1
|
Feng Y, Wu Y, Zhu Y, He Y, Weng W. Progress in single-cell sequencing of retinal vein occlusion or ischemic hypoxic retinopathy. Exp Eye Res 2025; 257:110436. [PMID: 40414336 DOI: 10.1016/j.exer.2025.110436] [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: 02/11/2025] [Revised: 04/15/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Retinal vein occlusion (RVO) and ischemic hypoxic retinopathy (IHR) are leading cause of irreversible vision loss worldwide, compelled by complex microvascular dysfunction, neuroinflammation, and tissue hypoxia. Despite advances in imaging and treatment, a comprehensive understanding of cellular and molecular heterogeneity underlying these pathologies remains limited. Recently, single-cell RNA sequencing (scRNA-seq) has emerged as a transformative technology, enabling unprecedented resolution of cellular dynamics, transcriptomic landscapes, and intracellular communication within the retina. Single-cell technologies continue to evolve, they are poised to revolutionize our understanding of retinal vascular diseases, ultimately paving the way for precision diagnostics and targeted interventions. This technique has revolutionized our understanding regarding complex biological systems and enables proper analysis of cellular heterogeneity. This review highlights the recent progress for the application SCS to dissect the pathophysiology of RVO and IHR. Moreover, current study summarizes findings on altered gene expression endothelial cells, Muller glia, micro glia and photoreceptors under ischemic and hypoxic stress, shedding light on potential therapeutic targets and biomarkers. Furthermore, this study explores the integration of snRNA-seq, spatial transcriptomics, and multi-omics approaches to enhance the spatial and temporal mapping of retinal responses. Additionally, discuss the current challenges, including sample preservation, retinal cell-type annotation, and cross-species translation, while offering insights into future directions such as personalized medicine and regenerative strategies. This paper aims to provide clinicians and researchers with a comprehensive update on the rapidly expanding frontier of single-cell analysis in retinal ischemic diseases.
Collapse
Affiliation(s)
- Yanbing Feng
- Department of Ophthalmology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China
| | - Yibo Wu
- Department of Ophthalmology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China
| | - Yixing Zhu
- Department of Ophthalmology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China
| | - Yanyan He
- Department of Ophthalmology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China
| | - Wenqing Weng
- Department of Ophthalmology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China.
| |
Collapse
|
2
|
Li T, Kowal TJ, Zhao J, Li L, Wang Q, Ning K, Lo CH, Liu Z, Shen Y, Yu J, Jin H, Sun Y. Effect of brimonidine on retinal ganglion cell function by in vivo calcium imaging of optic nerve crush in mice. Exp Eye Res 2025; 255:110355. [PMID: 40127747 PMCID: PMC12058395 DOI: 10.1016/j.exer.2025.110355] [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: 01/26/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Brimonidine has shown neuroprotective effects in animal studies, but clinical trials failed to demonstrate effective endpoints. Here, we used a newly developed in vivo calcium imaging method to measure RGC function of brimonidine in mice optic nerve crush (ONC) models. To transduce RGCs in vivo, wild-type C57Bl/6j mice were treated with intravitreal AAV2-mSncg-jGCaMP7s, a live-cell Ca2+ tracer. RGCs are defined as 10 subtypes according to different responses to UV light. Mice were treated with topical brimonidine or placebo three times daily for two weeks after ONC. The calcium signals of live-cell RGCs were measured with the Heidelberg cSLO system. Ganglion cell complex (GCC) thickness and IOP were examined at different timepoints after treatment. RGCs were counted after RBPMS immunostaining. Live calcium imaging showed ONC significantly decreased RGC number at 14 days post-ONC compared to controls. The topical brimonidine administration changed calcium signal responses of RGC to UV light in ONC mice. It showed brimonidine partly prevented the decrease of survival ON-RGCs percent after ONC. Single RGC analysis showed a lower conversion percent of ON-RGCs to OFF-RGCs with brimonidine administration after ONC. However, no significant differences in RGC survival, IOP or GCC thickness were noted between eyes treated with brimonidine or placebo. In the acute ONC mice model, in vivo calcium imaging revealed that brimonidine maintained the Ca2+ activation of ON-RGCs to UV stimulation, inhibiting the conversion of survival ON-RGCs to OFF-RGCs. This indicates that ON-RGCs may be more resilient to acute optic nerve injury based on the calcium imaging method.
Collapse
Affiliation(s)
- Tingting Li
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Tia J Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Jingyu Zhao
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Liang Li
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Qing Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Chien-Hui Lo
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Zhiquan Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Yingchun Shen
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA
| | - Jing Yu
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China.
| | - Haiying Jin
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94306, USA; Palo Alto Veterans Administration, Palo Alto, CA, USA.
| |
Collapse
|
3
|
Xu N, Li Z, Zeng X, Jiang Y, Sun T, Liu S, Li N, Li Z, Huang Y, Wang L. Reln-Dab1 pathway mitigates retinal ganglion cell apoptosis in retinal ischemia-reperfusion injury. Cell Death Dis 2025; 16:423. [PMID: 40442071 PMCID: PMC12122947 DOI: 10.1038/s41419-025-07742-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 05/06/2025] [Accepted: 05/19/2025] [Indexed: 06/02/2025]
Abstract
Ischemia-reperfusion (I/R) injury is associated with a variety of retinal diseases, resulting in loss of the number of ganglion cells (RGCs), retinal structural disorders, and retinal dysfunction. The Reelin protein is an important regulator of neuronal migration and synaptogenesis, and the Reln signaling pathway plays an essential role in regulating the targeted projection of RGC dendrites and neuronal survival, which has not been reported in retinal I/R injury. The aim of this study was to investigate the expression, role and mechanism of Reln in retinal I/R injury. By establishing Reln-CreERT2 mTmG transgenic mice, it was observed that the expression of Reln initially decreased and then increased after retinal I/R injury. After supplementing exogenous Reelin protein and adeno-associated virus (AAV)-targeted regulation of Reln in vivo, morphological and functional experiments demonstrated its effectiveness in protecting RGCs survival, maintaining morphological integrity of the retina, and inhibiting post-injury retinal dysfunction. Furthermore, integrin β1 (Itgb1) was identified as the main receptor through which Reelin exerts neuroprotective effects while regulating retinal I/R injury repair through the Dab1-PI3K-Akt pathway. These findings provide evidence supporting Reln pathway's role in maintaining retinal homeostasis and facilitating injury repair. Moreover, these findings have significant implications for identifying new targets for preventing and treating various retinal diseases.
Collapse
Affiliation(s)
- Ning Xu
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Zongyuan Li
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Xiangwen Zeng
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yilin Jiang
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Tunan Sun
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Shuyu Liu
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Na Li
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhao Li
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Yifei Huang
- Medical School of Chinese People's Liberation Army, Beijing, China.
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China.
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China.
| | - Liqiang Wang
- Medical School of Chinese People's Liberation Army, Beijing, China.
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China.
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China.
| |
Collapse
|
4
|
Tommasini D, Yoshimatsu T, Puthussery T, Baden T, Shekhar K. Comparative transcriptomic insights into the evolution of vertebrate photoreceptor types. Curr Biol 2025; 35:2228-2239.e4. [PMID: 40250432 DOI: 10.1016/j.cub.2025.03.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/20/2025]
Abstract
To explore the molecular similarities and potential evolutionary origins of vertebrate photoreceptor types, we analyzed single-cell and -nucleus transcriptomic atlases from six vertebrate species: zebrafish, chicken, lizard, opossum, ground squirrel, and human. Comparative analyses identified conserved transcriptional signatures for the five ancestral photoreceptor types: red, blue, green, and UV cones, as well as rods. We further identified and validated molecular markers of the principal and accessory members of the tetrapod double cone. Comparative transcriptomics suggests that the principal member originated from ancestral red cones, although the origin of the accessory member is less clear. The gene expression variation among cone types mirrors their spectral order (red → green → blue → UV). We find that rods are highly dissimilar to all cone types, suggesting that rods may have diverged prior to the spectral diversification of cones.
Collapse
Affiliation(s)
- Dario Tommasini
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Takeshi Yoshimatsu
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.
| | - Teresa Puthussery
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Vision Sciences Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA; Herbert Wertheim School of Optometry and Vision Science, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Tom Baden
- Center for Sensory Neuroscience and Computation, Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, Brighton BN1 9QG, UK.
| | - Karthik Shekhar
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Vision Sciences Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA; Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
5
|
Ding C, Ndiaye PS, Campbell SR, Fry MY, Gong J, Wienbar SR, Gibbs W, Morquette P, Chao LH, Do MTH, Schwarz T. SARM1 loss protects retinal ganglion cells in a mouse model of Autosomal Dominant Optic Atrophy. J Clin Invest 2025; 135:e191315. [PMID: 40344041 DOI: 10.1172/jci191315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
Autosomal Dominant Optic Atrophy (ADOA), the most prevalent hereditary optic neuropathy, leads to retinal ganglion cell (RGC) degeneration and vision loss. ADOA is primarily caused by mutations in the OPA1 gene, which encodes a conserved GTPase important for mitochondrial inner membrane dynamics. To date, the disease mechanism remains unclear, and no therapies are available. We generated a mouse model carrying the pathogenic Opa1R290Q/+ allele that recapitulated key features of human ADOA, including mitochondrial defects, age-related RGC loss, optic nerve degeneration, and reduced RGC functions. We identified SARM1, a neurodegeneration switch, as a key driver of RGC degeneration in these mice. Sarm1 knockout nearly completely suppressed all the degeneration phenotypes without reversing mitochondrial fragmentation. Additionally, we showed that a portion of SARM1 localized within the mitochondrial intermembrane space (IMS). These findings indicated that SARM1 was activated downstream of mitochondrial dysfunction in ADOA, highlighting it as a promising therapeutic target.
Collapse
Affiliation(s)
- Chen Ding
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Papa S Ndiaye
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Sydney R Campbell
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Michelle Y Fry
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States of America
| | - Jincheng Gong
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Sophia R Wienbar
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Whitney Gibbs
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Philippe Morquette
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Luke H Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States of America
| | - Michael Tri H Do
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| | - Thomas Schwarz
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States of America
| |
Collapse
|
6
|
Venanzi AW, McGee LD, Hackam AS. Evaluating the Evidence for Neuroprotective and Axonal Regenerative Activities of Different Inflammatory Cell Types After Optic Nerve Injury. Mol Neurobiol 2025; 62:6212-6227. [PMID: 39738875 PMCID: PMC11953096 DOI: 10.1007/s12035-024-04679-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
The optic nerve contains retinal ganglion cell (RGC) axons and functions to transmit visual stimuli to the brain. Injury to the optic nerve from ischemia, trauma, or disease leads to retrograde axonal degeneration and subsequent RGC dysfunction and death, causing irreversible vision loss. Inflammatory responses to neurological damage and axonal injuries in the central nervous system (CNS) are typically harmful to neurons and prevent recovery. However, recent evidence indicates that certain inflammatory cell types and signaling pathways are protective after optic nerve injury and promote RGC survival and axonal regeneration. The objective of this review is to examine the evidence for diverse effects of inflammatory cell types on the retina and optic nerve after injury. Additionally, we highlight promising avenues for further research.
