic products has contributed to several patent applications in recent years. This review outlines the current treatment options, outcomes of recent research studies, and patent details around the novel drug delivery approach for the treatment of AMD.
Collapse
Affiliation(s)
- Himanshu Paliwal
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana 384012, Gujarat, India
| | - Bhupendra Gopalbhai Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana 384012, Gujarat, India
- Correspondence: or ; Tel.: +91-9429225025
| | - Teerapol Srichana
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Ravish J. Patel
- Ramanbhai Patel College of Pharmacy (RPCP), Charotar University of Science and Technology, Anand 388421, Gujarat, India
| |
Collapse
|
34
|
Elseady WS, Keshk WA, Negm WA, Elkhalawany W, Elhanafy H, Ibrahim MAA, Radwan DA. Saffron extract attenuates Sofosbuvir-induced retinal neurodegeneration in albino rat. Anat Rec (Hoboken) 2023; 306:422-436. [PMID: 35451203 DOI: 10.1002/ar.24942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/27/2022] [Accepted: 04/11/2022] [Indexed: 01/25/2023]
Abstract
Sofosbuvir is a novel drug candidate for the treatment of hepatitis C viral infection; however, vision loss is one of its growing adverse effects. Saffron is a natural biomolecule with a high antioxidant potential that has been efficiently used in some diseases caused by oxidative stress. This study evaluated Sofosbuvir's neurodegenerative effect on the retina of albino rat and examined the potential protective role of saffron aqueous extract. Twenty-one adult male albino rats were randomly divided into three groups: Control, Sofosbuvir-treated (41.1 mg/kg /day for 6 weeks), and Sofosbuvir + Saffron co-treated groups. Retinal specimens were biochemically analyzed for malondialdehyde (MDA), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) levels. In addition, light and transmission electron microscopic examination, as well as immunohistochemical staining for Caspase-3, COX-2, and GFAP were performed. Sofosbuvir treatment caused a significant increase in retinal MDA, IL-6, and TNF-α levels coupling with a significant decrease in retinal total antioxidant capacity level. Histopathological findings revealed disturbed retinal architecture, detached pigment epithelium, vacuolated photoreceptors, in addition to a significant decrease in the thicknesses of both outer and inner nuclear layers, and the number of ganglionic cells. Ultrastructural examination revealed extensive degenerative changes in all retinal layers. Caspase-3, COX-2, and GFAP immunohistochemical expressions were significantly increased. Meanwhile, concomitant treatment with Saffron significantly improved retinal redox status, inflammation, histological, and ultrastructural parameters. Saffron may protect the retina from the hazardous effects of Sofosbuvir. Saffron could be used as an adjuvant therapy to protect patients receiving Sofosbuvir from retinal damage.
Collapse
Affiliation(s)
- Walaa S Elseady
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Walaa A Keshk
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Walaa A Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Walaa Elkhalawany
- Tropical Medicine & Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hend Elhanafy
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Marwa A A Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Doaa A Radwan
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
35
|
Cellular and Molecular Mechanisms of Pathogenesis Underlying Inherited Retinal Dystrophies. Biomolecules 2023; 13:biom13020271. [PMID: 36830640 PMCID: PMC9953031 DOI: 10.3390/biom13020271] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are congenital retinal degenerative diseases that have various inheritance patterns, including dominant, recessive, X-linked, and mitochondrial. These diseases are most often the result of defects in rod and/or cone photoreceptor and retinal pigment epithelium function, development, or both. The genes associated with these diseases, when mutated, produce altered protein products that have downstream effects in pathways critical to vision, including phototransduction, the visual cycle, photoreceptor development, cellular respiration, and retinal homeostasis. The aim of this manuscript is to provide a comprehensive review of the underlying molecular mechanisms of pathogenesis of IRDs by delving into many of the genes associated with IRD development, their protein products, and the pathways interrupted by genetic mutation.
