1
|
Simon CJ, Khabou H, Chaffiol A, Rucli M, Finzi M, Norberg N, Grimaud A, Mücher B, Desrosiers M, Sancho S, Bonilha VL, Grieve K, Duebel J, Paques M, Picaud S, Sahel JA, Audo I, Herlitze S, Dalkara D. Reactivating the phototransduction cascade with a mutation agnostic gene therapy preserves vision in rod-cone dystrophies. iScience 2025; 28:112106. [PMID: 40171489 PMCID: PMC11960651 DOI: 10.1016/j.isci.2025.112106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/01/2024] [Accepted: 01/21/2025] [Indexed: 04/03/2025] Open
Abstract
Rod-cone dystrophy (RCD) comprises genetic conditions where rod photoreceptor degeneration leads to cone loss, causing progressive vision loss. We investigated the phototransduction cascade in degenerating cones using two RCD mouse models and found that opsin and arrestin expression continues in the cell body during outer segment degeneration. Based on this observation, we explored reactivating cones through G-protein-coupled inwardly rectifying K (GIRK) channel expression. Using adeno-associated viral delivery of GIRK channels, we achieved improved visual function in both mouse models. Additionally, we examined human tissue from late-stage RCD patients and confirmed the presence of cone opsin and cone arrestin expression, supporting the potential therapeutic application of this approach. This GIRK-channel-based strategy offers a promising method to preserve high-quality vision in RCD patients, regardless of their specific genetic mutation.
Collapse
Affiliation(s)
- Cardillia-Joe Simon
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Hanen Khabou
- Gamut Therapeutics, 4 rue Thénard, 75005 Paris, France
| | - Antoine Chaffiol
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Marco Rucli
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Marion Finzi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Nat Norberg
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
| | - Anaïs Grimaud
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
| | - Brix Mücher
- Department of Zoology and Neurobiology, Ruhr University Bochum, 44780 Bochum, Germany
- Neuronal Circuits and Behaviour Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom
| | - Mélissa Desrosiers
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Serge Sancho
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
| | - Vera Lucia Bonilha
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Kate Grieve
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
| | - Jens Duebel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | - Michel Paques
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, 75012 Paris, France
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| |
Collapse
|
2
|
Kim DE, Kim S, Kim M, Min BK, Im M. Retinal degeneration increases inter-trial variabilities of light-evoked spiking activities in ganglion cells. Exp Eye Res 2025; 253:110305. [PMID: 39983973 DOI: 10.1016/j.exer.2025.110305] [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: 11/28/2024] [Revised: 02/03/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Retinal ganglion cells (RGCs) transmit visual information to the brain in the form of spike trains, which form visual perception. The reliabilities of spike timing and count are thought to play a crucial role in generating stable percepts. However, the effect of retinal degeneration on spike reproducibility remains underexplored. In this study, we examined longitudinal changes of both spike timing and count across different RGC types in response to repeated presentations of an identical light stimulus in retinal degeneration 10 (rd10) mice (B6.CXBl-Pde6brd10/J), a well-established model of retinitis pigmentosa (RP). We recorded the spiking responses of RGC populations to repeated white flashes using 256-channel multi-electrode array (MEA) at four rd10 age groups representing various stages of retinal degeneration. Our experimental results revealed a significant reduction in both spike timing and count consistencies compared to those in wild-type RGC recordings. Furthermore, the inter-trial variability patterns of different RGC types were found to differ throughout the degeneration process. For instance, when the spike time tiling coefficient (STTC) was used to evaluate inter-trial spike timing consistency, contrast-sensitive RGCs (ON, OFF, and ON-OFF types) exhibited a systematic decrease in spike timing consistency as degeneration progressed, whereas the remaining units did not show similar trends. Thus, we concluded that light-evoked spike trains become less consistent as degeneration progresses, with variability in spike timing and spike count varying across cell types. Given the critical role of spiking reliability in visual perception, our findings highlight the importance of accounting for cell type-specific degeneration patterns and inter-trial spiking inconsistencies when developing visual rehabilitation therapies to achieve enhanced performance. The underlying mechanism(s) driving the inter-trial spiking inconsistencies warrant further investigation.
Collapse
Affiliation(s)
- Da Eun Kim
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea; Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Sein Kim
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea
| | - Minju Kim
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea
| | - Byoung-Kyong Min
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Maesoon Im
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science & Technology, Seoul, Republic of Korea; KHU-KIST Department of Converging Science & Technology, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Corsi F, Galante A, Maggi MA, Mazziotti R, Bisti S, Piano I, Gargini C. The efficacy of Saffron Repron® in counteracting the progression of retinitis pigmentosa: Neuroprotection and resilience. Asia Pac J Ophthalmol (Phila) 2025:100192. [PMID: 40122468 DOI: 10.1016/j.apjo.2025.100192] [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: 02/02/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
The mutations observed in the various forms of retinitis pigmentosa (RP) affect genes coding for rod-specific proteins and direct the progression of different forms of dystrophy toward total blindness. The present investigation aims to explore the protective effects of Saffron Repron®, in a mouse model of RP. Saffron was administered orally to pregnant females and weaned pups for 120 days. At different time points (P30, P60, P90, and P120), visual function, retinal function, cone lifespan, morphology, gene expression, and protein level were analyzed. The results indicate that chronic saffron treatment effectively slows the progression of long-term damage caused by genetic mutations in both the morphology and function of retinal neurons. Cellular mechanisms responsible for this action appear complex and, probably, due to coordinated and synergistic activities by its chemical components. Here we provide evidence that saffron is able to modulate the epigenetic pathway involved in neuroinflammation. Biochemical and molecular measures suggest that early saffron treatment may induce a form of adaptation known as acquired resilience.
Collapse
Affiliation(s)
- Francesca Corsi
- University of Pisa, Department of Pharmacy, Italy; National Institute of Biostructure and Biosystem (INBB), Italy
| | | | - Maria Anna Maggi
- Hortus Novus srl, via Campo Sportivo 2, Italy; Department of Physical and Chemical Sciences, University of L'Aquila, Italy
| | - Raffaele Mazziotti
- Department of Neuroscience, Psychology, Pharmacology, and Child Health, University of Florence, Italy; Institute of Neuroscience, National Research Council (CNR), Italy
| | - Silvia Bisti
- National Institute of Biostructure and Biosystem (INBB), Italy
| | - Ilaria Piano
- University of Pisa, Department of Pharmacy, Italy; National Institute of Biostructure and Biosystem (INBB), Italy.
| | - Claudia Gargini
- University of Pisa, Department of Pharmacy, Italy; National Institute of Biostructure and Biosystem (INBB), Italy
| |
Collapse
|
4
|
Bernardo-Colón A, Bighinati A, Parween S, Debnath S, Piano I, Adani E, Corsi F, Gargini C, Vergara N, Marigo V, Patricia Becerra S. H105A peptide eye drops promote photoreceptor survival in murine and human models of retinal degeneration. COMMUNICATIONS MEDICINE 2025; 5:81. [PMID: 40118996 PMCID: PMC11928584 DOI: 10.1038/s43856-025-00789-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 02/27/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Photoreceptor death leads to inherited blinding retinal diseases, such as retinitis pigmentosa (RP). As disease progression often outpaces therapeutic advances, developing effective treatments is urgent. This study evaluates the efficacy of small peptides derived from pigment epithelium-derived factor (PEDF), which are known to restrict common cell death pathways associated with retinal diseases. METHODS We tested chemically synthesized peptides (17-mer and H105A) with affinity for the PEDF receptor, PEDF-R, delivered as eye drops to two RP mouse models: rd10 (phosphodiesterase 6b mutation) and RhoP23H/+ (rhodopsin P23H mutation). Additionally, we engineered AAV-H105A vectors for intravitreal delivery in RhoP23H/+ mice. To assess peptide effects in human tissue, we used retinal organoids exposed to cigarette smoke extract, a model of oxidative stress. Photoreceptor survival, morphology and function were evaluated. RESULTS Here we show that peptides 17-mer and H105A delivered via eye drops successfully reach the retina, promote photoreceptor survival, and improve retinal function in both RP mouse models. Intravitreal delivery of a AAV-H105A vector delays photoreceptor degeneration in RhoP23H/+ mice up to six months. In human retinal organoids, peptide H105A specifically prevents photoreceptor death induced by oxidative stress, a contributing factor to RP progression. CONCLUSIONS PEDF peptide-based eye drops offer a promising, minimally invasive therapy to prevent photoreceptor degeneration in retinal disorders, with a favorable safety profile.
Collapse
Grants
- Z01 EY000306 Intramural NIH HHS
- Intramural Research Program of the National Eye Institute, National Institutes of Health, United States of America (Project #EY000306, SPB); the Prevention of Blindness Society (SPB); Fondazione Telethon (Project #GGP19113, VM), the National Center for “Gene Therapy and Drugs based on RNA Technology” cod. Progetto CN00000041 and “Health Extended Alliance for Innovative Therapies, Advanced Lab-research, and Integrated Approaches of Precision Medicine - HEAL ITALIA” tematica 6 “Innovative diagnostics and therapies in precision medicine” cod. Progetto PE0000019 PIANO NAZIONALE DI RIPRESA E RESILIENZA (PNRR) – MISSIONE 4 “Istruzione Ricerca” COMPONENTE 2, “Dalla ricerca all’impresa” INVESTIMENTO 1.4, “Potenziamento strutture di ricerca e creazione di "campioni nazionali di R&S” su alcune Key enabling technologies”, finanziato dall’Unione europea – NextGenerationEU (VM and AB); The CellSight Development Fund (NV); and a Challenge Grant to the Department of Ophthalmology at the University of Colorado from Research to Prevent Blindness (NV).
Collapse
Affiliation(s)
- Alexandra Bernardo-Colón
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Bighinati
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Shama Parween
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - Subrata Debnath
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Elisa Adani
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Francesca Corsi
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
- Gates Center for Regenerative Medicine, Linda Crnic Institute for Down Syndrome and University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy.
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
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
|
6
|
Thompson SL, Crowder SM, Hekmatara M, Sechrest ER, Deng WT, Robichaux MA. P23H rhodopsin aggregation in the ER causes synaptic protein imbalance in rod photoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619115. [PMID: 39484588 PMCID: PMC11526887 DOI: 10.1101/2024.10.18.619115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Rod photoreceptor neurons in the retina detect scotopic light through the visual pigment rhodopsin (Rho) in their outer segments (OS). Efficient Rho trafficking to the OS through the inner rod compartments is critical for long-term rod health. Given the importance of protein trafficking to the OS, less is known about the trafficking of rod synaptic proteins. Furthermore, the subcellular impact of Rho mislocalization on rod synapses (i.e., "spherules") has not been investigated. In this study we used super-resolution and electron microscopies, along with proteomics, to perform a subcellular analysis of Rho synaptic mislocalization in P23H-Rho-RFP mutant mice. We discovered that mutant P23H-Rho-RFP protein mislocalized in distinct ER aggregations within the spherule cytoplasm, which we confirmed with AAV overexpression. Additionally, we found synaptic protein abundance differences in P23H-Rho-RFP mice. By comparison, Rho mislocalized along the spherule plasma membrane in WT and rd10 mutant rods, in which there was no synaptic protein disruption. Throughout the study, we also identified a network of ER membranes within WT rod presynaptic spherules. Together, our findings indicate that photoreceptor synaptic proteins are sensitive to ER dysregulation.
Collapse
Affiliation(s)
- Samantha L Thompson
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Sophie M Crowder
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Maryam Hekmatara
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Emily R Sechrest
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Wen-Tao Deng
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Michael A Robichaux
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV 26506, United States
| |
Collapse
|
7
|
Liu S, Matsuo T, Matsuo C, Abe T, Chen J, Sun C, Zhao Q. Perspectives of traditional herbal medicines in treating retinitis pigmentosa. Front Med (Lausanne) 2024; 11:1468230. [PMID: 39712182 PMCID: PMC11660805 DOI: 10.3389/fmed.2024.1468230] [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: 07/21/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Medicinal plants, also known as herbs, have been discovered and utilized in traditional medical practice since prehistoric times. Medicinal plants have been proven rich in thousands of natural products that hold great potential for the development of new drugs. Previously, we reviewed the types of Chinese traditional medicines that a Tang Dynasty monk Jianzhen (Japanese: Ganjin) brought to Japan from China in 742. This article aims to review the origin of Kampo (Japanese traditional medicine), and to present the overview of neurodegenerative diseases and retinitis pigmentosa as well as medicinal plants in some depth. Through the study of medical history of the origin of Kampo, we found that herbs medicines contain many neuroprotective ingredients. It provides us a new perspective on extracting neuroprotective components from herbs medicines to treat neurodegenerative diseases. Retinitis pigmentosa (one of the ophthalmic neurodegenerative diseases) is an incurable blinding disease and has become a popular research direction in global ophthalmology. To date, treatments for retinitis pigmentosa are very limited worldwide. Therefore, we intend to integrate the knowledge and skills from different disciplines, such as medical science, pharmaceutical science and plant science, to take a new therapeutic approach to treat neurodegenerative diseases. In the future, we will use specific active ingredients extracted from medicinal plants to treat retinitis pigmentosa. By exploring the potent bioactive ingredients present in medicinal plants, a valuable opportunity will be offered to uncover novel approaches for the development of drugs which target for retinitis pigmentosa.