Collapse
Affiliation(s)
- Alexander W Venanzi
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10Th Ave, Rm 404, Miami, FL, 33136, USA
| | - Laura D McGee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10Th Ave, Rm 404, Miami, FL, 33136, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10Th Ave, Rm 404, Miami, FL, 33136, USA.
| |
Collapse
|
7
|
Hu Y, Grodzki LM, Bartsch U. Survival and Axonal Regeneration of Retinal Ganglion Cells in a Mouse Optic Nerve Crush Model After a Cell-Based Intravitreal Co-Administration of Ciliary Neurotrophic Factor and Glial Cell Line-Derived Neurotrophic Factor at Different Post-Lesion Time Points. Cells 2025; 14:643. [PMID: 40358167 PMCID: PMC12071274 DOI: 10.3390/cells14090643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
We recently showed, in a mouse optic nerve crush model, that a sustained cell-based intravitreal administration of ciliary neurotrophic factor (CNTF) and glial cell line-derived neurotrophic factor (GDNF) synergistically slowed the lesion-induced degeneration of retinal ganglion cells (RGCs), resulting in the presence of approximately 35% viable RGCs eight months after the lesion. However, the combinatorial neuroprotective treatment was initiated shortly after the lesion. To mimic a more clinically relevant situation, we co-administered both factors either three or five days after an intraorbital nerve crush when approximately 35% or 57% of the RGCs were degenerated, respectively. Analyses of the retinas at different time points after the lesion consistently revealed the presence of significantly more surviving RGCs in retinas co-treated with CNTF and GDNF than in retinas treated with either factor alone. For example, when the neurotrophic factors were administered five days after the nerve crush and the animals were analyzed two months after the lesion, retinas co-treated with CNTF and GDNF contained approximately 40% of the RGCs present at the start of treatment. In comparison, monotherapy with either CNTF or GDNF protected only about 15% or 10% of the RGCs present at baseline, respectively. The number of regenerating axons in the distal nerve stumps was similar in CNTF- and CNTF/GDNF-treated animals, despite the significantly higher number of rescued RGCs in the latter group. These findings have potential implications for studies aimed at developing neuroprotective treatments for optic neuropathies such as glaucoma.
Collapse
Affiliation(s)
| | | | - Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (Y.H.); (L.M.G.)
| |
Collapse
|
8
|
Brunchault MR, Hesse AM, Schaeffer J, Fröhlich A, Saintpierre A, Decourt C, Combes F, Nawabi H, Couté Y, Belin S. Proteomics-based characterization of ribosome heterogeneity in adult mouse organs. Cell Mol Life Sci 2025; 82:175. [PMID: 40272563 PMCID: PMC12022211 DOI: 10.1007/s00018-025-05708-7] [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: 05/13/2024] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
The translation process, leading to protein synthesis from mRNA, has been long thought to be invariable in all cellular organisms. Increasing evidence shows that it is finely regulated by variable features of the translation machinery. Notably, ribosomes, the functional units of protein synthesis, are suggested to display variations in their composition, depending on the developmental stage, cell type or physio-pathological context, thus hinting a new level of actionable regulation of gene expression. Yet, a comprehensive map of the heterogeneity of ribosome composition in ribosomal proteins (RPs) in different organs and tissues is not available. In this work, we explored tissue-specific ribosome heterogeneity using mass spectrometry-based quantitative proteomic characterization of ribosomal fractions purified from 14 adult mouse organs and tissues. We performed crossed clustering and statistical analyses of RP composition to highlight stable, variable and tissue-specific RPs across organs and tissues. Focusing on specific RPs, we validated their varying abundances using a targeted proteomic approach and western blot analyses, providing further insights into the tissue-specific ribosome RP signature. Finally, we investigated the origin of RP variations in ribosome fraction of the different tissues, by comparing RP relative amounts in our ribosomal proteomic dataset with their corresponding transcript abundances in three independent transcriptomic datasets. Interestingly, we found that, in some tissues, the RP abundance in purified ribosomes does not always correlate with the corresponding RP transcript level, arguing for a translational regulation of RP expression, and/or a regulated incorporation of RPs into ribosomes. Altogether, our data support the notion of a tissue-specific RP signature of ribosomes, which opens avenues to study how specific ribosomal composition provides an additional level of regulation to control gene expression in different tissues and organs.
Collapse
Affiliation(s)
- Marie R Brunchault
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Anne-Marie Hesse
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France
| | - Julia Schaeffer
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
- IBDM, CNRS, UMR 7288, Aix-Marseille Université, Marseille, France
| | - Albrecht Fröhlich
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Ana Saintpierre
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Charlotte Decourt
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Florence Combes
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France
| | - Homaira Nawabi
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France.
| | - Stephane Belin
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France.
| |
Collapse
|
9
|
Norrie JL, Lupo MS, Little DR, Shirinifard A, Mishra A, Zhang Q, Geiger N, Putnam D, Djekidel N, Ramirez C, Xu B, Dundee JM, Yu J, Chen X, Dyer MA. Latent epigenetic programs in Müller glia contribute to stress and disease response in the retina. Dev Cell 2025; 60:1199-1216.e7. [PMID: 39753128 PMCID: PMC12014377 DOI: 10.1016/j.devcel.2024.12.014] [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: 10/31/2023] [Revised: 07/09/2024] [Accepted: 12/06/2024] [Indexed: 04/24/2025]
Abstract
Previous studies have demonstrated the dynamic changes in chromatin structure during retinal development correlate with changes in gene expression. However, those studies lack cellular resolution. Here, we integrate single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) with bulk data to identify cell-type-specific changes in chromatin structure during human and murine development. Although promoter activity is correlated with chromatin accessibility, we discovered several hundred genes that were transcriptionally silent but had accessible chromatin at their promoters. Most of those silent/accessible gene promoters were in Müller glial cells, which function to maintain retinal homeostasis and respond to stress, injury, or disease. We refer to these as "pliancy genes" because they allow the Müller glia to rapidly change their gene expression and cellular state in response to retinal insults. The Müller glial cell pliancy program is established during development, and we demonstrate that pliancy genes are important for regulating inflammation in the murine retina in vivo.
Collapse
Affiliation(s)
- Jackie L Norrie
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marybeth S Lupo
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Danielle R Little
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Akhilesh Mishra
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Qiong Zhang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Natalie Geiger
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel Putnam
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nadhir Djekidel
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Cody Ramirez
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jacob M Dundee
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiang Yu
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael A Dyer
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
10
|
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
|
11
|
Allen AE, Hahn J, Richardson R, Pantiru A, Mouland J, Babu A, Baño-Otalora B, Monavarfeshani A, Yan W, Williams C, Wynne J, Rodgers J, Milosavljevic N, Orlowska-Feuer P, Storchi R, Sanes JR, Shekhar K, Lucas RJ. Altered proportions of retinal cell types and distinct visual codes in rodents occupying divergent ecological niches. Curr Biol 2025; 35:1446-1458.e5. [PMID: 40043699 DOI: 10.1016/j.cub.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/04/2024] [Accepted: 02/07/2025] [Indexed: 04/10/2025]
Abstract
Vertebrate retinas share a basic blueprint comprising 5 neuronal classes arranged according to a common wiring diagram. Yet, vision is aligned with species differences in behavior and ecology, raising the question of how evolution acts on this circuit to adjust its computational characteristics. We address that problem by comparing the thalamic visual code and retinal cell composition in closely related species occupying different niches: Rhabdomys pumilio, which are day-active murid rodents, and nocturnal laboratory mice (Mus musculus). Using high-density electrophysiological recordings, we compare visual responses at both single-unit and population levels in the thalamus of these two species. We find that Rhabdomys achieves a higher spatiotemporal resolution visual code through the selective expansion of information channels characterized by non-linear spatiotemporal summation. Comparative analysis of single-cell transcriptomic atlases reveals that this difference originates with the increased relative abundance of retinal bipolar and ganglion cell types supporting OFF and ON-OFF responses. These findings demonstrate that evolution may drive changes in neural computation by adjusting the proportions of shared cell types rather than inventing new types and show the power of matching high-density physiological recordings with transcriptomic cell atlases to study evolution in the brain.
Collapse
Affiliation(s)
- Annette E Allen
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK.
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rose Richardson
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Andreea Pantiru
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Josh Mouland
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Aadhithyan Babu
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Beatriz Baño-Otalora
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Aboozar Monavarfeshani
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Wenjun Yan
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Christopher Williams
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Jonathan Wynne
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Jessica Rodgers
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Nina Milosavljevic
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Patrycja Orlowska-Feuer
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Riccardo Storchi
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Joshua R Sanes
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, Vision Science Graduate Group, Center for Computational Biology, Biophysics Graduate Group, California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA
| | - Robert J Lucas
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
12
|
Yang S, Li C, Wang X, Huang T, Qian C, Li Q, Zhao L, Zhou S, Ding C, Nie R, Saijilafu, Hong Y, Liu C, Zhou F. Roles of Kdm6a and Kdm6b in Regulation of Mammalian Neural Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405537. [PMID: 39951327 PMCID: PMC12021076 DOI: 10.1002/advs.202405537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 01/21/2025] [Indexed: 02/16/2025]
Abstract
Epigenetic regulation of neuronal transcriptomic landscape is emerging to be a key coordinator of mammalian neural regeneration. The roles of two histone 3 lysine 27 (H3K27) demethylases, Kdm6a/b, in controlling neuroprotection and axon regeneration are investigated here. Deleting either Kdm6a or Kdm6b leads to enhanced sensory axon regeneration in the peripheral nervous system (PNS), whereas in the central nervous system (CNS), only deleting Kdm6a in retinal ganglion cells (RGCs) significantly enhances optic nerve regeneration. Moreover, both Kdm6a and Kdm6b function to regulate RGC survival but with different mechanisms. Mechanistically, Kdm6a regulates RGC regeneration via distinct pathway from that of Pten, and co-deleting Kdm6a and Pten results in long distance optic nerve regeneration passing the optic chiasm. In addition, RNA-seq profiling reveals that Kdm6a deletion switches the RGC transcriptomics into a developmental-like state and suppresses several known repressors of neural regeneration. Klf4 is identified as a direct downstream target of Kdm6a-H3K27me3 signaling in both sensory neurons and RGCs to regulate axon regeneration. These findings not only reveal different roles of Kdm6a and Kdm6b in regulation of neural regeneration and their underlying mechanisms, but also identify Kdm6a-mediated histone demethylation signaling as a novel epigenetic target for supporting CNS neural regeneration.