Collapse
|
36
|
Ji Y, Zhao M, Qiao X, Peng GH. Decitabine improves MMS-induced retinal photoreceptor cell damage by targeting DNMT3A and DNMT3B. Front Mol Neurosci 2023; 15:1057365. [PMID: 36704326 PMCID: PMC9872157 DOI: 10.3389/fnmol.2022.1057365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Retinitis pigmentosa (RP) is a group of neurodegenerative retinopathies causing blindness due to progressive and irreversible photoreceptor cell death. The alkylating agent methyl methanesulfonate (MMS) can induce selective photoreceptor cell death, which is used to establish RP animal models. MMS induces DNA base damage by adding alkyl groups to DNA, and epigenetic modifications influence DNA damage response. Here, we aimed to explore the relationship between DNA methylation and DNA damage response in dying photoreceptors of RP. Methods The mouse RP model was established by a single intraperitoneal injection of MMS. The retinal structure and function were assessed by H&E, OCT, TUNEL, and ERG at several time points. The expression of DNA methylation regulators was assessed by qPCR and Western blot. DNMT inhibitor 5-aza-dC was applied to inhibit the activity of DNA methyltransferases and improve the retinal photoreceptor damage. Results The outer nuclear layer (ONL) and IS/OS layer were significantly thinner and the retinal function was impaired after MMS treatment. The cell death was mainly located in the ONL. The retinal damage induced by MMS was accompanied by hyperexpression of DNMT3A/3B. The application of DNMT inhibitor 5-aza-dC could suppress the expression level of DNMT3A/3B, resulting in the remission of MMS-induced photoreceptor cell damage. The ONL and IS/OS layers were thicker than that of the control group, and the retinal function was partially restored. This protective effect of 5-aza-dC was associated with the down-regulated expression of DNMT3A/3B. Conclusion These findings identified a functional role of DNMT3A/3B in MMS-induced photoreceptor cell damage and provided novel evidence to support DNMTs as potential therapeutic targets in retinal degenerative diseases.Graphical Abstract.
Collapse
Affiliation(s)
- Yanli Ji
- Laboratory of Visual Cell Differentiation and Regulation, Basic Medical College, Zhengzhou University, Zhengzhou, China,Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Meng Zhao
- Laboratory of Visual Cell Differentiation and Regulation, Basic Medical College, Zhengzhou University, Zhengzhou, China,Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Xiaomeng Qiao
- Department of Forensic Medicine, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Guang-Hua Peng
- Laboratory of Visual Cell Differentiation and Regulation, Basic Medical College, Zhengzhou University, Zhengzhou, China,Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, China,*Correspondence: Guang-Hua Peng, ✉
| |
Collapse
|
37
|
John MC, Quinn J, Hu ML, Cehajic-Kapetanovic J, Xue K. Gene-agnostic therapeutic approaches for inherited retinal degenerations. Front Mol Neurosci 2023; 15:1068185. [PMID: 36710928 PMCID: PMC9881597 DOI: 10.3389/fnmol.2022.1068185] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
Inherited retinal diseases (IRDs) are associated with mutations in over 250 genes and represent a major cause of irreversible blindness worldwide. While gene augmentation or gene editing therapies could address the underlying genetic mutations in a small subset of patients, their utility remains limited by the great genetic heterogeneity of IRDs and the costs of developing individualised therapies. Gene-agnostic therapeutic approaches target common pathogenic pathways that drive retinal degeneration or provide functional rescue of vision independent of the genetic cause, thus offering potential clinical benefits to all IRD patients. Here, we review the key gene-agnostic approaches, including retinal cell reprogramming and replacement, neurotrophic support, immune modulation and optogenetics. The relative benefits and limitations of these strategies and the timing of clinical interventions are discussed.