Collapse
Affiliation(s)
- Shihui Liu
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Toshihiko Matsuo
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
- Department of Ophthalmology, Okayama University Hospital, Okayama, Japan
| | - Chie Matsuo
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Takumi Abe
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jinghua Chen
- Department of Ophthalmology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Qing Zhao
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
8
|
Dibas A, Batabyal S, Kim S, Carlson M, Mohanty S, Sharif NA. Efficacy of Intravitreal Multi-Characteristic Opsin (MCO-010) Optogenetic Gene Therapy in a Mouse Model of Leber Congenital Amaurosis. J Ocul Pharmacol Ther 2024; 40:702-708. [PMID: 39441604 DOI: 10.1089/jop.2024.0084] [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] [Indexed: 10/25/2024] Open
Abstract
Purpose: Leber congenital amaurosis (LCA) is a sight-threatening inherited retinal disorder (IRD) caused by numerous genetic mutations. Multi-characteristic opsin (MCO)-based optogenetic therapy allows the recruitment of residual cells of the retina in LCA for alternative vision transduction while being mutation-agnostic. Using rd12 mice, we investigated the in vivo efficacy of an adeno-associated virus2 (AAV2)-transduced ambient light-activatable MCO (MCO-010) containing a metabotropic glutamate receptor-6 bipolar cell-specific promoter/enhancer. Methods: Mice requiring > 40 s to reach and board a dimly lit hidden platform in a water-maze were selected and randomly divided into 2 cohorts. These mice were intravitreally (IVT) injected with either 1.7E9 gene copies/eye of MCO-010 or control AAV2 and re-tested in the water-maze. Spectral-domain optical coherence tomography (SD-OCT), hematoxylin and eosin staining of retinas, and electroretinographic (ERG) studies were also conducted. Results: Safety of MCO-010 in rd12 mice was confirmed by the lack of significant detrimental changes in the mouse behavior, b-wave amplitudes and in retinal thickness. rd12 control mice performed relatively poorly in the water-maze test requiring ≥ 30-60 s to find and board the platform. MCO-010-treated rd12 mice reached the platform much faster than the AAV2-treated rd12 mice, with some mice only requiring < 5 s to achieve this goal (P < 0.01-0.0024). Conclusions: IVT MCO-010 treatment was well tolerated by rd12 mice, and it prevented the decrease in retinal thickness, and preserved ERG parameters. It also significantly improved the vision in rd12 mice relative to control AAV2-injected mice. MCO-010 therefore represents a novel and efficacious optogenetic therapeutic to treat LCA and other IRDs irrespective of the genetic defect(s).
Collapse
Affiliation(s)
- Adnan Dibas
- Nanoscope Technologies LLC, Bedford, Texas, USA
| | | | | | | | - Samarendra Mohanty
- Nanoscope Technologies LLC, Bedford, Texas, USA
- Nanoscope Therapeutics Inc, Dallas, Texas, USA
| | | |
Collapse
|
9
|
Batabyal S, Kim S, Carlson M, Narcisse D, Tchedre K, Dibas A, Sharif NA, Mohanty S. Multi-Characteristic Opsin Therapy to Functionalize Retina, Attenuate Retinal Degeneration, and Restore Vision in Mouse Models of Retinitis Pigmentosa. Transl Vis Sci Technol 2024; 13:25. [PMID: 39412768 PMCID: PMC11486081 DOI: 10.1167/tvst.13.10.25] [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: 12/13/2023] [Accepted: 08/13/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose Retinal degeneration 1 and 10 (rd1 and rd10) mice are useful animal models of retinitis pigmentosa (RP) with rapidly and slowly progressive pathologies, respectively. Our study aims were to determine the effect of adeno-associated viral vector 2 (AAV2)-delivered multi-characteristic opsin (MCO-010; under the control of a metabotropic glutamate receptor-6 promoter enhancer) on the morphological and functional characteristics of vision in both rd1 and rd10 mice. Methods Various retinal measures of MCO-010 transduction and electrophysiological, behavioral, and other routine blood analyses were performed in the rd1 and/or rd10 mice after intravitreal injection of 1 µL of MCO-010 or AAV2 vehicle. Functional tests included electroretinogram, visually evoked potential, and behavior assay (optomotor and water maze). Retinal thickness, intraocular pressure, and plasma cytokine levels were also determined. Results Following intravitreal MCO-010 injection, approximately 80% of bipolar cells were transduced in the retina, and no alterations in retinal thickness were observed at 4 months post-injection. However, retinal thickness significantly decreased in control mice. MCO-010 treatment increased head movements and induced faster navigation of mice to the platform in a water-maze test. The MCO-010 gene therapy helped preserve visually evoked electrical response in the retina and visual cortex. No ocular toxicity, immunotoxicity, or phototoxicity was observed in the MCO-010-treated mice, even under chronic intense light conditions. Conclusions Intravitreal MCO-010 was well tolerated in rd1 and rd10 mice models of RP, and it appeared to attenuate retinal photoreceptor degeneration based on retinal structure and functional outcome measures. Translational Relevance As reported here, optogenetic treatment of the inner retina attenuates further retinal degeneration in addition to photosensitizing higher order neurons, and this disease-modifying aspect should be evaluated in optogenetic clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Adnan Dibas
- Nanoscope Technologies LLC, Bedford, TX, USA
| | | | - Samarendra Mohanty
- Nanoscope Technologies LLC, Bedford, TX, USA
- Nanoscope Therapeutics, Inc., Dallas, TX, USA
| |
Collapse
|
10
|
Napoli D, Orsini N, Salamone G, Calvello MA, Capsoni S, Cattaneo A, Strettoi E. Human NGF "Painless" Ocular Delivery for Retinitis Pigmentosa: An In Vivo Study. eNeuro 2024; 11:ENEURO.0096-24.2024. [PMID: 39293937 PMCID: PMC11412101 DOI: 10.1523/eneuro.0096-24.2024] [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: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 09/20/2024] Open
Abstract
Retinitis pigmentosa (RP) is a family of genetically heterogeneous diseases still without a cure. Despite the causative genetic mutation typically not expressed in cone photoreceptors, these cells inevitably degenerate following the primary death of rods, causing blindness. The reasons for the "bystander" degeneration of cones are presently unknown but decrement of survival factors, oxidative stress, and inflammation all play a role. Targeting these generalized biological processes represents a strategy to develop mutation-agnostic therapies for saving vision in large populations of RP individuals. A classical method to support neuronal survival is by employing neurotrophic factors, such as NGF. This study uses painless human NGF (hNGFp), a TrkA receptor-biased variant of the native molecule with lower affinity for nociceptors and limited activity as a pain inducer; the molecule has identical neurotrophic power of the native form but a reduced affinity for the p75NTR receptors, known to trigger apoptosis. hNGFp has a recognized activity on brain microglial cells, which are induced to a phenotype switch from a highly activated to a more homeostatic configuration. hNGFp was administered to RP-like mice in vivo with the aim of decreasing retinal inflammation and also providing retinal neuroprotection. However, the ability of this treatment to counteract the bystander degeneration of cones remained limited.
Collapse
Affiliation(s)
- Debora Napoli
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | - Noemi Orsini
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | | | | | - Simona Capsoni
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), Roma 00161, Italy
| | | |
Collapse
|
11
|
Yang M, Yao J, Jia L, Kocab AJ, Zacks DN. Preservation of retinal structure and function in two mouse models of inherited retinal degeneration by ONL1204, an inhibitor of the Fas receptor. Cell Death Dis 2024; 15:576. [PMID: 39117629 PMCID: PMC11310419 DOI: 10.1038/s41419-024-06970-6] [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: 04/15/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Due to the large number of genes and mutations that result in inherited retinal degenerations (IRD), there has been a paucity of therapeutic options for these patients. There is a large unmet need for therapeutic approaches targeting shared pathophysiologic pathways in a mutation-independent manner. The Fas receptor is a major activator and regulator of retinal cell death and inflammation in a variety of ocular diseases. We previously reported the activation of Fas-mediated photoreceptor (PR) cell death in two different IRD mouse models, rd10 and P23H, and demonstrated the protective effect of genetic Fas inhibition. The purpose of this study was to examine the effects of pharmacologic inhibition of Fas in these two models by intravitreal injection with a small peptide inhibitor of the Fas receptor, ONL1204. A single intravitreal injection of ONL1204 was given to one eye of rd10 mice at P14. Two intravitreal injections of ONL1204 were given to the P23H mice, once at P14 and again at 2-months of age. The fellow eyes were injected with vehicle alone. Fas activation, rate of PR cell death, retinal function, and the activation of immune cells in the retina were evaluated. In both rd10 and P23H mice, ONL1204 treatment resulted in decreased number of TUNEL (+) PRs, decreased caspase 8 activity, enhanced photoreceptor cell counts, and improved visual function compared with vehicle treated fellow eyes. Treatment with ONL1204 also reduced immune cell activation in the retinas of both rd10 and P23H mice. The protective effect of pharmacologic inhibition of Fas by ONL1204 in two distinct mouse models of retinal degeneration suggests that targeting this common pathophysiologic mechanism of cell death and inflammation represents a potential therapeutic approach to preserve the retina in patients with IRD, regardless of the genetic underpinning.
Collapse
Affiliation(s)
- Mengling Yang
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
- Eye Center of Xiangya Hospital, Xiangya School of medicine, Central South University, Changsha, Hunan, China
| | - Jingyu Yao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
| | - Lin Jia
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
| | | | - David N Zacks
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Bernardo-Colón A, Bighinati A, Parween S, Debnath S, Piano I, Adani E, Corsi F, Gargini C, Vergara N, Marigo V, Becerra SP. H105A peptide eye drops promote photoreceptor survival in murine and human models of retinal degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602890. [PMID: 39109177 PMCID: PMC11302621 DOI: 10.1101/2024.07.10.602890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Photoreceptor death causes blinding inheritable retinal diseases, such as retinitis pigmentosa (RP). As disease progression often outpaces therapeutic advances, finding effective treatments is urgent. This study focuses on developing a targeted approach by evaluating the efficacy of small peptides derived from pigment epithelium-derived factor (PEDF), known to restrict common cell death pathways associated with retinal diseases. Peptides with affinity for the PEDF receptor, PEDF-R, (17-mer and H105A) delivered via eye drops reached the retina, efficiently promoted photoreceptor survival, and improved retinal function in RP mouse models based on both the rd10 mutation and the rhodopsin P23H mutation. Additionally, intravitreal delivery of AAV-H105A vectors delayed photoreceptor degeneration in the latter RP mouse model. Furthermore, peptide H105A specifically prevented photoreceptor death induced by oxidative stress, a contributing factor to RP progression, in human retinal organoids. This promising approach for peptide eye drop delivery holds significant potential as a therapeutic for preventing photoreceptor death in retinal disorders, offering a high safety profile, low invasiveness and multiple delivery options.