Collapse
Affiliation(s)
- Shu‐Guang Yang
- Center for Translational Neural Regeneration ResearchSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
- Department of Orthopedic SurgeryThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Chang‐Ping Li
- Center for Translational Neural Regeneration ResearchSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
- Key Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesInstitute for Stem Cell and RegenerationChinese Academy of SciencesBeijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xue‐Wei Wang
- Department of Orthopedic SurgeryThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- Byrd Alzheimer's Center and Research InstituteUniversity of South FloridaTampaFL33613USA
- Department of Molecular MedicineUniversity of South Florida Morsani College of MedicineTampaFL33612USA
| | - Tao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoning110016China
| | - Cheng Qian
- Department of Orthopedic SurgeryThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Qiao Li
- Department of Orthopedic SurgeryThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Ling‐Rui Zhao
- Department of Orthopedic SurgeryThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Si‐Yu Zhou
- Center for Translational Neural Regeneration ResearchSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
| | - Chen‐Yun Ding
- Center for Translational Neural Regeneration ResearchSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
| | - Rui Nie
- Center for Translational Neural Regeneration ResearchSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
| | - Saijilafu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouZhejiang310015China
| | - Yu‐Cai Hong
- Department of Emergency MedicineSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
| | - Chang‐Mei Liu
- Key Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesInstitute for Stem Cell and RegenerationChinese Academy of SciencesBeijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Feng‐Quan Zhou
- Center for Translational Neural Regeneration ResearchSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
- Department of Orthopedic SurgeryThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- The Solomon H. Department of NeuroscienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| |
Collapse
|
13
|
Gallego-Ortega A, Galindo-Romero C, Vidal-Villegas B, Bernal-Garro JM, de la Villa P, Avilés-Trigueros M, Vidal-Sanz M. The action of 7,8-dihydroxyflavone preserves retinal ganglion cell survival and visual function via the TrkB pathway in NMDA-induced retinal excitotoxicity. Biomed Pharmacother 2025; 185:117944. [PMID: 40056826 DOI: 10.1016/j.biopha.2025.117944] [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: 11/26/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025] Open
Abstract
PURPOSE To analyze the response of different retinal ganglion cell (RGC) populations to NMDA-induced retinal excitotoxicity and the effect of an intraperitoneal treatment with 7,8-Dihydroxyflavone (DHF), a potent selective TrkB agonist. METHODS Adult albino rats were treated the day prior to NMDA injection and the three following days with intraperitoneal vehicle (1 %DMSO in 0.09 %NaCl) or DHF (5 mg/kg in vehicle) injections. DHF-afforded protection was studied in the population of Brn3a+RGCs, OPN+RGCs (α-RGCs), OPN+ Tbr2+RGCs (αONs-RGCs), OPN+ Tbr2-Brn3a-RGCs (αONt-RGCs) and OPN+Brn3a+RGCs (αOFF-RGCs) at 3,7,14, or 21 days. The functional response was analyzed longitudinally with full-field electroretinograms. The mechanisms underlying DHF-afforded neuroprotection were assessed by western blot (WB) analysis of the levels of phosphorylated and total TrkB, phosphatidylinositol 3 kinase (PIK3/AKT) and mitogen-activated protein kinase (MAPK). RESULTS NMDA intravitreal injection resulted in a significant diminution of the mean amplitudes of the pSTR and b-waves, as well as in severe depletion of all RGCs studied except αONt-RGCs. DHF treatment resulted in rescued mean amplitudes of the pSTR and b-waves up to 21 days after NMDA. WB analysis revealed an increase in p-TrkB which correlates to the increase of TRKB protein and an increase in normalized pAKT/AKT. pMAPK/MAPK was upregulated earlier and significantly higher in DHF-treated retinas. DHF afforded survival of up to 49 % of the Brn3a+RGCs versus 25 % of the vehicle group at 21 days after NMDA, and improved survival of the α-RGC and αONs-RGCs but did not rescue the αOFF-RGCs. CONCLUSION Different RGC types exhibit variable susceptibilities to NMDA injury, and DHF-mediated activation of TrkB affords neuroprotection.
Collapse
Affiliation(s)
- Alejandro Gallego-Ortega
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain; Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, USA.
| | - Caridad Galindo-Romero
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain.
| | - Beatriz Vidal-Villegas
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain; Moorfields Eye Hospital, London, United Kingdom
| | - José Manuel Bernal-Garro
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Pedro de la Villa
- Department of Systems Biology, Laboratory of Visual Neurophysiology, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Marcelino Avilés-Trigueros
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Manuel Vidal-Sanz
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| |
Collapse
|
14
|
Olguin AGR, Rochon PL, Theriault C, Brown T, Yao H, Cayouette M, Cook EP, Krishnaswamy A. Cadherin 4 assembles a family of color-preferring retinal circuits that respond to light offset. Curr Biol 2025; 35:1298-1310.e7. [PMID: 40081378 DOI: 10.1016/j.cub.2025.02.008] [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: 05/10/2024] [Revised: 11/22/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025]
Abstract
Retinal interneurons and projection neurons (retinal ganglion cells, RGCs) connect in specific combinations in a specialized neuropil called the inner plexiform layer (IPL). The IPL is divided into multiple sublaminae, with neurites of each neuronal type confined to one or a few layers. This laminar specificity is a major determinant of circuit specificity and circuit function. Using a combination of approaches, we show that RGCs targeting IPL sublaminae 1 and 3a (s1-s3a) express the cell adhesion molecule cadherin 4 (Cdh4). Using calcium imaging and iterative immunostaining, we classified Cdh4 RGCs into nine types that each encode unique aspects of dark visual stimuli. Cdh4 loss selectively disrupted the layer targeting of these RGCs, reduced their synaptic inputs from interneurons, and severely altered their visual responses. Overexpression of Cdh4 in other retinal neurons directed their neurites to s1-s3a through homophilic interactions. Taken together, these results demonstrate that Cdh4 is a novel layer-targeting system for nearly a quarter of all RGCs.
Collapse
Affiliation(s)
| | - Pierre-Luc Rochon
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | | | - Thomas Brown
- Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Houwen Yao
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Michel Cayouette
- Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Erik P Cook
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
15
|
Wang Z, Zhao C, Xu S, McCracken S, Apte RS, Williams PR. Energetic diversity in retinal ganglion cells is modulated by neuronal activity and correlates with resilience to degeneration. RESEARCH SQUARE 2025:rs.3.rs-5989609. [PMID: 40162221 PMCID: PMC11952644 DOI: 10.21203/rs.3.rs-5989609/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Neuronal function requires high energy expenditure that is likely customized to meet specific signaling demands. However, little is known about diversity of metabolic homeostasis among divergently-functioning types of neurons. To this end, we examined retinal ganglion cells (RGCs), a population of closely related, yet electrophysiologically distinct excitatory projection neurons. Using in vivo 2-photon imaging to measure ATP with single cell resolution, we identified differential homeostatic energy maintenance in the RGC population that correspond to distinct RGC types. In the presence of circuit activity, the most active RGC type (Alpha RGCs), had lower homeostatic ATP levels than other types and exhibited the greatest magnitude of ATP decline when ATP synthesis was inhibited. By simultaneously manipulating circuit activity and mitochondrial function, we found that while oxidative phosphorylation was required to meet ATP demands during circuit activity, it was expendable to maintain resting ATP levels. We also examined ATP signatures associated with survival and injury response after axotomy and report a correlation between low homeostatic ATP and increased survival. In addition, we observed transient ATP increases in RGCs following axon injury. Together, these findings identify diversity of energy handling capabilities of dynamically active neurons with implications for neuronal resilience.
Collapse
Affiliation(s)
- Zelun Wang
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
- Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Zhao
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shelly Xu
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sean McCracken
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
- Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rajendra S. Apte
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Philip R. Williams
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, US
| |
Collapse
|
16
|
Sarich SC, Sreevidya VS, Udvadia AJ, Svoboda KR, Gutzman JH. The transcription factor Jun is necessary for optic nerve regeneration in larval zebrafish. PLoS One 2025; 20:e0313534. [PMID: 40063628 PMCID: PMC11892826 DOI: 10.1371/journal.pone.0313534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 10/25/2024] [Indexed: 05/13/2025] Open
Abstract
Damage to the axons of the adult mammalian central nervous system (CNS) from traumatic injury or neurodegenerative diseases often results in permanent loss of function due to failure of axons to regenerate. Zebrafish, however, can express regeneration-associated genes to revert CNS neurons to a growth-competent state and regenerate damaged axons to functionality. An established model for CNS axon regeneration is optic nerve injury in zebrafish, where it was previously shown that thousands of genes are temporally expressed during the regeneration time course. It is likely that hubs of key transcription factors, rather than individual factors regulate the temporal clusters of expression after injury to facilitate cell survival, regrowth, and synaptic targeting in the brain. One transcription factor of interest in orchestrating CNS axon regeneration is jun. However, it remains unclear if CNS regeneration can progress without Jun. To test this, a transgenic zebrafish line was developed to express a heat-shock inducible dominant negative Jun. Induction of dominant negative Jun downregulated endogenous jun expression and larvae with functional jun knockdown demonstrated impaired retinal ganglion cell axon regeneration. Analysis of select putative Jun target genes, previously shown to be upregulated in adult zebrafish optic nerve regeneration, demonstrated that with functional Jun knockdown, atf3 and ascl1a were significantly downregulated, and sox11a was upregulated at distinct time points. These results position jun as a key regulator for successful optic nerve regeneration, further distinguish the regeneration program from development, and advance our knowledge for the formation of future therapies to treat CNS damage.
Collapse
Affiliation(s)
- Sarah C. Sarich
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Virinchipuram S. Sreevidya
- Joseph J. Zilber College of Public Health, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Ava J. Udvadia
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| | - Kurt R. Svoboda
- Joseph J. Zilber College of Public Health, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Jennifer H. Gutzman
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
17
|
Hu M, Veldman MB. Srebf2 mediates successful optic nerve axon regeneration via the mevalonate synthesis pathway. Mol Neurodegener 2025; 20:28. [PMID: 40045384 PMCID: PMC11883989 DOI: 10.1186/s13024-025-00807-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Axon regeneration within the mammalian central nervous system is extremely limited. In optic neuropathy conditions like glaucoma, the inability of retinal ganglion cell (RGC) axons to regenerate is a major impediment to functional recovery. In contrast, adult teleost fish such as zebrafish can fully regenerate RGC axons enabling visual recovery from optic nerve (ON) injury making it an ideal model to probe the mechanisms of successful axon regeneration. METHODS Laser Capture Microdissection followed by RNA-sequencing (LCM-seq) was used to identify genes and pathways differentially expressed in RGCs during ON regeneration. We validate these findings by in situ hybridization and qRT-PCR. Using loss- and gain-of-function experiments we demonstrate the necessity of srebf2 for efficient ON regeneration and recovery of visual function. Finally, we use LCM-seq coupled with experimental manipulations to identify downstream srebf2 target genes and test the role of hmgcra/b and mevalonate in this process. Statistical analysis was performed using Student's t-test, two-way ANOVA, or repeated measures with appropriate post-hoc tests where applicable. RESULTS LCM-seq comparison of uninjured versus 3-day post ON injury RGCs identified significant upregulation of the cholesterol synthesis pathway during axon regeneration. The master regulator of this pathway, the transcription factor srebf2, is upregulated throughout the regeneration phase. Chemical inhibition or morpholino-based gene knockdown of srebf2 decreased axon regeneration into the ON and optic tectum and delayed recovery of visual behavior over the course of normal optic nerve regeneration without causing a significant loss of RGCs. Constitutively active srebf2 can fully rescue axon regeneration and visual behavior losses caused by inhibition of endogenous srebf2 but does not accelerate regeneration compared to the control group. LCM-seq confirms the expected regulation of predicted srebf2 target genes after loss- or gain-of-function in vivo. Downstream of srebf2, hmgcra/b knockdown or simvastatin treatment delayed axon regeneration and this effect was rescued by supplemental mevalonate. Mevalonate treatment alone was sufficient to accelerate ON regeneration. CONCLUSIONS These results demonstrate that srebf2 and the downstream mevalonate synthesis pathway plays an important role in regulating efficient axon regeneration in the zebrafish visual system. Involvement of this pathway should be closely examined in failed mammalian ON regeneration.