Collapse
Affiliation(s)
- Molly C. John
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Joel Quinn
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Monica L. Hu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
38
|
Olivares-González L, Velasco S, Campillo I, Millán JM, Rodrigo R. Redox Status in Retinitis Pigmentosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:443-448. [PMID: 37440070 DOI: 10.1007/978-3-031-27681-1_65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Retinitis pigmentosa (RP) is the most common form of inherited retinal dystrophy characterized by the progressive loss of vision. It is a rare disease. Despite being a genetic disease, its progression is influenced by oxidative damage and chemokines and cytokines released by the activated immune cells (e.g., macrophages or microglia). The role of oxidative stress is very important in the retina. Rods are the main consumers of oxygen (O2), so they are constantly exposed to oxidative stress and lipid peroxidation. According to the oxidative hypothesis, after rod death in the early stages of the disease, O2 would accumulate in large quantities in the retina, producing hyperoxia and favoring the accumulation of reactive oxygen species and reactive nitrogen species that would cause oxidative damage to lipids, proteins, and DNA, exacerbating the process of retinal degeneration. Evidence shows alterations in the antioxidant-oxidant state in patients and in animal models of RP. In recent years, therapeutic approaches aimed at reducing oxidative stress have emerged as useful therapies to slow down the progression of RP. We focus this review on oxidative stress and its relationship with the progression of RP.
Collapse
Affiliation(s)
- L Olivares-González
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| | - S Velasco
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| | - I Campillo
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| | - J M Millán
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER), Madrid, Spain
- Molecular, Cellular and Genomic Biomedicine, Health Research Institute La Fe, Valencia, Spain
| | - R Rodrigo
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain.
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain.
- Rare Diseases Networking Biomedical Research Centre (CIBERER), Madrid, Spain.
| |
Collapse
|
39
|
Völkner M, Wagner F, Kurth T, Sykes AM, Del Toro Runzer C, Ebner LJA, Kavak C, Alexaki VI, Cimalla P, Mehner M, Koch E, Karl MO. Modeling inducible neuropathologies of the retina with differential phenotypes in organoids. Front Cell Neurosci 2023; 17:1106287. [PMID: 37213216 PMCID: PMC10196395 DOI: 10.3389/fncel.2023.1106287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/06/2023] [Indexed: 05/23/2023] Open
Abstract
Neurodegenerative diseases remain incompletely understood and therapies are needed. Stem cell-derived organoid models facilitate fundamental and translational medicine research. However, to which extent differential neuronal and glial pathologic processes can be reproduced in current systems is still unclear. Here, we tested 16 different chemical, physical, and cell functional manipulations in mouse retina organoids to further explore this. Some of the treatments induce differential phenotypes, indicating that organoids are competent to reproduce distinct pathologic processes. Notably, mouse retina organoids even reproduce a complex pathology phenotype with combined photoreceptor neurodegeneration and glial pathologies upon combined (not single) application of HBEGF and TNF, two factors previously associated with neurodegenerative diseases. Pharmacological inhibitors for MAPK signaling completely prevent photoreceptor and glial pathologies, while inhibitors for Rho/ROCK, NFkB, and CDK4 differentially affect them. In conclusion, mouse retina organoids facilitate reproduction of distinct and complex pathologies, mechanistic access, insights for further organoid optimization, and modeling of differential phenotypes for future applications in fundamental and translational medicine research.
Collapse
Affiliation(s)
- Manuela Völkner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Felix Wagner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Alex M. Sykes
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Lynn J. A. Ebner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Cagri Kavak
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Vasileia Ismini Alexaki
- Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Dresden, Germany
| | - Peter Cimalla
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mirko Mehner
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Edmund Koch
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mike O. Karl
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- *Correspondence: Mike O. Karl, ,
| |
Collapse
|
40
|
Huang Y, Yuan L, He G, Cao Y, Deng X, Deng H. Novel compound heterozygous variants in the USH2A gene associated with autosomal recessive retinitis pigmentosa without hearing loss. Front Cell Dev Biol 2023; 11:1129862. [PMID: 36875754 PMCID: PMC9974670 DOI: 10.3389/fcell.2023.1129862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/25/2023] [Indexed: 02/17/2023] Open
Abstract
Background: Retinitis pigmentosa (RP) is a group of progressive inherited retinal dystrophies characterized by the primary degeneration of rod photoreceptors and the subsequent loss of cone photoreceptors because of cell death. It is caused by different mechanisms, including inflammation, apoptosis, necroptosis, pyroptosis, and autophagy. Variants in the usherin gene (USH2A) have been reported in autosomal recessive RP with or without hearing loss. In the present study, we aimed to identify causative variants in a Han-Chinese pedigree with autosomal recessive RP. Methods: A six-member, three-generation Han-Chinese family with autosomal recessive RP was recruited. A full clinical examination, whole exome sequencing, and Sanger sequencing, as well as co-segregation analysis were performed. Results: Three heterozygous variants in the USH2A gene, c.3304C>T (p.Q1102*), c.4745T>C (p.L1582P), and c.14740G>A (p.E4914K), were identified in the proband, which were inherited from parents and transmitted to the daughters. Bioinformatics analysis supported the pathogenicity of the c.3304C>T (p.Q1102*) and c.4745T>C (p.L1582P) variants. Conclusions: Novel compound heterozygous variants in the USH2A gene, c.3304C>T (p.Q1102*) and c.4745T>C (p.L1582P), were identified as the genetic causes of autosomal recessive RP. The findings may enhance the current knowledge of the pathogenesis of USH2A-associated phenotypes, expand the spectrum of the USH2A gene variants, and contribute to improved genetic counseling, prenatal diagnosis, and disease management.