Collapse
Affiliation(s)
- Alexandra Bernardo-Colón
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health; Bethesda, MD, USA
| | - Andrea Bighinati
- Department of Life Sciences, University of Modena and Reggio Emilia; 41125 Modena, Italy
| | - Shama Parween
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - Subrata Debnath
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health; Bethesda, MD, USA
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa; 56126 Pisa, Italy
| | - Elisa Adani
- Department of Life Sciences, University of Modena and Reggio Emilia; 41125 Modena, Italy
| | - Francesca Corsi
- Department of Pharmacy, University of Pisa; 56126 Pisa, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa; 56126 Pisa, Italy
| | - Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
- Gates Center for Regenerative Medicine, Linda Crnic Institute for Down Syndrome and University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia; 41125 Modena, Italy
| | - S. Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health; Bethesda, MD, USA
| |
Collapse
|
13
|
Borchert GA, Shamsnajafabadi H, Ng BWJ, Xue K, De Silva SR, Downes SM, MacLaren RE, Cehajic-Kapetanovic J. Age-related macular degeneration: suitability of optogenetic therapy for geographic atrophy. Front Neurosci 2024; 18:1415575. [PMID: 39010943 PMCID: PMC11246919 DOI: 10.3389/fnins.2024.1415575] [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: 04/10/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Age-related macular degeneration (AMD) is a growing public health concern given the aging population and it is the leading cause of blindness in developed countries, affecting individuals over the age of 55 years. AMD affects the retinal pigment epithelium (RPE) and Bruch's membrane in the macula, leading to secondary photoreceptor degeneration and eventual loss of central vision. Late AMD is divided into two forms: neovascular AMD and geographic atrophy (GA). GA accounts for around 60% of late AMD and has been the most challenging subtype to treat. Recent advances include approval of new intravitreally administered therapeutics, pegcetacoplan (Syfovre) and avacincaptad pegol (Iveric Bio), which target complement factors C3 and C5, respectively, which slow down the rate of enlargement of the area of atrophy. However, there is currently no treatment to reverse the central vision loss associated with GA. Optogenetics may provide a strategy for rescuing visual function in GA by imparting light-sensitivity to the surviving inner retina (i.e., retinal ganglion cells or bipolar cells). It takes advantage of residual inner retinal architecture to transmit visual stimuli along the visual pathway, while a wide range of photosensitive proteins are available for consideration. Herein, we review the anatomical changes in GA, discuss the suitability of optogenetic therapeutic sensors in different target cells in pre-clinical models, and consider the advantages and disadvantages of different routes of administration of therapeutic vectors.
Collapse
Affiliation(s)
- Grace A. Borchert
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Hoda Shamsnajafabadi
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Benjamin W. J. Ng
- Nuffield Laboratory of Ophthalmology, 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, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Samantha R. De Silva
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Susan M. Downes
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
14
|
Pfeffer ME, DiFrancesco ML, Marchesi A, Galluzzi F, Moschetta M, Rossini A, Francia S, Franz CM, Fok Y, Valotteau C, Paternò GM, Redondo Morata L, Vacca F, Mattiello S, Magni A, Maragliano L, Beverina L, Mattioli G, Lanzani G, Baldelli P, Colombo E, Benfenati F. Nanoactuator for Neuronal Optoporation. ACS NANO 2024; 18:12427-12452. [PMID: 38687909 DOI: 10.1021/acsnano.4c01672] [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: 05/02/2024]
Abstract
Light-driven modulation of neuronal activity at high spatial-temporal resolution is becoming of high interest in neuroscience. In addition to optogenetics, nongenetic membrane-targeted nanomachines that alter the electrical state of the neuronal membranes are in demand. Here, we engineered and characterized a photoswitchable conjugated compound (BV-1) that spontaneously partitions into the neuronal membrane and undergoes a charge transfer upon light stimulation. The activity of primary neurons is not affected in the dark, whereas millisecond light pulses of cyan light induce a progressive decrease in membrane resistance and an increase in inward current matched to a progressive depolarization and action potential firing. We found that illumination of BV-1 induces oxidation of membrane phospholipids, which is necessary for the electrophysiological effects and is associated with decreased membrane tension and increased membrane fluidity. Time-resolved atomic force microscopy and molecular dynamics simulations performed on planar lipid bilayers revealed that the underlying mechanism is a light-driven formation of pore-like structures across the plasma membrane. Such a phenomenon decreases membrane resistance and increases permeability to monovalent cations, namely, Na+, mimicking the effects of antifungal polyenes. The same effect on membrane resistance was also observed in nonexcitable cells. When sustained light stimulations are applied, neuronal swelling and death occur. The light-controlled pore-forming properties of BV-1 allow performing "on-demand" light-induced membrane poration to rapidly shift from cell-attached to perforated whole-cell patch-clamp configuration. Administration of BV-1 to ex vivo retinal explants or in vivo primary visual cortex elicited neuronal firing in response to short trains of light stimuli, followed by activity silencing upon prolonged light stimulations. BV-1 represents a versatile molecular nanomachine whose properties can be exploited to induce either photostimulation or space-specific cell death, depending on the pattern and duration of light stimulation.
Collapse
Affiliation(s)
- Marlene E Pfeffer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132 Genova, Italy
| | | | - Arin Marchesi
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Torrette di Ancona, Italy
| | - Filippo Galluzzi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Via Morego 30, 16163 Genova, Italy
| | - Matteo Moschetta
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
| | - Andrea Rossini
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
| | - Simona Francia
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Clemens M Franz
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yulia Fok
- Aix-Marseille University, INSERM, DyNaMo, Turing Centre for Living Systems, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Claire Valotteau
- Aix-Marseille University, INSERM, DyNaMo, Turing Centre for Living Systems, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Giuseppe Maria Paternò
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza Leonardo Da Vinci, 32, 20133 Milan, Italy
| | - Lorena Redondo Morata
- Aix-Marseille University, INSERM, DyNaMo, Turing Centre for Living Systems, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Francesca Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Sara Mattiello
- Department of Material Science, Bicocca University, Via Roberto Cozzi 55, 20126 Milano, Italy
| | - Arianna Magni
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza Leonardo Da Vinci, 32, 20133 Milan, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Luca Beverina
- Department of Material Science, Bicocca University, Via Roberto Cozzi 55, 20126 Milano, Italy
| | - Giuseppe Mattioli
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (CNR-ISM), Via Salaria km 29.300, 00015 Monterotondo (RM), Italy
| | - Guglielmo Lanzani
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza Leonardo Da Vinci, 32, 20133 Milan, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Elisabetta Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
15
|
Li X, Sedlacek M, Nath A, Szatko KP, Grimes WN, Diamond JS. A metabotropic glutamate receptor agonist enhances visual signal fidelity in a mouse model of retinitis pigmentosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591881. [PMID: 38746092 PMCID: PMC11092665 DOI: 10.1101/2024.04.30.591881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Many inherited retinal diseases target photoreceptors, which transduce light into a neural signal that is processed by the downstream visual system. As photoreceptors degenerate, physiological and morphological changes to retinal synapses and circuitry reduce sensitivity and increase noise, degrading visual signal fidelity. Here, we pharmacologically targeted the first synapse in the retina in an effort to reduce circuit noise without sacrificing visual sensitivity. We tested a strategy to partially replace the neurotransmitter lost when photoreceptors die with an agonist of receptors that ON bipolars cells use to detect glutamate released from photoreceptors. In rd10 mice, which express a photoreceptor mutation that causes retinitis pigmentosa (RP), we found that a low dose of the mGluR6 agonist l-2-amino-4-phosphonobutyric acid (L-AP4) reduced pathological noise induced by photoreceptor degeneration. After making in vivo electroretinogram recordings in rd10 mice to characterize the developmental time course of visual signal degeneration, we examined effects of L-AP4 on sensitivity and circuit noise by recording in vitro light-evoked responses from individual retinal ganglion cells (RGCs). L-AP4 decreased circuit noise evident in RGC recordings without significantly reducing response amplitudes, an effect that persisted over the entire time course of rod photoreceptor degeneration. Subsequent in vitro recordings from rod bipolar cells (RBCs) showed that RBCs are more depolarized in rd10 retinas, likely contributing to downstream circuit noise and reduced synaptic gain, both of which appear to be ameliorated by hyperpolarizing RBCs with L-AP4. These beneficial effects may reduce pathological circuit remodeling and preserve the efficacy of therapies designed to restore vision.
Collapse
Affiliation(s)
- Xiaoyi Li
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA 21218
| | - Miloslav Sedlacek
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - Amurta Nath
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - Klaudia P. Szatko
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - William N. Grimes
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - Jeffrey S. Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| |
Collapse
|
16
|
Wang Y, Liu X, Wang B, Sun H, Ren Y, Zhang H. Compounding engineered mesenchymal stem cell-derived exosomes: A potential rescue strategy for retinal degeneration. Biomed Pharmacother 2024; 173:116424. [PMID: 38471273 DOI: 10.1016/j.biopha.2024.116424] [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: 01/06/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
The prevalence of retinal degenerative diseases, including age-related macular degeneration and retinitis pigmentosa, has been increasing globally and is linked to the aging population and improved life expectancy. These diseases are characterized by chronic, progressive neuronal damage or depletion of the photoreceptor cells in the retina, and limited effective treatment options are currently available. Mesenchymal stem cell-derived exosomes (MSC-EXOs) containing cytokines, growth factors, lipids, mRNA, and miRNA, which act as mediators of intercellular communication transferring bioactive molecules to recipient cells, offer an appealing, non-cellular nanotherapeutic approach for retinal degenerative diseases. However, treatment specificity is compromised due to their high heterogeneity in size, content, functional effects, and parental cellular source. To improve this, engineered MSC-EXOs with increased drug-loading capacity, targeting ability, and resistance to bodily degradation and elimination have been developed. This review summarizes the recent advances in miRNAs of MSC-EXOs as a treatment for retinal degeneration, discussing the strategies and methods for engineering therapeutic MSC-EXOs. Notably, to address the single functional role of engineered MSC-EXOs, we propose a novel concept called "Compound Engineered MSC-EXOs (Co-E-MSC-EXOs)" along with its derived potential therapeutic approaches. The advantages and challenges of employing Co-E-MSC-EXOs for retinal degeneration in clinical applications, as well as the strategies and issues related to them, are also highlighted.
Collapse
Affiliation(s)
- Yao Wang
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China.
| | - Xianning Liu
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China
| | - Bei Wang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Hanhan Sun
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yiqian Ren
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China
| | - Hongbing Zhang
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China.
| |
Collapse
|
17
|
Grubaugh CR, Dhingra A, Prakash B, Montenegro D, Sparrow JR, Daniele LL, Curcio CA, Bell BA, Hussain MM, Boesze-Battaglia K. Microsomal triglyceride transfer protein is necessary to maintain lipid homeostasis and retinal function. FASEB J 2024; 38:e23522. [PMID: 38445789 PMCID: PMC10949407 DOI: 10.1096/fj.202302491r] [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: 12/02/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024]
Abstract
Lipid processing by the retinal pigment epithelium (RPE) is necessary to maintain retinal health and function. Dysregulation of retinal lipid homeostasis due to normal aging or age-related disease triggers lipid accumulation within the RPE, on Bruch's membrane (BrM), and in the subretinal space. In its role as a hub for lipid trafficking into and out of the neural retina, the RPE packages a significant amount of lipid into lipid droplets for storage and into apolipoprotein B (APOB)-containing lipoproteins (Blps) for export. Microsomal triglyceride transfer protein (MTP), encoded by the MTTP gene, is essential for Blp assembly. Herein we test the hypothesis that MTP expression in the RPE is essential to maintain lipid balance and retinal function using the newly generated RPEΔMttp mouse model. Using non-invasive ocular imaging, electroretinography, and histochemical and biochemical analyses we show that genetic depletion of Mttp from the RPE results in intracellular lipid accumulation, increased photoreceptor-associated cholesterol deposits, and photoreceptor cell death, and loss of rod but not cone function. RPE-specific reduction in Mttp had no significant effect on plasma lipids and lipoproteins. While APOB was decreased in the RPE, most ocular retinoids remained unchanged, with the exception of the storage form of retinoid, retinyl ester. Thus suggesting that RPE MTP is critical for Blp synthesis and assembly but is not directly involved in plasma lipoprotein metabolism. These studies demonstrate that RPE-specific MTP expression is necessary to establish and maintain retinal lipid homeostasis and visual function.