Collapse
Affiliation(s)
- Mengming Hu
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Matthew B Veldman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, USA.
| |
Collapse
|
18
|
Zhang X, Yang C, Zhang C, Wu J, Zhang X, Gao J, Wang X, Chan LT, Zhou Y, Chen Y, Tam SST, Chen S, Ma Y, Yung WH, Duan L, Jiang L, Wang Y, Liu K. Functional optic tract rewiring via subtype- and target-specific axonal regeneration and presynaptic activity enhancement. Nat Commun 2025; 16:2174. [PMID: 40038284 PMCID: PMC11880380 DOI: 10.1038/s41467-025-57445-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
Mechanisms underlying functional axonal rewiring after adult mammalian central nervous system (CNS) injuries remain unclear partially due to limited models. Here we develop a mouse intracranial pre-olivary pretectal nucleus (OPN) optic tract injury model and demonstrate that Pten/Socs3 knockout and CNTF expression in retinal ganglion cells (RGCs) promotes optic tract regeneration and OPN reinnervation. Revealed by transmission electron microscopy, trans-synaptic labeling, and electrophysiology, functional synapses are formed in OPN mainly by intrinsically photosensitive RGCs, thereby partially restoring the pupillary light reflex (PLR). Moreover, combining with Lipin1 knockdown accelerates the recovery and achieves functional reconnection after chronic injury. PLR can be further boosted by increasing RGC photosensitivity with melanopsin overexpression, and it can also be enhanced by treatment of a voltage-gated calcium channel modulator to augment presynaptic release. These findings highlight the importance of neuronal types and presynaptic activity for functional reconnection after CNS injuries.
Collapse
Grants
- AoE/M-604/16 Research Grants Council, University Grants Committee (RGC, UGC)
- C6034-21G Research Grants Council, University Grants Committee (RGC, UGC)
- T13-602/21N Research Grants Council, University Grants Committee (RGC, UGC)
- 16102524 Research Grants Council, University Grants Committee (RGC, UGC)
- JLFS/M-604/24 Research Grants Council, University Grants Committee (RGC, UGC)
- PDFS2223-6S04 Research Grants Council, University Grants Committee (RGC, UGC)
- C4001-22Y Research Grants Council, University Grants Committee (RGC, UGC)
- C4002-21EF Research Grants Council, University Grants Committee (RGC, UGC)
- C4014-23G Research Grants Council, University Grants Committee (RGC, UGC)
- CRS_CUHK405/23 Research Grants Council, University Grants Committee (RGC, UGC)
- ITCPD/17-9 Innovation and Technology Commission (ITF)
- 82171384 National Natural Science Foundation of China (National Science Foundation of China)
Collapse
Affiliation(s)
- Xin Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Chao Yang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | - Chengle Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Junqiang Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiang Zhang
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jiayang Gao
- School of Life Sciences, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, Sha Tin, China
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Leung Ting Chan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yiren Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yujun Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Sindy Sing Ting Tam
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shuhang Chen
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuqian Ma
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wing-Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, Sha Tin, China
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, Sha Tin, China
| | - Yiwen Wang
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China.
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
19
|
Sahoo PK, Agrawal M, Hanovice N, Ward PJ, Desai M, Smith TP, SiMa H, Dulin JN, Vaughn LS, Tuszynski MH, Welshhans K, Benowitz LI, English AW, Houle JD, Twiss JL. Disruption of G3BP1 granules promotes mammalian CNS and PNS axon regeneration. Proc Natl Acad Sci U S A 2025; 122:e2411811122. [PMID: 40014573 PMCID: PMC11892601 DOI: 10.1073/pnas.2411811122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/25/2025] [Indexed: 03/01/2025] Open
Abstract
Depletion or inhibition of core stress granule proteins, G3BP1 in mammals and TIAR-2 in Caenorhabditis elegans, increases the growth of spontaneously regenerating axons. Inhibition of G3BP1 by expression of its acidic or "B-domain" accelerates axon regeneration after nerve injury, bringing a potential therapeutic strategy for peripheral nerve repair. Here, we asked whether G3BP1 inhibition is a viable strategy to promote regeneration in injured mammalian central nervous system (CNS) where axons do not regenerate spontaneously. G3BP1 B-domain expression was found to promote axon regeneration in the transected spinal cord provided with a permissive peripheral nerve graft (PNG) as well as in crushed optic nerve. Moreover, a cell-permeable peptide (CPP) to a subregion of B-domain (rodent G3BP1 amino acids 190 to 208) accelerated axon regeneration after peripheral nerve injury and promoted regrowth of reticulospinal axons into the distal transected spinal cord through a bridging PNG. G3BP1 CPP promoted axon growth from rodent and human neurons cultured on permissive substrates, and this function required alternating Glu/Asp-Pro repeats that impart a unique predicted tertiary structure. The G3BP1 CPP disassembles axonal G3BP1, G3BP2, and FMRP, but not FXR1, granules and selectively increases axonal protein synthesis in cortical neurons. These studies identify G3BP1 granules as a key regulator of axon growth in CNS neurons and demonstrate that disassembly of these granules promotes retinal axon regeneration in injured optic nerve and reticulospinal axon elongation into permissive environments after CNS injury. This work highlights G3BP1 granule disassembly as a potential therapeutic strategy for enhancing axon growth and neural repair.
Collapse
Affiliation(s)
- Pabitra K. Sahoo
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Rutgers University–Newark, Newark, NJ07102
| | - Manasi Agrawal
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Rutgers University–Newark, Newark, NJ07102
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH44242
| | - Nicholas Hanovice
- Departments of Neurosurgery and Ophthalmology, Boston Children’s Hospital, Cambridge, MA02115
| | - Patricia J. Ward
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, GA30332
| | - Meghal Desai
- Department of Biological Sciences, Rutgers University–Newark, Newark, NJ07102
| | - Terika P. Smith
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
| | - HaoMin SiMa
- Departments of Neurosurgery and Ophthalmology, Boston Children’s Hospital, Cambridge, MA02115
| | - Jennifer N. Dulin
- Department of Neurosciences, University of California–San Diego, La Jolla, CA92093
- Department of Biology, Texas A&M University, College Station, TX77843
| | - Lauren S. Vaughn
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
| | - Mark H. Tuszynski
- Department of Neurosciences, University of California–San Diego, La Jolla, CA92093
| | - Kristy Welshhans
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC29208
| | - Larry I. Benowitz
- Departments of Neurosurgery and Ophthalmology, Boston Children’s Hospital, Cambridge, MA02115
| | - Arthur W. English
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, GA30332
| | - John D. Houle
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Jeffery L. Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC29208
| |
Collapse
|
20
|
Avilés EC, Wang SK, Patel S, Cordero S, Shi S, Lin L, Kefalov VJ, Goodrich LV, Cepko CL, Xue Y. ERG responses to high-frequency flickers require FAT3 signaling in mouse retinal bipolar cells. J Gen Physiol 2025; 157:e202413642. [PMID: 39903280 PMCID: PMC11793021 DOI: 10.1085/jgp.202413642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/17/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Vision is initiated by the reception of light by photoreceptors and subsequent processing via downstream retinal neurons. Proper circuit organization depends on the multifunctional tissue polarity protein FAT3, which is required for amacrine cell connectivity and retinal lamination. Here, we investigated the retinal function of Fat3 mutant mice and found decreases in both electroretinography and perceptual responses to high-frequency flashes. These defects did not correlate with abnormal amacrine cell wiring, pointing instead to a role in bipolar cell subtypes that also express FAT3. The role of FAT3 in the response to high temporal frequency flashes depends upon its ability to transduce an intracellular signal. Mechanistically, FAT3 binds to the synaptic protein PTPσ intracellularly and is required to localize GRIK1 to OFF-cone bipolar cell synapses with cone photoreceptors. These findings expand the repertoire of FAT3's functions and reveal its importance in bipolar cells for high-frequency light response.
Collapse
Affiliation(s)
- Evelyn C. Avilés
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sean K. Wang
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Sarina Patel
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sebastian Cordero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Shuxiang Shi
- Lingang Laboratory, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lucas Lin
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Vladimir J. Kefalov
- Gavin Herbert Eye Institute and Center for Translational Vision Research, University of California, Irvine, Irvine, CA, USA
| | - Lisa V. Goodrich
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Constance L. Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Yunlu Xue
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Lingang Laboratory, Shanghai, China
| |
Collapse
|
21
|
Santos JR, Li C, Andries L, Masin L, Nuttin B, Reinhard K, Moons L, Cuntz H, Farrow K. Predicting the Regenerative Potential of Retinal Ganglion Cells Based on Developmental Growth Trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640775. [PMID: 40060504 PMCID: PMC11888416 DOI: 10.1101/2025.02.28.640775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Retinal ganglion cells in the mammalian central nervous system fail to regenerate following injury, with the capacity to survive and regrow varying by cell type. This variability may be linked to differences in developmental programs that overlap with the genetic pathways that mediate regeneration. To explore this correlation, we compared the structural changes in mouse retinal ganglion cells during development with those occurring after axonal injury. The dendritic trees of over 1,000 ganglion cells were reconstructed at different developmental stages, revealing that each cell type follows a distinct timeline. ON-sustained (sONα) cells reach maturity by P14, whereas ON-transient (tONα) cells achieve their maximum dendritic size by P10. Modeling of the dendritic changes indicate that while sONα and tONα follow similar growth programs the onset of growth was later in sONα. After optic nerve crush, the remodeling of dendritic architecture differed between the two cell-types. sONα cells exhibited rapid dendritic shrinkage, while tONα cells shrank more gradually with changes in branching features. Following injury, sONα cells reverted to an earlier developmental state than tONα cells. In addition, after co-deletion of PTEN and SOC3, neurons appeared to regress further back in developmental time. Our results provide evidence that a ganglion cell's resilience to injury and regenerative potential is predicted by its maturation timeline. Understanding these intrinsic differences could inform targeted neuroprotective interventions.