Collapse
Affiliation(s)
- Yanxia Huang
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China.,Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| | - Lamei Yuan
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China.,Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| | - Guiyun He
- Department of Ophthalmology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yanna Cao
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China.,Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| | - Hao Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China.,Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China.,Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Weinberg J, Gaur M, Swaroop A, Taylor A. Proteostasis in aging-associated ocular disease. Mol Aspects Med 2022; 88:101157. [PMID: 36459837 PMCID: PMC9742340 DOI: 10.1016/j.mam.2022.101157] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
Abstract
Vision impairment has devastating consequences for the quality of human life. The cells and tissues associated with the visual process must function throughout one's life span and maintain homeostasis despite exposure to a variety of insults. Maintenance of the proteome is termed proteostasis, and is vital for normal cellular functions, especially at an advanced age. Here we describe basic aspects of proteostasis, from protein synthesis and folding to degradation, and discuss the current status of the field with a particular focus on major age-related eye diseases: age-related macular degeneration, cataract, and glaucoma. Our intent is to allow vision scientists to determine where and how to harness the proteostatic machinery for extending functional homeostasis in the aging retina, lens, and trabecular meshwork. Several common themes have emerged despite these tissues having vastly different metabolisms. Continued exposure to insults, including chronic stress with advancing age, increases proteostatic burden and reduces the fidelity of the degradation machineries including the ubiquitin-proteasome and the autophagy-lysosome systems that recognize and remove damaged proteins. This "double jeopardy" results in an exponential accumulation of cytotoxic proteins with advancing age. We conclude with a discussion of the challenges in maintaining an appropriate balance of protein synthesis and degradation pathways, and suggest that harnessing proteostatic capacities should provide new opportunities to design interventions for attenuating age-related eye diseases before they limit sight.
Collapse
Affiliation(s)
- Jasper Weinberg
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Mohita Gaur
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Allen Taylor
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
42
|
Chidlow G, Chan WO, Wood JPM, Casson RJ. Investigations into photoreceptor energy metabolism during experimental retinal detachment. Front Cell Neurosci 2022; 16:1036834. [PMID: 36467607 PMCID: PMC9716104 DOI: 10.3389/fncel.2022.1036834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/01/2022] [Indexed: 08/27/2023] Open
Abstract
Retinal detachment is a sight-threatening disorder, which occurs when the photoreceptors are separated from their vascular supply. The aim of the present study was to shed light on photoreceptor energy metabolism during experimental detachment in rats. Retinal detachment was induced in the eyes of rats via subretinal injection of sodium hyaluronate. Initially, we investigated whether detachment caused hypoxia within photoreceptors, as evaluated by the exogenous and endogenous biomarkers pimonidazole and HIF-1α, as well as by qPCR analysis of HIF target genes. The results showed no unequivocal staining for pimonidazole or HIF-1α within any detached retina, nor upregulation of HIF target genes, suggesting that any reduction in pO2 is of insufficient magnitude to produce hypoxia-induced covalent protein adducts or HIF-1α stabilisation. Subsequently, we analysed expression of cellular bioenergetic enzymes in photoreceptors during detachment. We documented loss of mitochondrial, and downregulation of glycolytic enzymes during detachment, indicating that photoreceptors have reduced energetic requirements and/or capacity. Given that detachment did not cause widespread hypoxia, but did result in downregulated expression of bioenergetic enzymes, we hypothesised that substrate insufficiency may be critical in terms