Collapse
Affiliation(s)
- Catharina R. Grubaugh
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Binu Prakash
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Diego Montenegro
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10027 USA
| | - Janet R. Sparrow
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10027 USA
| | - Lauren L. Daniele
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brent A. Bell
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Xu F, Zheng C, Xu W, Zhang S, Liu S, Chen X, Yao K. Breaking genetic shackles: The advance of base editing in genetic disorder treatment. Front Pharmacol 2024; 15:1364135. [PMID: 38510648 PMCID: PMC10953296 DOI: 10.3389/fphar.2024.1364135] [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: 01/01/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
The rapid evolution of gene editing technology has markedly improved the outlook for treating genetic diseases. Base editing, recognized as an exceptionally precise genetic modification tool, is emerging as a focus in the realm of genetic disease therapy. We provide a comprehensive overview of the fundamental principles and delivery methods of cytosine base editors (CBE), adenine base editors (ABE), and RNA base editors, with a particular focus on their applications and recent research advances in the treatment of genetic diseases. We have also explored the potential challenges faced by base editing technology in treatment, including aspects such as targeting specificity, safety, and efficacy, and have enumerated a series of possible solutions to propel the clinical translation of base editing technology. In conclusion, this article not only underscores the present state of base editing technology but also envisions its tremendous potential in the future, providing a novel perspective on the treatment of genetic diseases. It underscores the vast potential of base editing technology in the realm of genetic medicine, providing support for the progression of gene medicine and the development of innovative approaches to genetic disease therapy.
Collapse
Affiliation(s)
- Fang Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Caiyan Zheng
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Weihui Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shiyao Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shanshan Liu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaopeng Chen
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Seo HW, Cha S, Jeong Y, Ahn J, Lee KJ, Kim S, Goo YS. Focal stimulation of retinal ganglion cells using subretinal 3D microelectrodes with peripheral electrodes of opposite current. Biomed Eng Lett 2024; 14:355-365. [PMID: 38374901 PMCID: PMC10874361 DOI: 10.1007/s13534-023-00342-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/10/2023] [Accepted: 12/07/2023] [Indexed: 02/21/2024] Open
Abstract
Subretinal prostheses have been developed to stimulate survived retinal ganglion cells (RGCs), indirectly following the physiological visual pathways. However, current spreading from the prosthesis electrode causes the activation of unintended RGCs, thereby limiting the spatial resolution of artificial vision. This study proposes a strategy for focal stimulation of RGCs using a subretinal electrode array, in which six hexagonally arranged peripheral electrodes surround a stimulating electrode. RGCs in an in-vitro condition were subretinally stimulated using a fabricated electrode array coated with iridium oxide, following the three different stimulation configurations (with no peripheral, six electrodes of opposite current, and six ground). In-vitro experiments showed that the stimulation with six electrodes of opposite current was most effective in controlling RGC responses with a high spatial resolution. The results suggest that the effective utilization of return electrodes, such as by applying an opposite current to them, could help reduce current spreading beyond the local area targeted for stimulation and elicit RGC responses only in the vicinity of the stimulating electrode. Supplementary Information The online version contains supplementary material available at 10.1007/s13534-023-00342-3.
Collapse
Affiliation(s)
- Hee Won Seo
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Seongkwang Cha
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, Republic of Korea
| | - Yurim Jeong
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, Republic of Korea
| | - Jungryul Ahn
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, Republic of Korea
| | - Kyeong Jae Lee
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sohee Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yong Sook Goo
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, Republic of Korea
| |
Collapse
|
20
|
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
|
21
|
Ding J, Kim TH, Ma G, Yao X. Intrinsic signal optoretinography of dark adaptation abnormality due to rod photoreceptor degeneration. Exp Biol Med (Maywood) 2024; 249:10024. [PMID: 38463390 PMCID: PMC10911128 DOI: 10.3389/ebm.2024.10024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/04/2024] [Indexed: 03/12/2024] Open
Abstract
This research aims to investigate the potential of using intrinsic optical signal (IOS) optoretinography (ORG) to objectively detect dark adaptation (DA) abnormalities related to rod photoreceptor degeneration. Functional optical coherence tomography (OCT) was employed in both wild-type (WT) and retinal degeneration 10 (rd10) mice to conduct this assessment. Dynamic OCT measurements captured the changes in retinal thickness and reflectance from light-to-dark transition. Comparative analysis revealed significant IOS alterations within the outer retina. Specifically, a reduction in thickness from external limiting membrane (ELM) peak to retinal pigment epithelium (RPE) peak was observed (WT: 1.13 ± 0.69 µm, 30 min DA; rd10: 2.64 ± 0.86 µm, 30 min DA), as well as a decrease in the intensity of the inner segment ellipsoid zone (EZ) in 30 min DA compared to light adaptation (LA). The reduction of relative EZ intensity was notable in rd10 after 5 min DA and in WT after 15 min DA, with a distinguishable difference between rd10 and WT after 10 min DA. Furthermore, our findings indicated a significant decrease in the relative intensity of the hypo-reflective band between EZ and RPE in rd10 retinas during DA, which primarily corresponds to the outer segment (OS) region. In conclusion, the observed DA-IOS abnormalities, including changes in ELM-RPE thickness, EZ, and OS intensity, hold promise as differentiators between WT and rd10 mice before noticeable morphological abnormalities occur. These findings suggest the potential of this non-invasive imaging technique for the early detection of dysfunction in retinal photoreceptors.
Collapse
Affiliation(s)
- Jie Ding
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Tae-Hoon Kim
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Guangying Ma
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Xincheng Yao
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
22
|
Su E, Kesavamoorthy N, Junge JA, Zheng M, Craft CM, Ameri H. Comparison of Retinal Metabolic Activity and Structural Development between rd10 Mice and Normal Mice Using Multiphoton Fluorescence Lifetime Imaging Microscopy. Curr Issues Mol Biol 2024; 46:612-620. [PMID: 38248341 PMCID: PMC10813981 DOI: 10.3390/cimb46010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Fluorescence lifetime imaging microscopy (FLIM) is a technique that analyzes the metabolic state of tissues based on the spatial distribution of fluorescence lifetimes of certain interacting molecules. We used multiphoton FLIM to study the metabolic state of developing C57BL6/J and rd10 retinas based on the fluorescence lifetimes of free versus bound nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide phosphate (NAD(P)H), with free NAD(P)H percentages suggesting increased glycolysis and bound NAD(P)H percentages indicating oxidative phosphorylation. The mice were sacrificed and enucleated at various time points throughout their first 3 months of life. The isolated eyecups were fixed, sectioned using a polyacrylamide gel embedding technique, and then analyzed with FLIM. The results suggested that in both C57BL6/J mice and rd10 mice, oxidative phosphorylation initially decreased and then increased, plateauing over time. This trend, however, was accelerated in rd10 mice, with its turning point occurring at p10 versus the p30 turning point in C57BL6/J mice. There was also a noticeable difference in oxidative phosphorylation rates between the outer and inner retinas in both strains, with greater oxidative phosphorylation present in the latter. A greater understanding of rd10 and WT metabolic changes during retinal development may provide deeper insights into retinal degeneration and facilitate the development of future treatments.
Collapse
Affiliation(s)
- Erin Su
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (E.S.); (N.K.); (C.M.C.)
| | - Niranjana Kesavamoorthy
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (E.S.); (N.K.); (C.M.C.)
| | - Jason A. Junge
- Department of Biological Sciences, David Dornsife College of Letters Arts and Sciences, University of Southern California Dana, Los Angeles, CA 90089, USA;
| | - Mengmei Zheng
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (E.S.); (N.K.); (C.M.C.)
| | - Cheryl Mae Craft
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (E.S.); (N.K.); (C.M.C.)
| | - Hossein Ameri
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (E.S.); (N.K.); (C.M.C.)
| |
Collapse
|
23
|
Kim H, Roh H, Kim SH, Lee K, Im M, Oh SJ. Effective protection of photoreceptors using an inflammation-responsive hydrogel to attenuate outer retinal degeneration. NPJ Regen Med 2023; 8:68. [PMID: 38097595 PMCID: PMC10721838 DOI: 10.1038/s41536-023-00342-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Retinitis pigmentosa (RP) is an outer retinal degenerative disease that can lead to photoreceptor cell death and profound vision loss. Although effective regulation of intraretinal inflammation can slow down the progression of the disease, an efficient anti-inflammatory treatment strategy is still lacking. This study reports the fabrication of a hyaluronic acid-based inflammation-responsive hydrogel (IRH) and its epigenetic regulation effects on retinal degeneration. The injectable IRH was designed to respond to cathepsin overexpression in an inflammatory environment. The epigenetic drug, the enhancer of zeste homolog 2 (EZH2) inhibitors, was loaded into the hydrogel to attenuate inflammatory factors. On-demand anti-inflammatory effects of microglia cells via the drug-loaded IRH were verified in vitro and in vivo retinal degeneration 10 (rd10) mice model. Therefore, our IRH not only reduced intraretinal inflammation but also protected photoreceptors morphologically and functionally. Our results suggest the IRH reported here can be used to considerably delay vision loss caused by RP.
Collapse
Affiliation(s)
- Hyerim Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea
| | - Hyeonhee Roh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
- School of Electrical Engineering, College of Engineering, Korea University, Seoul, 02841, South Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute, KIST, Seoul, 02792, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, South Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea.
- Research Institute for Convergence Science, Seoul National University, Seoul, 08826, South Korea.
| | - Maesoon Im
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea.
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, South Korea.
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, South Korea.
| | - Seung Ja Oh
- Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, South Korea.
| |
Collapse
|
24
|
Grubaugh CR, Dhingra A, Prakash B, Montenegro D, Sparrow JR, Daniele LL, Curcio CA, Bell BA, Hussain MM, Boesze-Battaglia K. Microsomal triglyceride transfer protein is necessary to maintain lipid homeostasis and retinal function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570418. [PMID: 38105975 PMCID: PMC10723417 DOI: 10.1101/2023.12.06.570418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Lipid processing by the retinal pigment epithelium (RPE) is necessary to maintain retinal health and function. Dysregulation of retinal lipid homeostasis due to normal aging or to age-related disease triggers lipid accumulation within the RPE, on Bruch's membrane (BrM), and in the subretinal space. In its role as a hub for lipid trafficking into and out of the neural retina, the RPE packages a significant amount of lipid into lipid droplets for storage and into apolipoprotein B (apoB)-containing lipoproteins (Blps) for export. Microsomal triglyceride transfer protein (MTP), encoded by the MTTP gene, is essential for Blp assembly. Herein we test the hypothesis that MTP expression in the RPE is essential to maintain lipid balance and retinal function using the newly generated RPEΔMttp mouse model. Using non-invasive ocular imaging, electroretinography, and histochemical and biochemical analyses we show that genetic deletion of Mttp from the RPE results in intracellular lipid accumulation, increased photoreceptor -associated cholesterol deposits and photoreceptor cell death, and loss of rod but not cone function. RPE-specific ablation of Mttp had no significant effect on plasma lipids and lipoproteins. While, apoB was decreased in the RPE, ocular retinoid concentrations remained unchanged. Thus suggesting that RPE MTP is critical for Blp synthesis and assembly but not directly involved in ocular retinoid and plasma lipoprotein metabolism. These studies demonstrate that RPE-specific MTP expression is necessary to establish and maintain retinal lipid homeostasis and visual function.
Collapse
Affiliation(s)
- Catharina R. Grubaugh
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Binu Prakash
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Diego Montenegro
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY,10027 USA
| | - Janet R. Sparrow
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY,10027 USA
| | - Lauren L. Daniele
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brent A. Bell
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Sukkar B, Oktay L, Sahaboglu A, Moayedi A, Zenouri S, Al-Maghout T, Cantó A, Miranda M, Durdagi S, Hosseinzadeh Z. Inhibition of altered Orai1 channels in Müller cells protects photoreceptors in retinal degeneration. Glia 2023; 71:2511-2526. [PMID: 37533369 DOI: 10.1002/glia.24429] [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: 12/23/2022] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 08/04/2023]
Abstract
The expressions of ion channels by Müller glial cells (MGCs) may change in response to various retinal pathophysiological conditions. There remains a gap in our understanding of MGCs' responses to photoreceptor degeneration towards finding therapies. The study explores how an inhibition of store-operated Ca2+ entry (SOCE) and its major component, Orai1 channel, in MGCs protects photoreceptors from degeneration. The study revealed increased Orai1 expression in the MGCs of retinal degeneration 10 (rd10) mice. Enhanced expression of oxidative stress markers was confirmed as a crucial pathological mechanism in rd10 retina. Inducing oxidative stress in rat MGCs resulted in increasing SOCE and Ca2+ release-activated Ca2+ (CRAC) currents. SOCE inhibition by 2-Aminoethoxydiphenyl borate (2-APB) protected photoreceptors in degenerated retinas. Finally, molecular simulations proved the structural and dynamical features of 2-APB to the target structure Orai1. Our results provide new insights into the physiology of MGCs regarding retinal degeneration and shed a light on SOCE and Orai1 as new therapeutic targets.