Collapse
Affiliation(s)
- Joana Rf Santos
- VIB - Neuro-Electronics Research Flanders, Leuven, Belgium
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
| | - Chen Li
- VIB - Neuro-Electronics Research Flanders, Leuven, Belgium
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
- Current address: Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Lien Andries
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
| | - Luca Masin
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
| | - Bram Nuttin
- VIB - Neuro-Electronics Research Flanders, Leuven, Belgium
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
| | - Katja Reinhard
- VIB - Neuro-Electronics Research Flanders, Leuven, Belgium
- Current address: SISSA, Trieste, Italy
| | - Lieve Moons
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
| | - Hermann Cuntz
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
- Ernst Strüngmann Institute (ESI) for Neuroscience in cooperation with the Max Planck Society, 60528 Frankfurt am Main, Germany
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University Gießen, 35392 Giessen, Germany
| | - Karl Farrow
- VIB - Neuro-Electronics Research Flanders, Leuven, Belgium
- KU Leuven, Department of Biology & Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
22
|
Duda S, Block CT, Pradhan DR, Arzhangnia Y, Klaiber A, Greschner M, Puller C. Spatial distribution and functional integration of displaced retinal ganglion cells. Sci Rep 2025; 15:7123. [PMID: 40016499 PMCID: PMC11868576 DOI: 10.1038/s41598-025-91045-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
The retina contains distinct types of ganglion cells, which form mosaics with cells of each type at each position of the visual field. Displaced retinal ganglion cells (dRGCs) occur with cell bodies in the inner nuclear layer (INL), and regularly placed RGCs with cell bodies in the ganglion cell layer. An example of mammalian dRGCs are M1-type intrinsically photosensitive ganglion cells (ipRGCs). Little is known, however, about their relationship with regularly placed ipRGCs. We identified mouse ipRGC types M1, M2, and M4/sONɑ by immunohistochemistry and light microscopy. Reconstruction of immunolabeled mosaics from M1 and sONɑ RGCs indicated that dRGCs tiled the retina with their regular RGC partners. Multi-electrode array recordings revealed conventional receptive fields of displaced sONɑ RGCs which fit into the mosaic of their regular counterparts. An RGC distribution analysis showed type-specific dRGC patterns which followed neither the global density distribution of all RGCs nor the local densities of corresponding cell types. The displacement of RGC bodies into the INL occurs in a type-dependent manner, where dRGCs are positioned to form complete mosaics with their regular partners. Our data suggest that dRGCs and regular RGCs serve the same functional role within their corresponding population of RGCs.
Collapse
Affiliation(s)
- Sabrina Duda
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Christoph T Block
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Dipti R Pradhan
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Yousef Arzhangnia
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Alina Klaiber
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Martin Greschner
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Christian Puller
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany.
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior - caesar, Bonn, Germany.
| |
Collapse
|
23
|
Zapadka TE, Tran NM, Demb JB. Optic nerve injury impairs intrinsic mechanisms underlying electrical activity in a resilient retinal ganglion cell. J Physiol 2025. [PMID: 39985791 DOI: 10.1113/jp286414] [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: 02/09/2024] [Accepted: 01/22/2025] [Indexed: 02/24/2025] Open
Abstract
Retinal ganglion cells (RGCs) are the sole output neurons of the retina and convey visual information to the brain via their axons in the optic nerve. Following injury to the optic nerve, RGC axons degenerate and many cells die. For example, a model of axon injury, the optic nerve crush (ONC), kills ∼80% of RGCs after 2 weeks. Surviving cells are biased towards 'resilient' types, including several with sustained firing to light stimulation. RGC survival may depend on activity, and there is limited understanding of how or why activity changes following optic nerve injury. Here we quantified the electrophysiological properties of a highly resilient RGC type, the sustained ON-Alpha (AlphaONS) RGC, 7 days after ONC with extracellular and whole-cell patch clamp recording. Both light- and current-driven firing were reduced after ONC, but synaptic inputs were largely intact. Resting membrane potential and input resistance were relatively unchanged, while voltage-gated currents were impaired, including a reduction in voltage-gated sodium channel current and channel density in the axon initial segment. Hyperpolarization or chelation of intracellular calcium partially rescued firing rates. Extracellular recordings at 3 days following ONC showed normal light-evoked firing from AlphaONS RGCs and other Alpha RGCs, including susceptible types. These data suggest that an injured resilient RGC reduces its activity by 1 week after injury as a consequence of reduced voltage-gated current and downregulation of intrinsic excitability via a Ca2+-dependent mechanism. Reduced excitability may be due to degradation of the axon but could also be energetically beneficial, preserving energy for survival and regeneration. KEY POINTS: Retinal ganglion cell (RGC) types show diverse rates of survival after axon injury. A resilient RGC type (sustained ON-Alpha RGC) maintains its synaptic inputs 1 week after injury. The resilient RGC type shows diminished firing and reduced expression of axon initial segment genes 1 week after injury Activity deficits reflect dysfunction of intrinsic properties (Na+ channels, intracellular Ca2+), not changes to synaptic input. Both resilient and susceptible Alpha RGC types show intact firing at 3 days after injury, suggesting that activity at this time point does not predict resilience.
Collapse
Affiliation(s)
- Thomas E Zapadka
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Nicholas M Tran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan B Demb
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
24
|
Liu D, Webber HC, Bian F, Xu Y, Prakash M, Feng X, Yang M, Yang H, You IJ, Li L, Liu L, Liu P, Huang H, Chang CY, Liu L, Shah SH, La Torre A, Welsbie DS, Sun Y, Duan X, Goldberg JL, Braun M, Lansky Z, Hu Y. Optineurin-facilitated axonal mitochondria delivery promotes neuroprotection and axon regeneration. Nat Commun 2025; 16:1789. [PMID: 39979261 PMCID: PMC11842812 DOI: 10.1038/s41467-025-57135-8] [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: 04/10/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Optineurin (OPTN) mutations are linked to amyotrophic lateral sclerosis (ALS) and normal tension glaucoma (NTG), but a relevant animal model is lacking, and the molecular mechanisms underlying neurodegeneration are unknown. We find that OPTN C-terminus truncation (OPTN∆C) causes late-onset neurodegeneration of retinal ganglion cells (RGCs), optic nerve (ON), and spinal cord motor neurons, preceded by a decrease of axonal mitochondria in mice. We discover that OPTN directly interacts with both microtubules and the mitochondrial transport complex TRAK1/KIF5B, stabilizing them for proper anterograde axonal mitochondrial transport, in a C-terminus dependent manner. Furthermore, overexpressing OPTN/TRAK1/KIF5B prevents not only OPTN truncation-induced, but also ocular hypertension-induced neurodegeneration, and promotes robust ON regeneration. Therefore, in addition to generating animal models for NTG and ALS, our results establish OPTN as a facilitator of the microtubule-dependent mitochondrial transport necessary for adequate axonal mitochondria delivery, and its loss as the likely molecular mechanism of neurodegeneration.
Collapse
Affiliation(s)
- Dong Liu
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Hannah C Webber
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Fuyun Bian
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yangfan Xu
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences; Shanghai Research Center of Ophthalmology and Optometry, Shanghai, P.R. China
| | - Manjari Prakash
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Prague West, Czechia
| | - Xue Feng
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ming Yang
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Hang Yang
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - In-Jee You
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Liang Li
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Liping Liu
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Pingting Liu
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Haoliang Huang
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Chien-Yi Chang
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Liang Liu
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sahil H Shah
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - Derek S Welsbie
- Viterbi Family Department of Ophthalmology, University of California San Diego, San Diego, CA, USA
| | - Yang Sun
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey Louis Goldberg
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Prague West, Czechia
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Prague West, Czechia.
| | - Yang Hu
- Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute at Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
25
|
You T, Yang Y, A L, Cheng X, Lin X, Liang Q, Ge L, Xie J, Chen S, Liu N, He J, Xu H, Ma X. IFNγ preconditioning improves neuroprotection of MSC-derived vesicles on injured retinal ganglion cells by suppressing microglia activation via miRNA-dependent ribosome activity. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:87-111. [PMID: 40206798 PMCID: PMC11977360 DOI: 10.20517/evcna.2024.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
Aim: Microglial activation plays a pivotal role in the pathogenesis of retinal ganglion cell (RGC) degeneration resulting from optic nerve crush (ONC). Small extracellular vesicles (sEVs) secreted by mesenchymal stem cells (MSCs) have the potential to prevent retinal degeneration by modulating microglial activation. In this study, we elucidated the specific effects of sEVs derived from IFN-γ-primed MSCs on the phenotypic transition of microglia and the associated pathways in ONC mice. Methods: The ONC mice model was established and administered intravitreal injection with the sEVs derived from native MSCs (native sEVs) and the sEVs derived from MSCs primed with IFN-γ (IFNγ-sEVs). Their respective effects on the survival of the retinal ganglion cells (RGCs) and the transition of microglia phenotypes were determined through visual function testing and immunohistochemical staining. Combined with mRNA seq and microRNA seq techniques, we elucidated the mechanism of modulation of microglia phenotypic transformation by sEVs derived from MSCs primed by IFNγ. Results: It demonstrated that IFNγ-sEVs exhibited superior protective effects against RGC loss and reduced inflammatory responses in the ONC retina compared to native sEVs. Both types of sEVs promoted microglia activation to disease-associated microglia (DAM) phenotype, while IFNγ-sEVs especially suppressed interferon-responsive microglia (IRM) activation during RGCs degeneration. Subsequent miRNA sequencing suggested that miR-423-5p, which exhibited the most significant differential expression between the two sEVs types and elevated expression in IFNγ-sEVs, inhibited the expression of IRM and ribosomal genes. Conclusion: These findings suggest that IFN-γ-preconditioned MSCs may enhance sEVs of neuroprotection on RGCs by suppressing IRM activation through the secretion of sEVs containing specific microRNAs in ONC mice.
Collapse
Affiliation(s)
- Tianjing You
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian 116014, Liaoning, China
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Yuanxing Yang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Luodan A
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Xuan Cheng
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Xi Lin
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Qingle Liang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Lingling Ge
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Jing Xie
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Siyu Chen
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Na Liu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Juncai He
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Department of Ophthalmology, The 920 Hospital of PLA Joint Logistics Support Force, Kunming 650032, Yunnan, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Xiang Ma
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian 116014, Liaoning, China
| |
Collapse
|
26
|
Budoff SA, Poleg-Polsky A. A Complete Spatial Map of Mouse Retinal Ganglion Cells Reveals Density and Gene Expression Specializations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637538. [PMID: 39990332 PMCID: PMC11844403 DOI: 10.1101/2025.02.10.637538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Retinal ganglion cells (RGCs) transmit visual information from the eye to the brain. In mice, several RGC subtypes show nonuniform spatial distributions, potentially mediating specific visual functions. However, the full extent of RGC specialization remains unknown. Here, we used en-face cryosectioning, spatial transcriptomics, and machine learning to map the spatial distribution of all RGC subtypes identified in previous single-cell studies. While two-thirds of RGC subtypes were evenly distributed, others showed strong biases toward ventral or dorso-temporal regions associated with sky vision and the area retinae temporalis (ART), the predicted homolog of the area centralis. Additionally, we observed unexpected spatial variation in gene expression within several subtypes along the dorso-ventral axis or within vs. outside the ART, independent of RGC density profiles. Finally, we found limited correlations between the gene profiles of the ART and the primate macula, suggesting divergent specialization between the mouse and primate central vision.