of pathogenesis, and that boosting metabolic inputs may preserve photoreceptor bioenergetic production and, protect against their degeneration. Thus, we tested whether supplementation with the bioavailable energy substrate pyruvate mitigated rod and cone injury and degeneration. Despite protecting photoreceptors in culture from nutrient deprivation, pyruvate failed to protect against apoptotic death of rods, loss of cone opsins, and loss of inner segment mitochondria, in situ, when evaluated at 3 days after detachment. The regimen was also ineffective against cumulative photoreceptor deconstruction and degeneration when evaluated after 4 weeks. Retinal metabolism, particularly the bioenergetic profiles and pathological responses of the various cellular subtypes still presents a considerable knowledge gap that has important clinical consequences. While our data do not support the use of pyruvate supplementation as a means of protecting detached photoreceptors, they do provide a foundation and motivation for future research in this area.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
43
|
A 69 kb Deletion in chr19q13.42 including PRPF31 Gene in a Chinese Family Affected with Autosomal Dominant Retinitis Pigmentosa. J Clin Med 2022; 11:jcm11226682. [PMID: 36431159 PMCID: PMC9695658 DOI: 10.3390/jcm11226682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/20/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
We aimed to identify the genetic cause of autosomal dominant retinitis pigmentosa (adRP) and characterize the underlying molecular mechanisms of incomplete penetrance in a Chinese family affected with adRP. All enrolled family members underwent ophthalmic examinations. Whole-genome sequencing (WGS), multiplex ligation-dependent probe amplification (MLPA), linkage analysis and haplotype construction were performed in all participants. RNA-seq was performed to analyze the regulating mechanism of incomplete penetrance among affected patients, mutation carriers and healthy controls. In the studied family, 14 individuals carried a novel heterozygous large deletion of 69 kilobase (kb) in 19q13.42 encompassing exon 1 of the PRPF31 gene and five upstream genes: TFPT, OSCAR, NDUFA3, TARM1, and VSTM1. Three family members were sequenced and diagnosed as non-penetrant carriers (NPCs). RNA-seq showed significant differential expression of genes in deletion between mutation carriers and healthy control. The RP11 pedigree in this study was the largest pedigree compared to other reported RP11 pedigrees with large deletions. Early onset in all affected members in this pedigree was considered to be a special phenotype and was firstly reported in a RP11 family for the first time. Differential expression of PRPF31 between affected and unaffected subjects indicates a haploinsufficiency to cause the disease in the family. The other genes with significant differential expression might play a cooperative effect on the penetrance of RP11.
Collapse
|
44
|
Ren X, Léveillard T. Modulating antioxidant systems as a therapeutic approach to retinal degeneration. Redox Biol 2022; 57:102510. [PMID: 36274523 PMCID: PMC9596747 DOI: 10.1016/j.redox.2022.102510] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/21/2022] Open
Abstract
The human retina is facing a big challenge of reactive oxygen species (ROS) from endogenous and exogenous sources. Excessive ROS can cause damage to DNA, lipids, and proteins, triggering abnormal redox signaling, and ultimately lead to cell death. Thus, oxidative stress has been observed in inherited retinal diseases as a common hallmark. To counteract the detrimental effect of ROS, cells are equipped with various antioxidant defenses. In this review, we will focus on the antioxidant systems in the retina and how they can protect retina from oxidative stress. Both small antioxidants and antioxidant enzymes play a role in ROS removal. Particularly, the thioredoxin and glutaredoxin systems, as the major antioxidant systems in mammalian cells, exert functions in redox signaling regulation via modifying cysteines in proteins. In addition, the thioredoxin-like rod-derived cone viability factor (RdCVFL) and thioredoxin interacting protein (TXNIP) can modulate metabolism in photoreceptors and promote their survival. In conclusion, elevating the antioxidant capacity in retina is a promising therapy to curb the progress of inherited retinal degeneration.