Collapse
Affiliation(s)
- Basma Sukkar
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Lalehan Oktay
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Ayse Sahaboglu
- Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard Karls University, Tübingen, Germany
| | - Aylin Moayedi
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Shima Zenouri
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Tamer Al-Maghout
- Department of Cardiology and Vascular Medicine and Physiology, University of Tübingen, Tübingen, Germany
| | - Antolin Cantó
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - María Miranda
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
- Molecular Therapy Laboratory, School of Pharmacy, Bahcesehir University, Istanbul, Turkey
| | - Zohreh Hosseinzadeh
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
26
|
Matsevich C, Gopalakrishnan P, Chang N, Obolensky A, Beryozkin A, Salameh M, Kostic C, Sharon D, Arsenijevic Y, Banin E. Gene augmentation therapy attenuates retinal degeneration in a knockout mouse model of Fam161a retinitis pigmentosa. Mol Ther 2023; 31:2948-2961. [PMID: 37580905 PMCID: PMC10556223 DOI: 10.1016/j.ymthe.2023.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/14/2023] [Accepted: 08/10/2023] [Indexed: 08/16/2023] Open
Abstract
Photoreceptor cell degeneration and death is the major hallmark of a wide group of human blinding diseases including age-related macular degeneration and inherited retinal diseases such as retinitis pigmentosa. In recent years, inherited retinal diseases have become the "testing ground" for novel therapeutic modalities, including gene and cell-based therapies. Currently there is no available treatment for retinitis pigmentosa caused by FAM161A biallelic pathogenic variants. In this study, we injected an adeno-associated virus encoding for the longer transcript of mFam161a into the subretinal space of P24-P29 Fam161a knockout mice to characterize the safety and efficacy of gene augmentation therapy. Serial in vivo assessment of retinal function and structure at 3, 6, and 8 months of age using the optomotor response test, full-field electroretinography, fundus autofluorescence, and optical coherence tomography imaging as well as ex vivo quantitative histology and immunohistochemical studies revealed a significant structural and functional rescue effect in treated eyes accompanied by expression of the FAM161A protein in photoreceptors. The results of this study may serve as an important step toward future application of gene augmentation therapy in FAM161A-deficient patients by identifying a promising isoform to rescue photoreceptors and their function.
Collapse
Affiliation(s)
- Chen Matsevich
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Ning Chang
- Group for Retinal Disorder Research, Department of Ophthalmology, University Lausanne - Jules-Gonin Eye Hospital Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Alexey Obolensky
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Avigail Beryozkin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Manar Salameh
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Corinne Kostic
- Group for Retinal Disorder Research, Department of Ophthalmology, University Lausanne - Jules-Gonin Eye Hospital Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Dror Sharon
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | - Yvan Arsenijevic
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Eyal Banin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
27
|
Son Y, Chen ZC, Roh H, Lee BC, Im M. Effects on Retinal Stimulation of the Geometry and the Insertion Location of Penetrating Electrodes. IEEE Trans Neural Syst Rehabil Eng 2023; 31:3803-3812. [PMID: 37729573 DOI: 10.1109/tnsre.2023.3317496] [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: 09/22/2023]
Abstract
Retinal implants have been developed and implanted to restore vision from outer retinal degeneration, but their performance is still limited due to the poor spatial resolution. To improve the localization of stimulation, microelectrodes in various three-dimensional (3D) shapes have been investigated. In particular, computational simulation is crucial for optimizing the performance of a novel microelectrode design before actual fabrication. However, most previous studies have assumed a uniform conductivity for the entire retina without testing the effect of electrodes placement in different layers. In this study, we used the finite element method to simulate electric fields created by 3D microelectrodes of three different designs in a retina model with a stratified conductivity profile. The three electrode designs included two conventional shapes - a conical electrode (CE) and a pillar electrode (PE); we also proposed a novel structure of pillar electrode with an insulating wall (PEIW). A quantitative comparison of these designs shows the PEIW generates a stronger and more confined electric field with the same current injection, which is preferred for high-resolution retinal prostheses. Moreover, our results demonstrate both the magnitude and the shape of potential distribution generated by a penetrating electrode depend not only on the geometry, but also substantially on the insertion depth of the electrode. Although epiretinal insertions are mainly discussed, we also compared results for subretinal insertions. The results provide valuable insights for improving the spatial resolution of retinal implants using 3D penetrating microelectrodes and highlight the importance of considering the heterogeneity of conductivities in the retina.
Collapse
|
28
|
Cha S, Ahn J, Kim SW, Choi KE, Yoo Y, Eom H, Shin D, Goo YS. The Variation of Electrical Pulse Duration Elicits Reliable Network-Mediated Responses of Retinal Ganglion Cells in Normal, Not in Degenerate Primate Retinas. Bioengineering (Basel) 2023; 10:1135. [PMID: 37892865 PMCID: PMC10604198 DOI: 10.3390/bioengineering10101135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
This study aims to investigate the efficacy of electrical stimulation by comparing network-mediated RGC responses in normal and degenerate retinas using a N-methyl-N-nitrosourea (MNU)-induced non-human primate (NHPs) retinitis pigmentosa (RP) model. Adult cynomolgus monkeys were used for normal and outer retinal degeneration (RD) induced by MNU. The network-mediated RGC responses were recorded from the peripheral retina mounted on an 8 × 8 multielectrode array (MEA). The amplitude and duration of biphasic current pulses were modulated from 1 to 50 μA and 500 to 4000 μs, respectively. The threshold charge density for eliciting a network-mediated RGC response was higher in the RD monkeys than in the normal monkeys (1.47 ± 0.13 mC/cm2 vs. 1.06 ± 0.09 mC/cm2, p < 0.05) at a 500 μs pulse duration. The monkeys required a higher charge density than rodents among the RD models (monkeys; 1.47 ± 0.13 mC/cm2, mouse; 1.04 ± 0.09 mC/cm2, and rat; 1.16 ± 0.16 mC/cm2, p < 0.01). Increasing the pulse amplitude and pulse duration elicited more RGC spikes in the normal primate retinas. However, only pulse amplitude variation elicited more RGC spikes in degenerate primate retinas. Therefore, the pulse strategy for primate RD retinas should be optimized, eventually contributing to retinal prosthetics. Given that RD NHP RGCs are not sensitive to pulse duration, using shorter pulses may potentially be a more charge-effective approach for retinal prosthetics.
Collapse
Affiliation(s)
- Seongkwang Cha
- Department of Physiology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (S.C.); (J.A.)
| | - Jungryul Ahn
- Department of Physiology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (S.C.); (J.A.)
| | - Seong-Woo Kim
- Horang-I Eye Center, Seoul 07999, Republic of Korea;
| | - Kwang-Eon Choi
- Department of Ophthalmology, College of Medicine, Korea University, Seoul 08308, Republic of Korea;
| | - Yongseok Yoo
- School of Computer Science and Engineering, Soongsil University, Seoul 06978, Republic of Korea;
| | - Heejong Eom
- Laboratory Animal Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea; (H.E.); (D.S.)
| | - Donggwan Shin
- Laboratory Animal Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea; (H.E.); (D.S.)
| | - Yong Sook Goo
- Department of Physiology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (S.C.); (J.A.)
| |
Collapse
|
29
|
Halfmann C, Rüland T, Müller F, Jehasse K, Kampa BM. Electrophysiological properties of layer 2/3 pyramidal neurons in the primary visual cortex of a retinitis pigmentosa mouse model ( rd10). Front Cell Neurosci 2023; 17:1258773. [PMID: 37780205 PMCID: PMC10540630 DOI: 10.3389/fncel.2023.1258773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
Retinal degeneration is one of the main causes of visual impairment and blindness. One group of retinal degenerative diseases, leading to the loss of photoreceptors, is collectively termed retinitis pigmentosa. In this group of diseases, the remaining retina is largely spared from initial cell death making retinal ganglion cells an interesting target for vision restoration methods. However, it is unknown how downstream brain areas, in particular the visual cortex, are affected by the progression of blindness. Visual deprivation studies have shown dramatic changes in the electrophysiological properties of visual cortex neurons, but changes on a cellular level in retinitis pigmentosa have not been investigated yet. Therefore, we used the rd10 mouse model to perform patch-clamp recordings of pyramidal neurons in layer 2/3 of the primary visual cortex to screen for potential changes in electrophysiological properties resulting from retinal degeneration. Compared to wild-type C57BL/6 mice, we only found an increase in intrinsic excitability around the time point of maximal retinal degeneration. In addition, we saw an increase in the current amplitude of spontaneous putative inhibitory events after a longer progression of retinal degeneration. However, we did not observe a long-lasting shift in excitability after prolonged retinal degeneration. Together, our results provide evidence of an intact visual cortex with promising potential for future therapeutic strategies to restore vision.
Collapse
Affiliation(s)
- Claas Halfmann
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Thomas Rüland
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
- Molecular and Cellular Physiology, Institute of Biological Information Processing (IBI-1), Forschungszentrum Jülich GmbH, Jülich, Germany
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany
| | - Frank Müller
- Molecular and Cellular Physiology, Institute of Biological Information Processing (IBI-1), Forschungszentrum Jülich GmbH, Jülich, Germany
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany
- Research Training Group 2610 Innoretvision, RWTH Aachen University, Aachen, Germany
| | - Kevin Jehasse
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Björn M. Kampa
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany
- Research Training Group 2610 Innoretvision, RWTH Aachen University, Aachen, Germany
- JARA BRAIN, Institute of Neuroscience and Medicine (INM-10), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
30
|
Kramer RH. Suppressing Retinal Remodeling to Mitigate Vision Loss in Photoreceptor Degenerative Disorders. Annu Rev Vis Sci 2023; 9:131-153. [PMID: 37713276 DOI: 10.1146/annurev-vision-112122-020957] [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] [Indexed: 09/17/2023]
Abstract
Rod and cone photoreceptors degenerate in retinitis pigmentosa and age-related macular degeneration, robbing the visual system of light-triggered signals necessary for sight. However, changes in the retina do not stop with the photoreceptors. A stereotypical set of morphological and physiological changes, known as remodeling, occur in downstream retinal neurons. Some aspects of remodeling are homeostatic, with structural or functional changes compensating for partial loss of visual inputs. However, other aspects are nonhomeostatic, corrupting retinal information processing to obscure vision mediated naturally by surviving photoreceptors or artificially by vision-restoration technologies. In this review, I consider the mechanism of remodeling and its consequences for residual and restored visual function; discuss the role of retinoic acid, a critical molecular trigger of detrimental remodeling; and discuss strategies for suppressing retinoic acid biosynthesis or signaling as therapeutic possibilities for mitigating vision loss.
Collapse
Affiliation(s)
- Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, USA;
| |
Collapse
|
31
|
Wu Y, Wan X, Zhao D, Chen X, Wang Y, Tang X, Li J, Li S, Sun X, Bi C, Zhang X. AAV-mediated base-editing therapy ameliorates the disease phenotypes in a mouse model of retinitis pigmentosa. Nat Commun 2023; 14:4923. [PMID: 37582961 PMCID: PMC10427680 DOI: 10.1038/s41467-023-40655-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 08/07/2023] [Indexed: 08/17/2023] Open
Abstract
Base editing technology is an ideal solution for treating pathogenic single-nucleotide variations (SNVs). No gene editing therapy has yet been approved for eye diseases, such as retinitis pigmentosa (RP). Here, we show, in the rd10 mouse model, which carries an SNV identified as an RP-causing mutation in human patients, that subretinal delivery of an optimized dual adeno-associated virus system containing the adenine base editor corrects the pathogenic SNV in the neuroretina with up to 49% efficiency. Light microscopy showed that a thick and robust outer nuclear layer (photoreceptors) was preserved in the treated area compared with the thin, degenerated outer nuclear layer without treatment. Substantial electroretinogram signals were detected in treated rd10 eyes, whereas control treated eyes showed minimal signals. The water maze experiment showed that the treatment substantially improved vision-guided behavior. Together, we construct and validate a translational therapeutic solution for the treatment of RP in humans. Our findings might accelerate the development of base-editing based gene therapies.