Collapse
Affiliation(s)
- Samuel A. Budoff
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| | - Alon Poleg-Polsky
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| |
Collapse
|
27
|
Vidal-Vázquez N, Hernández-Núñez I, Carballo-Pacoret P, Salisbury S, Villamayor PR, Hervas-Sotomayor F, Yuan X, Lamanna F, Schneider C, Schmidt J, Mazan S, Kaessmann H, Adrio F, Robledo D, Barreiro-Iglesias A, Candal E. A single-nucleus RNA sequencing atlas of the postnatal retina of the shark Scyliorhinus canicula. Sci Data 2025; 12:228. [PMID: 39920165 PMCID: PMC11806052 DOI: 10.1038/s41597-025-04547-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
The retina, whose basic cellular structure is highly conserved across vertebrates, constitutes an accessible system for studying the central nervous system. In recent years, single-cell RNA sequencing studies have uncovered cellular diversity in the retina of a variety of species, providing new insights on retinal evolution and development. However, similar data in cartilaginous fishes, the sister group to all other extant jawed vertebrates, are still lacking. Here, we present a single-nucleus RNA sequencing atlas of the postnatal retina of the catshark Scyliorhinus canicula, consisting of the expression profiles for 17,438 individual cells from three female, juvenile catshark specimens. Unsupervised clustering revealed 22 distinct cell types comprising all major retinal cell classes, as well as retinal progenitor cells (whose presence reflects the persistence of proliferative activity in postnatal stages in sharks) and oligodendrocytes. Thus, our dataset serves as a foundation for further studies on the development and function of the catshark retina. Moreover, integration of our atlas with data from other species will allow for a better understanding of vertebrate retinal evolution.
Collapse
Affiliation(s)
- Nicolás Vidal-Vázquez
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Ismael Hernández-Núñez
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Pablo Carballo-Pacoret
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Sarah Salisbury
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Paula R Villamayor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, 27002, Lugo, Spain
| | - Francisca Hervas-Sotomayor
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
- INRAE, LPGP, Rennes, France
| | - Xuefei Yuan
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Francesco Lamanna
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Céline Schneider
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Julia Schmidt
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Sylvie Mazan
- CNRS, Sorbonne Université, Biologie Intégrative des Organismes Marins, UMR7232-BIOM, Banyuls-sur-Mer, France
| | - Henrik Kaessmann
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Fátima Adrio
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
- Departamento de Zooloxía, Xenética e Antropoloxía Física, CIBUS, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Antón Barreiro-Iglesias
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Eva Candal
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
28
|
Kinder L, Lindner M. Expression of Osteopontin in M2 and M4 Intrinsically Photosensitive Retinal Ganglion Cells in the Mouse Retina. Invest Ophthalmol Vis Sci 2025; 66:14. [PMID: 39908128 PMCID: PMC11804889 DOI: 10.1167/iovs.66.2.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/12/2025] [Indexed: 02/07/2025] Open
Abstract
Purpose Melanopsin-expressing intrinsically photosensitive (ip) retinal ganglion cells (RGCs) can be divided into six different subtypes (M1 - M6). Yet, specific markers exist for only some of these subtypes that could be employed to study the function of individual subtypes. Osteopontin (Spp1) marks αRGC, suggesting that, across ipRGCs, it would only mark the M4-ipRGC subtype (synonymous to ON-sustained αRGCs). Recent evidence suggests that osteopontin expression could spread to other ipRGC subtypes. Therefore, this study aims to characterize the expression pattern of osteopontin across ipRGC subtypes in mice. Methods Single-cell RNA (scRNA-seq) sequencing data from murine RGCs were analyzed to identify expression patterns of Spp1 across ipRGCs. Immunohistochemistry (IHC) was performed on retinal cryosections and flatmounts from C57BL/6J mice to characterize the localization of osteopontin across ipRGCs. Neurite tracing was employed to study dendritic morphology and identify individual ipRGC subtypes. Results scRNA-seq analysis revealed Spp1 expression in two distinct clusters of ipRGCs. IHC confirmed osteopontin colocalization with neurofilament heavy chain, an established marker for αRGCs, including M4-ipRGCs. Spp1 immunoreactivity was moreover identified in one additional group of ipRGCs. By dendritic morphology and stratification, those cells were clearly identified as M2-ipRGCs. Conclusions Our findings demonstrate that osteopontin is expressed in both M2- and M4-ipRGCs, challenging the notion of osteopontin as a marker exclusively for αRGCs. IHC double-labeling for osteopontin and melanopsin provides a novel method to identify and differentiate M2 ipRGCs from other subtypes. This will support the study of ipRGC physiology in a subtype -specific manner and may, for instance, foster research in the field of optic nerve injury.
Collapse
Affiliation(s)
- Leonie Kinder
- Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps-University, Marburg, Germany
| | - Moritz Lindner
- Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps-University, Marburg, Germany
- The Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Department of Ophthalmology, Philipps-University, University Hospital of Giessen and Marburg GmbH, Marburg Campus, Marburg, Germany
| |
Collapse
|
29
|
Lee PY, Greferath U, Zhao D, Huang JY, Wang AYM, Vessey KA, Chrysostomou V, Fletcher EL, Crowston JG, Bui BV. Systemic TRPV4 inhibition worsens retinal response to acute intraocular pressure elevation in older but not younger mice. Optom Vis Sci 2025; 102:78-89. [PMID: 39882862 DOI: 10.1097/opx.0000000000002217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
SIGNIFICANCE Previous evidence showed that transient receptor potential vanilloid 4 (TRPV4) inhibition was protective of retinal ganglion cell (RGC) loss after chronic intraocular pressure (IOP) elevation in young animals. However, the role of TRPV4 in mechanosensing IOP changes in the aging eye is not well understood. PURPOSE This study compared the recovery of retinal function and structure after acute IOP elevation in 3- and 12-month-old mouse eyes with and without TRPV4 inhibition. METHODS We examined retinal TRPV4 expression in 2-month-old rodent eyes using immunohistochemistry and transcript analysis of isolated macroglia and RGCs. To modulate TRPV4, mice were treated daily with either vehicle or a TRPV4 antagonist (HC-067047 10 mg/kg) delivered intraperitoneally for 7 days before and 7 days after IOP elevation (50 mmHg for 30 minutes). Retinal function and structure were assessed using dark-adapted full-field electroretinography and optical coherence tomography, respectively. RESULTS We showed that Müller cells strongly expressed TRPV4. Seven days after IOP elevation, RGC functional recovery was significantly poorer in older mice treated with TRPV4 antagonist compared with age-matched vehicle controls (-54 ± 7% vs. -24 ± 10%, p=0.046) and their younger TRPV4 antagonist-treated counterparts (-5 ± 5%, p<0.001). CONCLUSIONS This study showed that there was an age-related deficit in RGC functional recovery from IOP elevation with TRPV4 inhibition.
Collapse
Affiliation(s)
- Pei Ying Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Da Zhao
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jin Y Huang
- School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Anna Y M Wang
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Vicki Chrysostomou
- Singapore Eye Research Institute, Duke-NUS Medical School, Singapore, Singapore
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
30
|
Roy S. Emerging strategies targeting genes and cells in glaucoma. Vision Res 2025; 227:108533. [PMID: 39644708 PMCID: PMC11788065 DOI: 10.1016/j.visres.2024.108533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Glaucoma comprises a heterogeneous set of eye conditions that cause progressive vision loss. Glaucoma has a complex etiology, with different genetic and non-genetic risk factors that differ across populations. Although difficult to diagnose in early stages, compromised cellular signaling, dysregulation of genes, and homeostatic imbalance are common precursors to injury and subsequent death of retinal ganglion cells (RGCs). Lowering intraocular pressure (IOP) remains the primary approach for managing glaucoma but IOP alone does not explain all glaucoma risks. Orthogonal approaches such as large-scale genetic screening, combined with studies of animal models have been instrumental in identifying genes and molecular pathways involved in glaucoma pathogenesis. Cell type dependent vulnerability among RGCs can reveal genetic basis for specific visual deficits. A growing body of knowledge and availability of modern tools to perform targeted assessments of cellular health in different animal models facilitate development of effective and timely interventions for vision rescue. This review highlights recent findings on genes, molecules, and cell types in the context of glaucoma pathophysiology and treatment.
Collapse
Affiliation(s)
- Suva Roy
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
31
|
Rudy MA, Watkins TA. A lead role for a "secondary" axonal injury response. Neural Regen Res 2025; 20:469-470. [PMID: 38819051 PMCID: PMC11317944 DOI: 10.4103/nrr.nrr-d-23-02070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 06/01/2024] Open
Affiliation(s)
- Melissa A. Rudy
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Trent A. Watkins
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
32
|
Yang X, Zhou B. Unleashing metabolic power for axonal regeneration. Trends Endocrinol Metab 2025; 36:161-175. [PMID: 39069446 DOI: 10.1016/j.tem.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024]
Abstract
Axon regeneration requires the mobilization of intracellular resources, including proteins, lipids, and nucleotides. After injury, neurons need to adapt their metabolism to meet the biosynthetic demands needed to achieve axonal regeneration. However, the exact contribution of cellular metabolism to this process remains elusive. Insights into the metabolic characteristics of proliferative cells may illuminate similar mechanisms operating in axon regeneration; therefore, unraveling previously unappreciated roles of metabolic adaptation is critical to achieving neuron regrowth, which is connected to the therapeutic strategies for neurological conditions necessitating nerve repairs, such as spinal cord injury and stroke. Here, we outline the metabolic role in axon regeneration and discuss factors enhancing nerve regrowth, highlighting potential novel metabolic treatments for restoring nerve function.
Collapse
Affiliation(s)
- Xiaoyan Yang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China.
| |
Collapse
|
33
|
El-Danaf RN, Kapuralin K, Rajesh R, Simon F, Drou N, Pinto-Teixeira F, Özel MN, Desplan C. Morphological and functional convergence of visual projection neurons from diverse neurogenic origins in Drosophila. Nat Commun 2025; 16:698. [PMID: 39814708 PMCID: PMC11735856 DOI: 10.1038/s41467-025-56059-7] [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] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
The Drosophila visual system is a powerful model to study the development of neural circuits. Lobula columnar neurons-LCNs are visual output neurons that encode visual features relevant to natural behavior. There are ~20 classes of LCNs forming non-overlapping synaptic optic glomeruli in the brain. To address their origin, we used single-cell mRNA sequencing to define the transcriptome of LCN subtypes and identified lines that are expressed throughout their development. We show that LCNs originate from stem cells in four distinct brain regions exhibiting different modes of neurogenesis, including the ventral and dorsal tips of the outer proliferation center, the ventral superficial inner proliferation center and the central brain. We show that this convergence of similar neurons illustrates the complexity of generating neuronal diversity, and likely reflects the evolutionary origin of each subtype that detects a specific visual feature and might influence behaviors specific to each species.
Collapse
Affiliation(s)
- Rana Naja El-Danaf
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE.
| | - Katarina Kapuralin
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE
- Faculty of Biotechnology and Drug Development, University of Rijeka, Rijeka, Croatia
| | - Raghuvanshi Rajesh
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE
- Department of Biology, New York University, 10 Washington Place, New York, NY, 10003, USA
| | - Félix Simon
- Department of Biology, New York University, 10 Washington Place, New York, NY, 10003, USA
| | - Nizar Drou
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE
| | - Filipe Pinto-Teixeira
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, UPS, CNRS, Toulouse, France
| | - Mehmet Neset Özel
- Department of Biology, New York University, 10 Washington Place, New York, NY, 10003, USA
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Claude Desplan
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE.