Collapse
Affiliation(s)
- Xiaoyuan Ren
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden.
| | - Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
45
|
Völkner M, Wagner F, Steinheuer LM, Carido M, Kurth T, Yazbeck A, Schor J, Wieneke S, Ebner LJA, Del Toro Runzer C, Taborsky D, Zoschke K, Vogt M, Canzler S, Hermann A, Khattak S, Hackermüller J, Karl MO. HBEGF-TNF induce a complex outer retinal pathology with photoreceptor cell extrusion in human organoids. Nat Commun 2022; 13:6183. [PMID: 36261438 PMCID: PMC9581928 DOI: 10.1038/s41467-022-33848-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
Human organoids could facilitate research of complex and currently incurable neuropathologies, such as age-related macular degeneration (AMD) which causes blindness. Here, we establish a human retinal organoid system reproducing several parameters of the human retina, including some within the macula, to model a complex combination of photoreceptor and glial pathologies. We show that combined application of TNF and HBEGF, factors associated with neuropathologies, is sufficient to induce photoreceptor degeneration, glial pathologies, dyslamination, and scar formation: These develop simultaneously and progressively as one complex phenotype. Histologic, transcriptome, live-imaging, and mechanistic studies reveal a previously unknown pathomechanism: Photoreceptor neurodegeneration via cell extrusion. This could be relevant for aging, AMD, and some inherited diseases. Pharmacological inhibitors of the mechanosensor PIEZO1, MAPK, and actomyosin each avert pathogenesis; a PIEZO1 activator induces photoreceptor extrusion. Our model offers mechanistic insights, hypotheses for neuropathologies, and it could be used to develop therapies to prevent vision loss or to regenerate the retina in patients suffering from AMD and other diseases.
Collapse
Affiliation(s)
- Manuela Völkner
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Felix Wagner
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lisa Maria Steinheuer
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Madalena Carido
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Ali Yazbeck
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Jana Schor
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Stephanie Wieneke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lynn J A Ebner
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | | | - David Taborsky
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Katja Zoschke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Marlen Vogt
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Sebastian Canzler
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Andreas Hermann
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- Department of Neurology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Shahryar Khattak
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Jörg Hackermüller
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Department of Computer Science, Leipzig University, Leipzig, Germany
| | - Mike O Karl
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| |
Collapse
|
46
|
Li R, Zhang J, Wang Q, Cheng M, Lin B. TPM1 mediates inflammation downstream of TREM2 via the PKA/CREB signaling pathway. J Neuroinflammation 2022; 19:257. [PMID: 36241997 PMCID: PMC9563125 DOI: 10.1186/s12974-022-02619-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
Background Microglia, the innate immune cells in the central nervous system, play an essential role in brain homeostasis, neuroinflammation and brain infections. Dysregulated microglia, on the other hand, are associated with neurodegenerative diseases, yet the mechanisms underlying pro-inflammatory gene expression in microglia are incompletely understood. Methods We investigated the role of the actin-associated protein tropomyosin 1 (TPM1) in regulating pro-inflammatory phenotype of microglia in the retina by using a combination of cell culture, immunocytochemistry, Western blot, qPCR, TUNEL, RNA sequencing and electroretinogram analysis. TREM2−/− mice were used to investigate whether TPM1 regulated pro-inflammatory responses downstream of TREM2. To conditionally deplete microglia, we backcrossed CX3CR1CreER mice with Rosa26iDTR mice to generate CX3CR1CreER:Rosa26iDTR mice. Results We revealed a vital role for TPM1 in regulating pro-inflammatory phenotype of microglia. We found that TPM1 drove LPS-induced inflammation and neuronal death in the retina via the PKA/CREB pathway. TPM1 knockdown ameliorated LPS-induced inflammation in WT retinas yet exaggerated the inflammation in TREM2−/− retinas. RNA sequencing revealed that genes associated with M1 microglia and A1 astrocytes were significantly downregulated in LPS-treated WT retinas but upregulated in LPS-treated TREM2−/− retinas after TPM1 knockdown. Mechanistically, we demonstrated that CREB activated by TPM1 knockdown mediated anti-inflammatory genes in LPS-treated WT retinas but pro-inflammatory genes in LPS-treated TREM2−/− retinas, suggesting a novel role for TREM2 as a brake on TPM1-mediated inflammation. Furthermore, we identified that TPM1 regulated inflammation downstream of TREM2 and in a microglia-dependent manner. Conclusions We demonstrate that TPM1 mediates inflammation downstream of TREM2 via the PKA/CREB signaling pathway. Our findings suggest that TPM1 could be a potential target for therapeutic intervention in brain diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02619-3.