Collapse
Affiliation(s)
- Yidong Wu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Eye Diseases, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
| | - Dongdong Zhao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Xuxu Chen
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Yujie Wang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Xinxin Tang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Ju Li
- College of Life Science, Tianjin Normal University, Tianjin, China
| | - Siwei Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Eye Diseases, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
| | - Changhao Bi
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Technology Innovation Center of Synthetic Biology, Tianjin, China.
| | - Xueli Zhang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Technology Innovation Center of Synthetic Biology, Tianjin, China.
| |
Collapse
|
32
|
Haq W, Zrenner E, Ueffing M, Paquet-Durand F. Using Micro-Electrode-Array Recordings and Retinal Disease Models to Elucidate Visual Functions: Simultaneous Recording of Local Electroretinograms and Ganglion Cell Action Potentials Reveals the Origin of Retinal Oscillatory Potentials. Bioengineering (Basel) 2023; 10:725. [PMID: 37370656 DOI: 10.3390/bioengineering10060725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The electroretinogram (ERG) is an essential diagnostic tool for visual function, both in clinical and research settings. Here, we establish an advanced in vitro approach to assess cell-type-specific ERG signal components. METHODS Retinal explant cultures, maintained under entirely controlled conditions, were derived from wild-type mice and rd10 rod- and cpfl1 cone-degeneration mouse models. Local micro-ERG (µERG) and simultaneous ganglion cell (GC) recordings were obtained from the retinal explants using multi-electrode arrays. Band-pass filtering was employed to distinguish photoreceptor, bipolar cell, amacrine cell (AC), and GC responses. RESULTS Scotopic and photopic stimulation discriminated between rod and cone responses in wild-type and mutant retina. The 25 kHz sampling rate allowed the visualization of oscillatory potentials (OPs) in extraordinary detail, revealing temporal correlations between OPs and GC responses. Pharmacological isolation of different retinal circuits found that OPs are generated by inner retinal AC electrical synapses. Importantly, this AC activity helped synchronise GC activity. CONCLUSION Our µERG protocol simultaneously records the light-dependent activities of the first-, second-, and third-order neurons within the native neuronal circuitry, providing unprecedented insights into retinal physiology and pathophysiology. This method now also enables complete in vitro retinal function testing of therapeutic interventions, providing critical guidance for later in vivo investigations.
Collapse
Affiliation(s)
- Wadood Haq
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| | - Eberhart Zrenner
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| | - François Paquet-Durand
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| |
Collapse
|
33
|
Ahn J, Jeong Y, Cha S, Lee JY, Yoo Y, Goo YS. High amplitude pulses on the same charge condition efficiently elicit bipolar cell-mediated retinal ganglion cell responses in the degenerate retina. Biomed Eng Lett 2023; 13:129-140. [PMID: 37124107 PMCID: PMC10130300 DOI: 10.1007/s13534-023-00260-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 02/01/2023] Open
Abstract
Retinal pigmentosa (RP) patients lose vision due to the loss of photoreceptors. Retinal prostheses bypass the dead photoreceptors by electrically stimulating surviving retinal neurons, such as bipolar cells or retinal ganglion cells (RGCs). In previous studies, stimulus charge has been mainly optimized to maximize the RGC response to electrical stimulation. This study aimed to investigate the effect of amplitude and duration even under the same charge condition on eliciting RGC spikes in the wild-type and degenerate retinas. Wild-type (WT) Sprague-Dawley rats were used as the normal retinal model, and Pde6b knockout rats were used as a retinal degeneration (RD) model. Electrically-evoked RGC spikes were recorded from isolated rat retinas using an 8 × 8 multielectrode array. The same charge was maintained (10 or 20 nC), and electrical stimulation was applied to WT and RD retinas, adjusting the amplitude and duration of the 1st phase of biphasic pulses. In the pulse modulation of the 1st phase, high amplitude (short duration) pulses induced more RGC spikes than low amplitude (long duration) pulses. Both WT and RD retinas showed a significant reduction in the number of RGC spikes upon stimulation with lower amplitude (longer duration) pulses. In clinical trials where stimulus charges are delivered to the degenerate retina of blind patients, high amplitude (short duration) pulses would help elicit more RGC spikes.
Collapse
Affiliation(s)
- Jungryul Ahn
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Yurim Jeong
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Seongkwang Cha
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Joo Yong Lee
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yongseok Yoo
- School of Computer Science and Engineering, Soongsil University, Seoul, South Korea
| | - Yong Sook Goo
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| |
Collapse
|
34
|
Barwick SR, Xiao H, Wolff D, Wang J, Perry E, Marshall B, Smith SB. Sigma 1 receptor activation improves retinal structure and function in the Rho P23H/+ mouse model of autosomal dominant retinitis pigmentosa. Exp Eye Res 2023; 230:109462. [PMID: 37003581 PMCID: PMC10155485 DOI: 10.1016/j.exer.2023.109462] [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: 12/12/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Retinitis pigmentosa (RP) is a group of devastating inherited retinal diseases that leads to visual impairment and oftentimes complete blindness. Currently no cure exists for RP thus research into prolonging vision is imperative. Sigma 1 receptor (Sig1R) is a promising small molecule target that has neuroprotective benefits in retinas of rapidly-degenerating mouse models. It is not clear whether Sig1R activation can provide similar neuroprotective benefits in more slowly-progressing RP models. Here, we examined Sig1R-mediated effects in the slowly-progressing RhoP23H/+ mouse, a model of autosomal dominant RP. We characterized the retinal degeneration of the RhoP23H/+ mouse over a 10 month period using three in vivo methods: Optomotor Response (OMR), Electroretinogram (ERG), and Spectral Domain-Optical Coherence Tomography (SD-OCT). A slow retinal degeneration was observed in both male and female RhoP23H/+ mice when compared to wild type. The OMR, which reflects visual acuity, showed a gradual decline through 10 months. Interestingly, female mice had more reduction in visual acuity than males. ERG assessment showed a gradual decline in scotopic and photopic responses in RhoP23H/+ mice. To investigate the neuroprotective benefits of Sig1R activation in the RhoP23H/+ mouse model, mutant mice were treated with a high-specificity Sig1R ligand (+)-pentazocine ((+)-PTZ) 3x/week at 0.5 mg/kg and examined using OMR, ERG, SD-OCT. A significant retention of visual function was observed in males and females at 10 months of age, with treated females retaining ∼50% greater visual acuity than non-treated mutant females. ERG revealed significant retention of scotopic and photopic b-wave amplitudes at 6 months in male and female RhoP23H/+ mice treated with (+)-PTZ. Further, in vivo analysis by SD-OCT revealed a significant retention of outer nuclear layer (ONL) thickness in male and female treated RhoP23H/+ mice. Histological studies showed significant retention of IS/OS length (∼50%), ONL thickness, and number of rows of photoreceptor cell nuclei at 6 months in (+)-PTZ-treated mutant mice. Interestingly, electron microscopy revealed preservation of OS discs in (+)-PTZ treated mutant mice compared to non-treated. Taken collectively, the in vivo and in vitro data provide the first evidence that targeting Sig1R can rescue visual function and structure in the RhoP23H/+ mouse. These results are promising and provide a framework for future studies to investigate Sig1R as a potential therapeutic target in retinal degenerative disease.
Collapse
Affiliation(s)
- Shannon R Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - David Wolff
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Elizabeth Perry
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Brendan Marshall
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
35
|
Ellis EM, Paniagua AE, Scalabrino ML, Thapa M, Rathinavelu J, Jiao Y, Williams DS, Field GD, Fain GL, Sampath AP. Cones and cone pathways remain functional in advanced retinal degeneration. Curr Biol 2023; 33:1513-1522.e4. [PMID: 36977418 PMCID: PMC10133175 DOI: 10.1016/j.cub.2023.03.007] [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: 09/20/2022] [Revised: 12/14/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
Abstract
Most defects causing retinal degeneration in retinitis pigmentosa (RP) are rod-specific mutations, but the subsequent degeneration of cones, which produces loss of daylight vision and high-acuity perception, is the most debilitating feature of the disease. To understand better why cones degenerate and how cone vision might be restored, we have made the first single-cell recordings of light responses from degenerating cones and retinal interneurons after most rods have died and cones have lost their outer-segment disk membranes and synaptic pedicles. We show that degenerating cones have functional cyclic-nucleotide-gated channels and can continue to give light responses, apparently produced by opsin localized either to small areas of organized membrane near the ciliary axoneme or distributed throughout the inner segment. Light responses of second-order horizontal and bipolar cells are less sensitive but otherwise resemble those of normal retina. Furthermore, retinal output as reflected in responses of ganglion cells is less sensitive but maintains spatiotemporal receptive fields at cone-mediated light levels. Together, these findings show that cones and their retinal pathways can remain functional even as degeneration is progressing, an encouraging result for future research aimed at enhancing the light sensitivity of residual cones to restore vision in patients with genetically inherited retinal degeneration.
Collapse
Affiliation(s)
- Erika M Ellis
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA
| | - Antonio E Paniagua
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA
| | - Miranda L Scalabrino
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mishek Thapa
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jay Rathinavelu
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yuekan Jiao
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA
| | - David S Williams
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA.
| | - Greg D Field
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Gordon L Fain
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA.
| | - Alapakkam P Sampath
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA.
| |
Collapse
|
36
|
Yamoah A, Tripathi P, Guo H, Scheve L, Walter P, Johnen S, Müller F, Weis J, Goswami A. Early Alterations of RNA Binding Protein (RBP) Homeostasis and ER Stress-Mediated Autophagy Contributes to Progressive Retinal Degeneration in the rd10 Mouse Model of Retinitis Pigmentosa (RP). Cells 2023; 12:cells12071094. [PMID: 37048167 PMCID: PMC10092976 DOI: 10.3390/cells12071094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
The retinal degeneration 10 (rd10) mouse model is widely used to study retinitis pigmentosa (RP) pathomechanisms. It offers a rather unique opportunity to study trans-neuronal degeneration because the cell populations in question are separated anatomically and the mutated Pde6b gene is selectively expressed in rod photoreceptors. We hypothesized that RNA binding protein (RBP) aggregation and abnormal autophagy might serve as early pathogenic events, damaging non-photoreceptor retinal cell types that are not primarily targeted by the Pde6b gene defect. We used a combination of immunohistochemistry (DAB, immunofluorescence), electron microscopy (EM), subcellular fractionation, and Western blot analysis on the retinal preparations obtained from both rd10 and wild-type mice. We found early, robust increases in levels of the protective endoplasmic reticulum (ER) calcium (Ca2+) buffering chaperone Sigma receptor 1 (SigR1) together with other ER-Ca2+ buffering proteins in both photoreceptors and non-photoreceptor neuronal cells before any noticeable photoreceptor degeneration. In line with this, we found markedly altered expression of the autophagy proteins p62 and LC3, together with abnormal ER widening and large autophagic vacuoles as detected by EM. Interestingly, these changes were accompanied by early, prominent cytoplasmic and nuclear aggregation of the key RBPs including pTDP-43 and FET family RBPs and stress granule formation. We conclude that progressive neurodegeneration in the rd10 mouse retina is associated with early disturbances of proteostasis and autophagy, along with abnormal cytoplasmic RBP aggregation.
Collapse
Affiliation(s)
- Alfred Yamoah
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- EURON-European Graduate School of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Priyanka Tripathi
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- EURON-European Graduate School of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Haihong Guo
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Leonie Scheve
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Sandra Johnen
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Frank Müller
- Institute of Biological Information Processing, Molecular and Cellular Physiology, IBI-1, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Anand Goswami
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
37
|
Maidana DE, Gonzalez-Buendia L, Miller JW, Vavvas DG. RIPK necrotic cell death pathway in both donor photoreceptor and host immune cells synergize to affect photoreceptor graft survival. FASEB J 2023; 37:e22847. [PMID: 36862516 PMCID: PMC10590064 DOI: 10.1096/fj.202201137r] [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: 07/16/2022] [Revised: 11/29/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Photoreceptor transplant has been put forward as a repair strategy to tackle degenerated retinas. Nonetheless, cell death and immune rejection seriously limit the success of this strategy, with only a small fraction of transplanted cells surviving. Improving the survival of transplanted cells is of critical importance. Recent evidence has identified receptor-interacting protein kinase 3 (RIPK3) as a molecular trigger controlling necroptotic cell death and inflammation. However, its role in photoreceptor transplantation and regenerative medicine has not been studied. We hypothesized that modulation of RIPK3 to address both cell death and immunity could have advantageous effects on photoreceptor survival. In a model of inherited retinal degeneration, deletion of RIPK3 in donor photoreceptor precursors significantly increases the survival of transplanted cells. Simultaneous RIPK3 deletion in donor photoreceptors and recipients maximizes graft survival. Lastly, to discern the role of RIPK3 in the host immune response, bone marrow transplant experiments demonstrated that peripheral immune cell RIPK3 deficiency is protective for both donor and host photoreceptor survival. Interestingly, this finding is independent of photoreceptor transplantation, as the peripheral protective effect is also observed in an additional retinal detachment photoreceptor degeneration model. Altogether, these results indicate that immunomodulatory and neuroprotective strategies targeting the RIPK3 pathway can aid regenerative therapies of photoreceptor transplantation.