- Department of Biology, New York University, 10 Washington Place, New York, NY, 10003, USA.
| |
Collapse
|
34
|
Yang T, Zhang N, Yang N. Single-cell sequencing in diabetic retinopathy: progress and prospects. J Transl Med 2025; 23:49. [PMID: 39806376 PMCID: PMC11727737 DOI: 10.1186/s12967-024-06066-x] [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: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic retinopathy is a major ocular complication of diabetes, characterized by progressive retinal microvascular damage and significant visual impairment in working-age adults. Traditional bulk RNA sequencing offers overall gene expression profiles but does not account for cellular heterogeneity. Single-cell RNA sequencing overcomes this limitation by providing transcriptomic data at the individual cell level and distinguishing novel cell subtypes, developmental trajectories, and intercellular communications. Researchers can use single-cell sequencing to draw retinal cell atlases and identify the transcriptomic features of retinal cells, enhancing our understanding of the pathogenesis and pathological changes in diabetic retinopathy. Additionally, single-cell sequencing is widely employed to analyze retinal organoids and single extracellular vesicles. Single-cell multi-omics sequencing integrates omics information, whereas stereo-sequencing analyzes gene expression and spatiotemporal data simultaneously. This review discusses the protocols of single-cell sequencing for obtaining single cells from retina and accurate sequencing data. It highlights the applications and advancements of single-cell sequencing in the study of normal retinas and the pathological changes associated with diabetic retinopathy. This underscores the potential of these technologies to deepen our understanding of the pathogenesis of diabetic retinopathy that may lead to the introduction of new therapeutic strategies.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China.
| |
Collapse
|
35
|
Gonschorek D, Goldin MA, Oesterle J, Schwerd-Kleine T, Arlinghaus R, Zhao Z, Schubert T, Marre O, Euler T. Nitric oxide modulates contrast suppression in a subset of mouse retinal ganglion cells. eLife 2025; 13:RP98742. [PMID: 39783858 PMCID: PMC11717361 DOI: 10.7554/elife.98742] [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] [Indexed: 01/12/2025] Open
Abstract
Neuromodulators have major influences on the regulation of neural circuit activity across the nervous system. Nitric oxide (NO) has been shown to be a prominent neuromodulator in many circuits and has been extensively studied in the retina. Here, it has been associated with the regulation of light adaptation, gain control, and gap junctional coupling, but its effect on the retinal output, specifically on the different types of retinal ganglion cells (RGCs), is still poorly understood. In this study, we used two-photon Ca2+ imaging and multi-electrode array (MEA) recordings to measure light-evoked activity of RGCs in the ganglion cell layer in the ex vivo mouse retina. This approach allowed us to investigate the neuromodulatory effects of NO on a cell type-level. Our findings reveal that NO selectively modulates the suppression of temporal responses in a distinct subset of contrast-suppressed RGC types, increasing their activity without altering the spatial properties of their receptive fields. Given that under photopic conditions, NO release is triggered by quick changes in light levels, we propose that these RGC types signal fast contrast changes to higher visual regions. Remarkably, we found that about one-third of the RGC types, recorded using two-photon Ca2+ imaging, exhibited consistent, cell type-specific adaptational response changes throughout an experiment, independent of NO. By employing a sequential-recording paradigm, we could disentangle those additional adaptational response changes from drug-induced modulations. Taken together, our research highlights the selective neuromodulatory effects of NO on RGCs and emphasizes the need of considering non-pharmacological activity changes, like adaptation, in such study designs.
Collapse
Affiliation(s)
- Dominic Gonschorek
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Matías A Goldin
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Jonathan Oesterle
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Hertie Institute for AI in Brain Health, Tübingen AI Center, University of TübingenTübingenGermany
| | - Tom Schwerd-Kleine
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Ryan Arlinghaus
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Zhijian Zhao
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Timm Schubert
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Olivier Marre
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Thomas Euler
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
- Bernstein Center for Computational Neuroscience, University of TübingenTübingenGermany
| |
Collapse
|
36
|
Liu ZG, Zhou LY, Sun YQ, Ma YH, Liu CM, Zhang BY. Unlocking the potential for optic nerve regeneration over long distances: a multi-therapeutic intervention. Front Neurol 2025; 15:1526973. [PMID: 39850731 PMCID: PMC11754882 DOI: 10.3389/fneur.2024.1526973] [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: 11/15/2024] [Accepted: 12/24/2024] [Indexed: 01/25/2025] Open
Abstract
Retinal ganglion cells (RGCs) generally fail to regenerate axons, resulting in irreversible vision loss after optic nerve injury. While many studies have shown that modulating specific genes can enhance RGCs survival and promote optic nerve regeneration, inducing long-distance axon regeneration in vivo through single-gene manipulation remains challenging. Nevertheless, combined multi-gene therapies have proven effective in significantly enhancing axonal regeneration. At present, research on promoting optic nerve regeneration remains slow, with most studies unable to achieve axonal growth beyond the optic chiasm or reestablish connections with the brain. Future research priorities include directing axonal growth along correct pathways, facilitating synapse formation and myelination, and modifying the inhibitory microenvironment. These strategies are crucial not only for optic nerve regeneration but also for broader applications in central nervous system repair. In this review, we discuss multifactors therapeutic strategies for optic nerve regeneration, offering insights into advancing nerve regeneration research.
Collapse
Affiliation(s)
- Zhen-Gang Liu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lai-Yang Zhou
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yong-Quan Sun
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yi-Hang Ma
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang-Mei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Bo-Yin Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Hernández-Núñez I, Clark BS. Experimental Framework for Assessing Mouse Retinal Regeneration Through Single-Cell RNA-Sequencing. Methods Mol Biol 2025; 2848:117-134. [PMID: 39240520 DOI: 10.1007/978-1-0716-4087-6_8] [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] [Indexed: 09/07/2024]
Abstract
Retinal degenerative diseases including age-related macular degeneration and glaucoma are estimated to currently affect more than 14 million people in the United States, with an increased prevalence of retinal degenerations in aged individuals. An expanding aged population who are living longer forecasts an increased prevalence and economic burden of visual impairments. Improvements to visual health and treatment paradigms for progressive retinal degenerations slow vision loss. However, current treatments fail to remedy the root cause of visual impairments caused by retinal degenerations-loss of retinal neurons. Stimulation of retinal regeneration from endogenous cellular sources presents an exciting treatment avenue for replacement of lost retinal cells. In multiple species including zebrafish and Xenopus, Müller glial cells maintain a highly efficient regenerative ability to reconstitute lost cells throughout the organism's lifespan, highlighting potential therapeutic avenues for stimulation of retinal regeneration in humans. Here, we describe how the application of single-cell RNA-sequencing (scRNA-seq) has enhanced our understanding of Müller glial cell-derived retinal regeneration, including the characterization of gene regulatory networks that facilitate/inhibit regenerative responses. Additionally, we provide a validated experimental framework for cellular preparation of mouse retinal cells as input into scRNA-seq experiments, including insights into experimental design and analyses of resulting data.
Collapse
Affiliation(s)
- Ismael Hernández-Núñez
- John F Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Clark
- John F Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
38
|
Zhang Q, Tang J, Liu L, Liu Z, Xue J, Ge J, Zhuo Y, Li Y. Emerging therapeutic strategies for optic nerve regeneration. Trends Pharmacol Sci 2025; 46:45-61. [PMID: 39694789 DOI: 10.1016/j.tips.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
The optic nerve, comprising axons from retinal ganglion cells (RGCs), is a component of the central nervous system (CNS) that generally exhibits a limited regeneration capacity following injury in mature mammals, resulting in permanent vision loss. Here, we summarize recent advances in interventions targeting cell-intrinsic and cell-extrinsic mechanisms to enhance RGC axon regeneration. Additionally, we summarize strategies for guiding the reconnection of regenerating axons with brain visual targets, aiming to restore partial visual function. Given the advent of high-throughput screening techniques and multiomics analyses, we discuss how these emerging methodologies deepen our understanding of regenerative mechanisms and expedite the development of innovative therapeutic approaches. Lastly, we explore the translational potential of these strategies in achieving clinically meaningful vision recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Liyan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhe Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Jingfei Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China; Department of Ophthalmology, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
39
|
Sripinun P, Lu W, Nikonov S, Patel S, Hennessy S, Yao T, Cui QN, Bell BA, Mitchell CH. Fluorescent identification of axons, dendrites and soma of neuronal retinal ganglion cells with a genetic marker as a tool for facilitating the study of neurodegeneration. FASEB Bioadv 2025; 7:e1478. [PMID: 39781424 PMCID: PMC11705399 DOI: 10.1096/fba.2024-00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 01/12/2025] Open
Abstract
This study characterizes a fluorescent Slc17a6-tdTomato neuronal reporter mouse line with strong labeling of axons throughout the optic nerve, of retinal ganglion cell (RGC) soma in the ganglion cell layer (GCL), and of RGC dendrites in the inner plexiform layer (IPL). The model facilitated assessment of RGC loss in models of degeneration and of RGC detection in mixed neural/glial cultures. The tdTomato signal showed strong overlap with >98% cells immunolabeled with RGC markers RBPMS or BRN3A, consistent with the ubiquitous presence of the vesicular glutamate transporter 2 (VGUT2, SLC17A6) in all RGC subtypes. There was no cross-labeling of ChAT-positive displaced amacrine cells in the GCL, although some signal emanated from the outer plexiform layer, consistent with horizontal cells. The fluorescence allowed rapid screening of RGC loss following optic nerve crush and intraocular pressure (IOP) elevation. The bright fluorescence also enabled non-invasive monitoring of extensive neurite networks and neuron/astrocyte interactions in culture. Robust Ca2+ responses to P2X7R agonist BzATP were detected from fluorescent RGCs using Ca2+-indicator Fura-2. Fluorescence from axons and soma was detected in vivo with a confocal scanning laser ophthalmoscope (cSLO); automatic RGC soma counts enhanced through machine learning approached the numbers found in retinal wholemounts. Controls indicated no impact of Slc17a6-tdTomato expression on light-dependent neuronal function as measured with a microelectrode array (MEA), or on retinal structure as measured with optical coherence tomography (OCT). In summary, the bright fluorescence in axons, dendrites and soma of ~all RGCs in the Slc17a6-tdTomato reporter mouse may facilitate the study of RGCs.