Collapse
Affiliation(s)
- Rong Li
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong. .,Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Shatin, Hong Kong.
| | - Jing Zhang
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Qiong Wang
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Shatin, Hong Kong
| | - Meng Cheng
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Bin Lin
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong. .,Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Shatin, Hong Kong. .,Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong.
| |
Collapse
|
47
|
Su T, Liang L, Zhang L, Wang J, Chen L, Su C, Cao J, Yu Q, Deng S, Chan HF, Tang S, Guo Y, Chen J. Retinal organoids and microfluidic chip-based approaches to explore the retinitis pigmentosa with USH2A mutations. Front Bioeng Biotechnol 2022; 10:939774. [PMID: 36185441 PMCID: PMC9524156 DOI: 10.3389/fbioe.2022.939774] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Retinitis pigmentosa (RP) is a leading cause of vision impairment and blindness worldwide, with limited medical treatment options. USH2A mutations are one of the most common causes of non-syndromic RP. In this study, we developed retinal organoids (ROs) and retinal pigment epithelium (RPE) cells from induced pluripotent stem cells (iPSCs) of RP patient to establish a sustainable in vitro RP disease model. RT-qPCR, western blot, and immunofluorescent staining assessments showed that USH2A mutations induced apoptosis of iPSCs and ROs, and deficiency of the extracellular matrix (ECM) components. Transcriptomics and proteomics findings suggested that abnormal ECM-receptor interactions could result in apoptosis of ROs with USH2A mutations via the PI3K-Akt pathway. To optimize the culture conditions of ROs, we fabricated a microfluidic chip to co-culture the ROs with RPE cells. Our results showed that this perfusion system could efficiently improve the survival rate of ROs. Further, ECM components such as laminin and collagen IV of ROs in the RP group were upregulated compared with those maintained in static culture. These findings illustrate the potential of microfluidic chip combined with ROs technology in disease modelling for RP.
Collapse
Affiliation(s)
- Ting Su
- Department of Ophthalmology, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Liying Liang
- Department of Ophthalmology, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Lan Zhang
- Department of Ophthalmology, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Jianing Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Luyin Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Caiying Su
- Department of Ophthalmology, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Jixing Cao
- Department of Ophthalmology, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Quan Yu
- Centric Laboratory, Medical College, Jinan University, Guangzhou, China
| | - Shuai Deng
- Institute for Tissue Engineering and Regenerative Medicine, Chinese University of Hong Kong, Hong Kong, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, Chinese University of Hong Kong, Hong Kong, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | | | - Yonglong Guo
- Department of Ophthalmology, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- *Correspondence: Jiansu Chen, ; Yonglong Guo,
| | - Jiansu Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Aier Eye Institute, Changsha, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
- *Correspondence: Jiansu Chen, ; Yonglong Guo,
| |
Collapse
|
48
|
Kovacs KD, Ciulla TA, Kiss S. Advancements in ocular gene therapy delivery: vectors and subretinal, intravitreal, and suprachoroidal techniques. Expert Opin Biol Ther 2022; 22:1193-1208. [PMID: 36062410 DOI: 10.1080/14712598.2022.2121646] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Ocular gene therapy represents fertile ground for rapid innovation, with ever-expanding therapeutic strategies, molecular targets, and indications. AREAS COVERED : Potential indications for ocular gene therapy have classically focused on inherited retinal disease (IRD), but more recently include acquired retinal diseases, such as neovascular age-related macular degeneration, geographic atrophy and diabetic retinopathy. Ocular gene therapy strategies have proliferated recently, and include gene augmentation, gene inactivation, gene editing, RNA modulation, and gene-independent gene augmentation. Viral vector therapeutic constructs include adeno-associated virus and lentivirus and continue to evolve through directed evolution and rationale design. Ocular gene therapy administration techniques have expanded beyond pars plana vitrectomy with subretinal injection to intravitreal injection and suprachoroidal injection. EXPERT OPINION : The success of treatment for IRD, paired with the promise of clinical research in acquired retinal diseases and in administration techniques, has raised the possibility of in-office gene therapy for common retinal disorders within the next five to ten years.