Collapse
Affiliation(s)
- Daniel E. Maidana
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lucia Gonzalez-Buendia
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Joan W. Miller
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
38
|
Rasmussen M, Tolone A, Paquet-Durand F, Welinder C, Schwede F, Ekström P. The photoreceptor protective cGMP-analog Rp-8-Br-PET-cGMPS interacts with cGMP-interactors PKGI, PDE1, PDE6, and PKAI in the degenerating mouse retina. J Comp Neurol 2023; 531:935-951. [PMID: 36989379 DOI: 10.1002/cne.25475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/12/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023]
Abstract
The inherited eye disease retinitis pigmentosa (RP) causes the loss of photoreceptors by a still unknown cell death mechanism. During this degeneration, cyclic guanosine-3',5'-monophosphate (cGMP) levels become elevated, leading to over-activation of the cGMP-binding protein cGMP-dependent protein kinase (PKG). cGMP analogs selectively modified to have inhibitory actions on PKG have aided in impeding photoreceptor death, and one such cGMP analog is Rp-8-Br-PET-cGMPS. However, cGMP analogs have previously been shown to interact with numerous targets, so to better understand the therapeutic action of Rp-8-Br-PET-cGMPS, it is necessary to elucidate its target-selectivity and hence what potential cellular mechanism(s) it may affect within the photoreceptors. Here, we, therefore, applied affinity chromatography together with mass spectrometry to isolate and identify Rp-8-Br-PET-cGMPS interactors from retinas derived from three different murine RP models (i.e., rd1, rd2, and rd10 mice). Our findings revealed that Rp-8-Br-PET-cGMPS bound seven known cGMP-binding proteins, including PKG1β, PDE1β, PDE1c, PDE6α, and PKA1α. Furthermore, an additional 28 proteins were found to be associated with Rp-8-Br-PET-cGMPS. This latter group included MAPK1/3, which is known to connect with cGMP/PKG in other systems. However, in organotypic retinal cultures, Rp-8-Br-PET-cGMPS had no effect on photoreceptor MAPK1/3 expression or activity. To summarize, Rp-8-Br-PET-cGMPS is more target specific compared to regular cGMP.
Collapse
Affiliation(s)
- Michel Rasmussen
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| | - Arianna Tolone
- Insitute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | | | - Charlotte Welinder
- Faculty of Medicine, Department of Clinical Sciences Lund, Mass Spectrometry, Lund University, Lund, Sweden
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co. KG, Bremen, Germany
| | - Per Ekström
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| |
Collapse
|
39
|
Cammalleri M, Amato R, Dal Monte M, Filippi L, Bagnoli P. The β3 adrenoceptor in proliferative retinopathies: "Cinderella" steps out of its family shadow. Pharmacol Res 2023; 190:106713. [PMID: 36863427 DOI: 10.1016/j.phrs.2023.106713] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/14/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
In the retina, hypoxic condition leads to overgrowing leaky vessels resulting in altered metabolic supply that may cause impaired visual function. Hypoxia-inducible factor-1 (HIF-1) is a central regulator of the retinal response to hypoxia by activating the transcription of numerous target genes, including vascular endothelium growth factor, which acts as a major player in retinal angiogenesis. In the present review, oxygen urge by the retina and its oxygen sensing systems including HIF-1 are discussed in respect to the role of the beta-adrenergic receptors (β-ARs) and their pharmacologic manipulation in the vascular response to hypoxia. In the β-AR family, β1- and β2-AR have long been attracting attention because their pharmacology is intensely used for human health, while β3-AR, the third and last cloned receptor is no longer increasingly emerging as an attractive target for drug discovery. Here, β3-AR, a main character in several organs including the heart, the adipose tissue and the urinary bladder, but so far a supporting actor in the retina, has been thoroughly examined in respect to its function in retinal response to hypoxia. In particular, its oxygen dependence has been taken as a key indicator of β3-AR involvement in HIF-1-mediated responses to oxygen. Hence, the possibility of β3-AR transcription by HIF-1 has been discussed from early circumstantial evidence to the recent demonstration that β3-AR acts as a novel HIF-1 target gene by playing like a putative intermediary between oxygen levels and retinal vessel proliferation. Thus, targeting β3-AR may implement the therapeutic armamentarium against neovascular pathologies of the eye.
Collapse
Affiliation(s)
| | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Luca Filippi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy.
| |
Collapse
|
40
|
Roh H, Otgondemberel Y, Eom J, Kim D, Im M. Electrically-evoked responses for retinal prostheses are differentially altered depending on ganglion cell types in outer retinal neurodegeneration caused by Crb1 gene mutation. Front Cell Neurosci 2023; 17:1115703. [PMID: 36814867 PMCID: PMC9939843 DOI: 10.3389/fncel.2023.1115703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/19/2023] [Indexed: 02/08/2023] Open
Abstract
Background Microelectronic prostheses for artificial vision stimulate neurons surviving outer retinal neurodegeneration such as retinitis pigmentosa (RP). Yet, the quality of prosthetic vision substantially varies across subjects, maybe due to different levels of retinal degeneration and/or distinct genotypes. Although the RP genotypes are remarkably diverse, prosthetic studies have primarily used retinal degeneration (rd) 1 and 10 mice, which both have Pde6b gene mutation. Here, we report the electric responses arising in retinal ganglion cells (RGCs) of the rd8 mouse model which has Crb1 mutation. Methods We first investigated age-dependent histological changes of wild-type (wt), rd8, and rd10 mice retinas by H&E staining. Then, we used cell-attached patch clamping to record spiking responses of ON, OFF and direction selective (DS) types of RGCs to a 4-ms-long electric pulse. The electric responses of rd8 RGCs were analyzed in comparison with those of wt RGCs in terms of individual RGC spiking patterns, populational characteristics, and spiking consistency across trials. Results In the histological examination, the rd8 mice showed partial retinal foldings, but the outer nuclear layer thicknesses remained comparable to those of the wt mice, indicating the early-stage of RP. Although spiking patterns of each RGC type seemed similar to those of the wt retinas, correlation levels between electric vs. light response features were different across the two mouse models. For example, in comparisons between light vs. electric response magnitudes, ON/OFF RGCs of the rd8 mice showed the same/opposite correlation polarity with those of wt mice, respectively. Also, the electric response spike counts of DS RGCs in the rd8 retinas showed a positive correlation with their direction selectivity indices (r = 0.40), while those of the wt retinas were negatively correlated (r = -0.90). Lastly, the spiking timing consistencies of late responses were largely decreased in both ON and OFF RGCs in the rd8 than the wt retinas, whereas no significant difference was found across DS RGCs of the two models. Conclusion Our results indicate the electric response features are altered depending on RGC types even from the early-stage RP caused by Crb1 mutation. Given the various degeneration patterns depending on mutation genes, our study suggests the importance of both genotype- and RGC type-dependent analyses for retinal prosthetic research.
Collapse
Affiliation(s)
- Hyeonhee Roh
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- School of Electrical Engineering, Korea University, Seoul, Republic of Korea
| | | | - Jeonghyeon Eom
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- School of Electrical Engineering, Kookmin University, Seoul, Republic of Korea
| | - Daniel Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Maesoon Im
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
41
|
Mercau ME, Akalu YT, Mazzoni F, Gyimesi G, Alberto EJ, Kong Y, Hafler BP, Finnemann SC, Rothlin CV, Ghosh S. Inflammation of the retinal pigment epithelium drives early-onset photoreceptor degeneration in Mertk-associated retinitis pigmentosa. SCIENCE ADVANCES 2023; 9:eade9459. [PMID: 36662852 PMCID: PMC9858494 DOI: 10.1126/sciadv.ade9459] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/19/2022] [Indexed: 05/17/2023]
Abstract
Severe, early-onset photoreceptor (PR) degeneration associated with MERTK mutations is thought to result from failed phagocytosis by retinal pigment epithelium (RPE). Notwithstanding, the severity and onset of PR degeneration in mouse models of Mertk ablation are determined by the hypomorphic expression or the loss of the Mertk paralog Tyro3. Here, we find that loss of Mertk and reduced expression/loss of Tyro3 led to RPE inflammation even before eye-opening. Incipient RPE inflammation cascaded to involve microglia activation and PR degeneration with monocyte infiltration. Inhibition of RPE inflammation with the JAK1/2 inhibitor ruxolitinib mitigated PR degeneration in Mertk-/- mice. Neither inflammation nor severe, early-onset PR degeneration was observed in mice with defective phagocytosis alone. Thus, inflammation drives severe, early-onset PR degeneration-associated with Mertk loss of function.
Collapse
Affiliation(s)
- Maria E. Mercau
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yemsratch T. Akalu
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Francesca Mazzoni
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Gavin Gyimesi
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Emily J. Alberto
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yong Kong
- Department of Molecular Biophysics and Biochemistry, W. M. Keck Foundation Biotechnology Resource Laboratory, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Brian P. Hafler
- Department of Ophthalmology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Carla V. Rothlin
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Sourav Ghosh
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
42
|
Paguaga ME, Penn JS, Uddin MDI. A novel optical imaging probe for targeted visualization of NLRP3 inflammasomes in a mouse model of age-related macular degeneration. Front Med (Lausanne) 2023; 9:1047791. [PMID: 36703888 PMCID: PMC9871584 DOI: 10.3389/fmed.2022.1047791] [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: 09/18/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Purpose Wet form of age-related macular degeneration (wet AMD) is a progressive vascular disease that mainly affects older adults and causes severe and irreversible vision loss. A key complication of wet AMD is choroidal neovascularization (CNV), which may be driven in part by NLRP3 inflammasomes that are associated with macrophages migration to CNV lesions. Since activated NLRP3 is correlated with CNV, visualizing NLRP3 inflammasomes and their associated macrophages is of great interest to monitor wet AMD progression and develop effective therapies against it. However, to the best of our knowledge, current ophthalmic imaging systems do not permit such targeted imaging. Therefore, in this study, we developed InflammaProbe-1, an optical imaging probe for targeted visualization of NLRP3 inflammasomes in CNV lesions. Methods InflammaProbe-1 was synthesized by conjugating a clinically relevant fluorophore, Oregon Green® 488, to the selective NLRP3 inhibitor, CY-09. The ability of InflammaProbe-1 to target NLRP3 was assessed with an enzyme-linked immunosorbent assay by comparing its ability to inhibit NLRP3-mediated secretion of IL-1β to that of CY-09 in LPS-primed and nigericin-stimulated BMDMs. In vitro confocal imaging of NLRP3 was performed on InflammaProbe-1-stained BMDMs that had been induced to express NLRP3 with LPS. In vivo imaging of NLRP3 was conducted on mouse laser induced choroidal neovascularization (LCNV), a model of AMD, 6 h after an intraperitoneal injection of InflammaProbe-1 at 10 mg/kg on day 4 post-LCNV. Results InflammaProbe-1 was just as effective as CY-09 at inhibiting IL-1β secretion (p < 0.01 at 10 μM for both the InflammaProbe-1 and CY-09 groups relative to the control). InflammaProbe-1-stained BMDMs that had been induced to express NLRP3 showed significantly brighter fluorescence than untreated cells (p < 0.0001 for LPS treatment group and p < 0.001 for LPS and nigericin treatment group). Furthermore, in vivo molecular imaging of NLRP3 was achieved in mouse LCNV. Conclusion We propose that InflammaProbe-1 may be a useful molecular imaging probe to monitor the onset, progression, and therapeutic response of AMD and other NLRP3-mediated diseases.