Collapse
Affiliation(s)
- Kyle D Kovacs
- Department of Ophthalmology, Retina Service, Weill Cornell Medical College, New York, NY, USA
| | | | - Szilárd Kiss
- Department of Ophthalmology, Retina Service, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
49
|
Ubiquitin Specific Protease USP48 Destabilizes NF-κB/p65 in Retinal Pigment Epithelium Cells. Int J Mol Sci 2022; 23:ijms23179682. [PMID: 36077078 PMCID: PMC9456453 DOI: 10.3390/ijms23179682] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022] Open
Abstract
Activation of NF-κB transcription factor is strictly regulated to accurately direct cellular processes including inflammation, immunity, and cell survival. In the retina, the modulation of the NF-κB pathway is essential to prevent excessive inflammatory responses, which plays a pivotal role in many retinal neurodegenerative diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), and inherited retinal dystrophies (IRDs). A critical cytokine mediating inflammatory responses in retinal cells is tumor necrosis factor-alpha (TNFα), leading to the activation of several transductional pathways, including NF-κB. However, the multiple factors orchestrating the appropriate regulation of NF-κB in retinal cells still remain unclear. The present study explores how the ubiquitin-specific protease 48 (USP48) downregulation impacts the stability and transcriptional activity of NF-κB/p65 in retinal pigment epithelium (RPE), at both basal conditions and following TNFα stimulation. We described that USP48 downregulation stabilizes p65. Notably, the accumulation of p65 is mainly detectable in the nuclear compartment and it is accompanied by an increased NF-κB transcriptional activity. These results delineate a novel role of USP48 in negatively regulating NF-κB in retinal cells, providing new opportunities for therapeutic intervention in retinal pathologies.
Collapse
|
50
|
Gegnaw ST, Sandu C, Mazzaro N, Mendoza J, Bergen AA, Felder-Schmittbuhl MP. Enhanced Robustness of the Mouse Retinal Circadian Clock Upon Inherited Retina Degeneration. J Biol Rhythms 2022; 37:567-574. [PMID: 35912966 DOI: 10.1177/07487304221112845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Daily biological rhythms are fundamental to retinal physiology and visual function. They are generated by a local circadian clock composed of a network of cell type/layer-specific, coupled oscillators. Animal models of retinal degeneration have been instrumental in characterizing the anatomical organization of the retinal clock. However, it is still unclear, among the multiple cell-types composing the retina, which ones are essential for proper circadian function. In this study, we used a previously well-characterized mouse model for autosomal dominant retinitis pigmentosa to examine the relationship between rod degeneration and the retinal circadian clock. This model carries the P23H mutation in rhodopsin, which induces mild rod degeneration in heterozygous and rapid loss of photoreceptors in homozygous genotypes. By measuring PER2::LUC bioluminescence rhythms, we show that the retinal clock in P23H/+ heterozygous mice displays circadian rhythms with significantly increased robustness and amplitude. By treating retinal explants with L-α aminoadipic acid, we further provide evidence that this enhanced rhythmicity might involve activation of Müller glial cells.
Collapse
Affiliation(s)
- Shumet T Gegnaw
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.,Departments of Clinical Genetics and Ophthalmology, University of Amsterdam, Amsterdam UMC, AMC, Amsterdam, The Netherlands
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Nadia Mazzaro
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Jorge Mendoza
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Arthur A Bergen
- Departments of Clinical Genetics and Ophthalmology, University of Amsterdam, Amsterdam UMC, AMC, Amsterdam, The Netherlands.,The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| |
Collapse
|