Collapse
Affiliation(s)
- Marcell E. Paguaga
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - John S. Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - MD Imam Uddin
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States,*Correspondence: MD Imam Uddin,
| |
Collapse
|
43
|
Cheng X, Gao H, Tao Z, Yin Z, Cha Z, Huang X, Zhang Y, Zeng Y, He J, Ge L, A L, Xu H, Peng GH. Repopulated retinal microglia promote Müller glia reprogramming and preserve visual function in retinal degenerative mice. Theranostics 2023; 13:1698-1715. [PMID: 37056562 PMCID: PMC10086209 DOI: 10.7150/thno.79538] [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: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 04/15/2023] Open
Abstract
Rationale: Müller glia (MG) play a key role in maintaining homeostasis of the retinal microenvironment. In zebrafish, MG reprogram into retinal progenitors and repair the injured retina, while this MG regenerative capability is suppressed in mammals. It has been revealed that microglia in zebrafish contribute to MG reprogramming, whereas those in mammals are over-activated during retinal injury or degeneration, causing chronic inflammation, acceleration of photoreceptor apoptosis, and gliosis of MG. Therefore, how to modulate the phenotype of microglia to enhance MG reprogramming rather than gliosis is critical. Methods: PLX3397, a colony-stimulating factor 1 receptor inhibitor, was applied to deplete microglia in the retinas of retinal degeneration 10 (rd10) mice, and withdrawal of PLX3397 was used to induce the repopulated microglia (Rep-MiG). The protective roles of the Rep-MiG on the degenerative retina were assessed using a light/dark transition test, and scotopic electroretinogram recordings. Immunofluorescence, western blot, transcriptomic sequencing, and bioinformatics analysis were performed to investigate the effects and mechanisms of microglia on MG reprogramming. Results: Following PLX3397 withdrawal, Rep-MiG replenished the entire retina with a ramified morphology and significantly improved the retinal outer nuclear layer structure, the electroretinography response, and the visual behavior of rd10 mice. Coincidentally, MG were activated, de-differentiated, and showed properties of retina progenitors in a spatial correlation with Rep-MiG. Morphological and transcriptomic analyses revealed Rep-MiG significantly enhanced protease inhibitor activity and suppressed extracellular matrix (ECM) levels during retinal degeneration. Conclusions: It suggested that Rep-MiG with the homeostasis characteristic stimulated the progenitor cell-like properties of MG, probably through regulating ECM remodeling, which protected photoreceptors and improved visual function of rd10 mice. It might be a potential protocol to reprogram MG and delay mammal retinal degeneration.
Collapse
Affiliation(s)
- Xuan Cheng
- Department of Ophthalmology, First medical center of Chinese PLA General Hospital, Beijing, 100853, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Hui Gao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Zui Tao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Zhiyuan Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Zhe Cha
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Xiaona Huang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Yikui Zhang
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Juncai He
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Lingling Ge
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Luodan A
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
- ✉ Corresponding authors: Haiwei Xu () and Guang-Hua Peng ()
| | - Guang-Hua Peng
- Department of Ophthalmology, First medical center of Chinese PLA General Hospital, Beijing, 100853, China
- Lab of Visual Cell Differentiation and Regulation, Basic Medical College, Zhengzhou University, Zhengzhou, China
- ✉ Corresponding authors: Haiwei Xu () and Guang-Hua Peng ()
| |
Collapse
|
44
|
Sigurdsson D, Grimm C. Single-Cell Transcriptomic Profiling of Müller Glia in the rd10 Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:377-381. [PMID: 37440060 DOI: 10.1007/978-3-031-27681-1_55] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Müller glia are the principal macroglia of the retina and support retinal neurons both in health and disease. In retinitis pigmentosa (RP), a highly heterogeneous inherited retinal disorder, the most common form of pathology involves primary rod degeneration, followed by secondary cone death. To investigate Müller glia responses to rod degeneration, we performed droplet-based single-cell RNA sequencing in the rd10 mouse model of RP during primary rod degeneration. We confirmed known MG behavior on gliosis, metabolic, and immune functions. Pde6brd10 Müller glia also exhibited an increased expression of histocompatibility complex members, which might arise from a novel immune function of Müller glia in RP. We also describe a possible decrease in glial lipid biogenesis, which might affect degenerating photoreceptors.
Collapse
Affiliation(s)
- Duygu Sigurdsson
- Lab for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
- Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| | - Christian Grimm
- Lab for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
45
|
Leinonen H, Fu Z, Bull E. Neural and Müller glial adaptation of the retina to photoreceptor degeneration. Neural Regen Res 2023; 18:701-707. [DOI: 10.4103/1673-5374.354511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
46
|
Cojocaru AE, Corna A, Reh M, Zeck G. High spatial resolution artificial vision inferred from the spiking output of retinal ganglion cells stimulated by optogenetic and electrical means. Front Cell Neurosci 2022; 16:1033738. [PMID: 36568888 PMCID: PMC9780279 DOI: 10.3389/fncel.2022.1033738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022] Open
Abstract
With vision impairment affecting millions of people world-wide, various strategies aiming at vision restoration are being undertaken. Thanks to decades of extensive research, electrical stimulation approaches to vision restoration began to undergo clinical trials. Quite recently, another technique employing optogenetic therapy emerged as a possible alternative. Both artificial vision restoration strategies reported poor spatial resolution so far. In this article, we compared the spatial resolution inferred ex vivo under ideal conditions using a computational model analysis of the retinal ganglion cell (RGC) spiking activity. The RGC spiking was stimulated in epiretinal configuration by either optogenetic or electrical means. RGCs activity was recorded from the ex vivo retina of transgenic late-stage photoreceptor-degenerated mice (rd10) using a high-density Complementary Metal Oxide Semiconductor (CMOS) based microelectrode array. The majority of retinal samples were stimulated by both, optogenetic and electrical stimuli using a spatial grating stimulus. A population-level analysis of the spiking activity of identified RGCs was performed and the spatial resolution achieved through electrical and optogenetic photo-stimulation was inferred using a support vector machine classifier. The best f1 score of the classifier for the electrical stimulation in epiretinal configuration was 86% for 32 micron wide gratings and increased to 100% for 128 microns. For optogenetically activated cells, we obtained high f1 scores of 82% for 10 microns grid width for a photo-stimulation frequency of 2.5 Hz and 73% for a photo-stimulation frequency of 10 Hz. A subsequent analysis, considering only the RGCs modulated in both electrical and optogenetic stimulation protocols revealed no significant difference in the prediction accuracy between the two stimulation modalities. The results presented here indicate that a high spatial resolution can be achieved for electrical or optogenetic artificial stimulation using the activated retinal ganglion cell output.
Collapse
Affiliation(s)
| | - Andrea Corna
- Institute of Biomedical Electronics, TU Wien, Vienna, Austria
| | - Miriam Reh
- Institute for Ophthalmic Research at the University of Tübingen, Tübingen, Germany
| | - Günther Zeck
- Institute of Biomedical Electronics, TU Wien, Vienna, Austria
| |
Collapse
|
47
|
Carleton M, Oesch NW. Differences in the spatial fidelity of evoked and spontaneous signals in the degenerating retina. Front Cell Neurosci 2022; 16:1040090. [PMID: 36419935 PMCID: PMC9676928 DOI: 10.3389/fncel.2022.1040090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/20/2022] [Indexed: 07/01/2024] Open
Abstract
Vision restoration strategies aim to reestablish vision by replacing the function of lost photoreceptors with optoelectronic hardware or through gene therapy. One complication to these approaches is that retinal circuitry undergoes remodeling after photoreceptor loss. Circuit remodeling following perturbation is ubiquitous in the nervous system and understanding these changes is crucial for treating neurodegeneration. Spontaneous oscillations that arise during retinal degeneration have been well-studied, however, other changes in the spatiotemporal processing of evoked and spontaneous activity have received less attention. Here we use subretinal electrical stimulation to measure the spatial and temporal spread of both spontaneous and evoked activity during retinal degeneration. We found that electrical stimulation synchronizes spontaneous oscillatory activity, over space and through time, thus leading to increased correlations in ganglion cell activity. Intriguingly, we found that spatial selectivity was maintained in rd10 retina for evoked responses, with spatial receptive fields comparable to wt retina. These findings indicate that different biophysical mechanisms are involved in mediating feed forward excitation, and the lateral spread of spontaneous activity in the rd10 retina, lending support toward the possibility of high-resolution vision restoration.
Collapse
Affiliation(s)
- Maya Carleton
- Department of Psychology, University of California, San Diego, La Jolla, CA, United States
| | - Nicholas W. Oesch
- Department of Psychology, University of California, San Diego, La Jolla, CA, United States
- Department of Ophthalmology, University of California, San Diego, La Jolla, CA, United States
- The Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
48
|
Retinal Structure and Function in a Knock-In Mouse Model for the FAM161A-p.Arg523* Human Nonsense Pathogenic Variant. OPHTHALMOLOGY SCIENCE 2022; 3:100229. [DOI: 10.1016/j.xops.2022.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/28/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
|
49
|
Hottin C, Perron M, Roger JE. GSK3 Is a Central Player in Retinal Degenerative Diseases but a Challenging Therapeutic Target. Cells 2022; 11:cells11182898. [PMID: 36139472 PMCID: PMC9496697 DOI: 10.3390/cells11182898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.
Collapse
Affiliation(s)
- Catherine Hottin
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
50
|
Eleftheriou CG, Corona C, Khattak S, Alam NM, Ivanova E, Bianchimano P, Liu Y, Sun D, Singh R, Batoki JC, Prusky GT, McAnany JJ, Peachey NS, Romano C, Sagdullaev BT. Retinoschisin Deficiency Induces Persistent Aberrant Waves of Activity Affecting Neuroglial Signaling in the Retina. J Neurosci 2022; 42:6983-7000. [PMID: 35906066 PMCID: PMC9464019 DOI: 10.1523/jneurosci.2128-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/21/2022] Open
Abstract
Genetic disorders that present during development make treatment strategies particularly challenging because there is a need to disentangle primary pathophysiology from downstream dysfunction caused at key developmental stages. To provide a deeper insight into this question, we studied a mouse model of X-linked juvenile retinoschisis, an early-onset inherited condition caused by mutations in the Rs1 gene encoding retinoschisin (RS1) and characterized by cystic retinal lesions and early visual deficits. Using an unbiased approach in expressing the fast intracellular calcium indicator GCaMP6f in neuronal, glial, and vascular cells of the retina of RS1-deficient male mice, we found that initial cyst formation is paralleled by the appearance of aberrant spontaneous neuroglial signals as early as postnatal day 15, when eyes normally open. These presented as glutamate-driven wavelets of neuronal activity and sporadic radial bursts of activity by Müller glia, spanning all retinal layers and disrupting light-induced signaling. This study confers a role to RS1 beyond its function as an adhesion molecule, identifies an early onset for dysfunction in the course of disease, establishing a potential window for disease diagnosis and therapeutic intervention.SIGNIFICANCE STATEMENT Developmental disorders make it difficult to distinguish pathophysiology due to ongoing disease from pathophysiology due to disrupted development. Here, we investigated a mouse model for X-linked retinoschisis, a well defined monogenic degenerative disease caused by mutations in the Rs1 gene, which codes for the protein retinoschisin. We evaluated the spontaneous activity of explanted retinas lacking retinoschisin at key stages of development using the unbiased approach of ubiquitously expressing GCaMP6f in all retinal neurons, vasculature, and glia. In mice lacking RS1, we found that an array of novel phenotypes, which present around eye opening, are linked to glutamatergic neurotransmission and affect visual processing. These data identify a novel pathophysiology linked to RS1, and define a window where treatments might be best targeted.
Collapse
Affiliation(s)
- Cyril G Eleftheriou
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
| | - Carlo Corona
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
| | | | - Nazia M Alam
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
| | - Elena Ivanova
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
- Regeneron Pharmaceuticals, Tarrytown, New York 10591
| | - Paola Bianchimano
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
| | - Yang Liu
- Regeneron Pharmaceuticals, Tarrytown, New York 10591
| | - Duo Sun
- Regeneron Pharmaceuticals, Tarrytown, New York 10591
| | - Rupesh Singh
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Julia C Batoki
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Glen T Prusky
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
| | - J Jason McAnany
- Department of Ophthalmology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Neal S Peachey
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio 44195
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio 44106
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195
| | | | - Botir T Sagdullaev
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York 10605
- Regeneron Pharmaceuticals, Tarrytown, New York 10591
| |
Collapse
|