Collapse
Affiliation(s)
- Puttipong Sripinun
- Department of Basic and Translational ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of OrthodonticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Orthodontics and Pediatric DentistryChiang Mai UniversityChiang MaiThailand
| | - Wennan Lu
- Department of Basic and Translational ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sergei Nikonov
- Department of NeuroscienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Suhani Patel
- Department of Basic and Translational ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sarah Hennessy
- Department of Basic and Translational ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Tianyuan Yao
- Department of OphthalmologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Qi N. Cui
- Department of OphthalmologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Brent A. Bell
- Department of OphthalmologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Claire H. Mitchell
- Department of Basic and Translational ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of PhysiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
40
|
Wang J, Zhang L, Cavallini M, Pahlevan A, Sun J, Morshedian A, Fain GL, Sampath AP, Peng YR. Molecular characterization of the sea lamprey retina illuminates the evolutionary origin of retinal cell types. Nat Commun 2024; 15:10761. [PMID: 39737973 PMCID: PMC11685597 DOI: 10.1038/s41467-024-55019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
The lamprey, a primitive jawless vertebrate whose ancestors diverged from all other vertebrates over 500 million years ago, offers a unique window into the ancient formation of the retina. Using single-cell RNA-sequencing, we characterize retinal cell types in the lamprey and compare them to those in mouse, chicken, and zebrafish. We find six cell classes and 74 distinct cell types, many shared with other vertebrate species. The conservation of cell types indicates their emergence early in vertebrate evolution, highlighting primordial designs of retinal circuits for the rod pathway, ON-OFF discrimination, and direction selectivity. The diversification of amacrine and some ganglion cell types appears, however, to be distinct in the lamprey. We further infer genetic regulators in specifying retinal cell classes and identify ancestral regulatory elements across species, noting decreased conservation in specifying amacrine cells. Altogether, our characterization of the lamprey retina illuminates the evolutionary origin of visual processing in the retina.
Collapse
Affiliation(s)
- Junqiang Wang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lin Zhang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Martina Cavallini
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ali Pahlevan
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Junwei Sun
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ala Morshedian
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Gordon L Fain
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Yi-Rong Peng
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Rao M, Luo Z, Liu CC, Chen CY, Wang S, Nahmou M, Tanasa B, Virmani A, Byrne L, Goldberg JL, Sahel JA, Chang KC. Tppp3 is a novel molecule for retinal ganglion cell identification and optic nerve regeneration. Acta Neuropathol Commun 2024; 12:204. [PMID: 39734233 PMCID: PMC11684310 DOI: 10.1186/s40478-024-01917-6] [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: 08/08/2024] [Accepted: 12/14/2024] [Indexed: 12/31/2024] Open
Abstract
Mammalian central nervous system (CNS) axons cannot spontaneously regenerate after injury, creating an unmet need to identify molecular regulators to promote axon regeneration and reduce the lasting impact of CNS injuries. While tubulin polymerization promoting protein family member 3 (Tppp3) is known to promote axon outgrowth in amphibians, its role in mammalian axon regeneration remains unknown. Here we investigated Tppp3 in retinal ganglion cells (RGCs) neuroprotection and axonal regeneration using an optic nerve crush (ONC) model in the rodent. Single-cell RNA sequencing identified the expression of Tppp3 in RGCs of mice, macaques, and humans. Tppp3 overexpression enhanced neurite outgrowth in mouse primary RGCs in vitro, promoted axon regeneration, and improved RGC survival after ONC. Bulk RNA sequencing indicated that Tppp3 overexpression upregulates axon regeneration genes such as Bmp4 and neuroinflammatory pathways. Our findings advance regenerative medicine by developing a new therapeutic strategy for RGC neuroprotection and axon regeneration.
Collapse
Affiliation(s)
- Mishal Rao
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA, 94305, USA
| | - Chia-Chun Liu
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
| | - Chi-Yu Chen
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
| | - Shining Wang
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
| | - Michael Nahmou
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA, 94305, USA
| | - Bogdan Tanasa
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA, 94305, USA
| | - Aman Virmani
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
| | - Leah Byrne
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA, 94305, USA
| | - José-Alain Sahel
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA
| | - Kun-Che Chang
- Department of Ophthalmology, UPMC Vision Institute, University of Pittsburgh School of Medicine, 1622 Locust Street, Pittsburgh, PA, 15219, USA.
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA, 94305, USA.
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
42
|
Peng XQ, Li YZ, Gu C, He XC, Li CP, Sun YQ, Du HZ, Teng ZQ, Liu CM. Marcks overexpression in retinal ganglion cells promotes optic nerve regeneration. Cell Death Dis 2024; 15:906. [PMID: 39695101 DOI: 10.1038/s41419-024-07281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
Regeneration of injured central nervous system (CNS) axons is highly restricted, leading to permanent neurological deficits. The myristoylated alanine-rich C-kinase substrate (MARCKS) is a membrane-associated protein kinase C (PKC) substrate ubiquitously expressed in eukaryotic cells, plays critical roles in development, brain plasticity, and tissues regeneration. However, little is known about the role of Marcks in CNS axon regeneration. Here we show that Marcks overexpression promotes robust axon regeneration either before or after optic nerve crush, but insignificantly impacts neuronal survival. Notably, immunostaining and RNA sequencing demonstrate that Marcks overexpression does not affect known regeneration-associated genes and pathways. Furthermore, combining CNTF which activates the JAK-STAT3 pathway and Marcks overexpression further enhances axon regeneration. Finally, we demonstrate functionally essential effector domain (ED) of MARCKS has similar effects on inducing axon regeneration in RGCs. These results suggest that manipulating Marcks and its ED may become a therapeutic approach to promote axon regeneration after CNS injury.
Collapse
Affiliation(s)
- Xue-Qi Peng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yan-Zhong Li
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Chen Gu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xuan-Cheng He
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Chang-Ping Li
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yong-Quan Sun
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hong-Zhen Du
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Zhao-Qian Teng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Chang-Mei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| |
Collapse
|
43
|
Tsai NY, Nimkar K, Zhao M, Lum MR, Yi Y, Garrett TR, Wang Y, Toma K, Caval-Holme F, Reddy N, Ehrlich AT, Kriegstein AR, Do MTH, Sivyer B, Shekhar K, Duan X. Molecular and spatial analysis of ganglion cells on retinal flatmounts: diversity, topography, and perivascularity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.15.628587. [PMID: 39763751 PMCID: PMC11702564 DOI: 10.1101/2024.12.15.628587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Diverse retinal ganglion cells (RGCs) transmit distinct visual features from the eye to the brain. Recent studies have categorized RGCs into 45 types in mice based on transcriptomic profiles, showing strong alignment with morphological and electrophysiological properties. However, little is known about how these types are spatially arranged on the two-dimensional retinal surface-an organization that influences visual encoding-and how their local microenvironments impact development and neurodegenerative responses. To address this gap, we optimized a workflow combining imaging-based spatial transcriptomics (MERFISH) and immunohistochemical co-staining on thin flatmount retinal sections. We used computational methods to register en face somata distributions of all molecularly defined RGC types. More than 75% (34/45) of types exhibited non-uniform distributions, likely reflecting adaptations of the retina's anatomy to the animal's visual environment. By analyzing the local neighborhoods of each cell, we identified perivascular RGCs located near blood vessels. Seven RGC types are enriched in the perivascular niche, including members of intrinsically photosensitive RGC (ipRGC) and direction-selective RGC (DSGC) subclasses. Orthologous human RGC counterparts of perivascular types - Melanopsin-enriched ipRGCs and ON DSGCs - were also proximal to blood vessels, suggesting their perivascularity may be evolutionarily conserved. Following optic nerve crush in mice, the perivascular M1-ipRGCs and ON DSGCs showed preferential survival, suggesting that proximity to blood vessels may render cell-extrinsic neuroprotection to RGCs through an mTOR-independent mechanism. Overall, our work offers a resource characterizing the spatial profiles of RGC types, enabling future studies of retinal development, physiology, and neurodegeneration at individual neuron type resolution across the two-dimensional space.
Collapse
Affiliation(s)
- Nicole Y Tsai
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Kushal Nimkar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- These authors contributed equally
| | - Mengya Zhao
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Matthew R Lum
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Yujuan Yi
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Tavita R Garrett
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Yixiao Wang
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Kenichi Toma
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Franklin Caval-Holme
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School. Boston, MA, USA
| | - Nikhil Reddy
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Aliza T Ehrlich
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Arnold R Kriegstein
- Department of Neurology and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Michael Tri H Do
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School. Boston, MA, USA
| | - Benjamin Sivyer
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; Center for Computational Biology; Biophysics Graduate Group, University of California, Berkeley, CA, USA
- These authors contributed equally
| | - Xin Duan
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
- Department of Physiology and Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
- Lead contact
| |
Collapse
|
44
|
Delpech C, Schaeffer J, Vilallongue N, Delaunay A, Benadjal A, Blot B, Excoffier B, Plissonnier E, Gascon E, Albert F, Paccard A, Saintpierre A, Gasnier C, Zagar Y, Castellani V, Belin S, Chédotal A, Nawabi H. Axon guidance during mouse central nervous system regeneration is required for specific brain innervation. Dev Cell 2024; 59:3213-3228.e8. [PMID: 39353435 DOI: 10.1016/j.devcel.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 07/11/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Reconstructing functional neuronal circuits is one major challenge of central nervous system repair. Through activation of pro-growth signaling pathways, some neurons achieve long-distance axon regrowth. Yet, functional reconnection has hardly been obtained, as these regenerating axons fail to resume their initial trajectory and reinnervate their proper target. Axon guidance is considered to be active only during development. Here, using the mouse visual system, we show that axon guidance is still active in the adult brain in regenerative conditions. We highlight that regenerating retinal ganglion cell axons avoid one of their primary targets, the suprachiasmatic nucleus (SCN), due to Slit/Robo repulsive signaling. Together with promoting regeneration, silencing Slit/Robo in vivo enables regenerating axons to enter the SCN and form active synapses. The newly formed circuit is associated with neuronal activation and functional recovery. Our results provide evidence that axon guidance mechanisms are required to reconnect regenerating axons to specific brain nuclei.
Collapse
Affiliation(s)
- Céline Delpech
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julia Schaeffer
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Noemie Vilallongue
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Apolline Delaunay
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Amin Benadjal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Beatrice Blot
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Blandine Excoffier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Elise Plissonnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eduardo Gascon
- Aix Marseille University, CNRS, INT, Institute of Neurosci Timone, Marseille, France
| | - Floriane Albert
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Antoine Paccard
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Ana Saintpierre
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Celestin Gasnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Valérie Castellani
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | - Stephane Belin
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France; Institut de pathologie, groupe hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Homaira Nawabi
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
45
|
Schaeffer J, Belin S. Axon regeneration: an issue of translation. C R Biol 2024; 347:249-258. [PMID: 39665232 DOI: 10.5802/crbiol.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024]
Abstract
In the mammalian central nervous system (CNS), adult neurons fail to regenerate spontaneously upon axon injury, which leads to a permanent and irreversible loss of neuronal functions. For more than 15 years, much effort was invested to unlock axon regrowth programs based on extensive transcriptomic characterization. However, it is now well described that mRNA and protein levels correlate only partially in cells, and that the transcription process (from DNA to mRNA) may not directly reflect protein expression. Conversely, the translation process (from mRNA to protein) provides an additional layer of gene regulation. This aspect has been overlooked in CNS regeneration. In this review, we discuss the limitations of transcriptomic approaches to promote CNS regeneration and we provide the rationale to investigate translational regulation in this context, and notably the regulatory role of the translational complex. Finally, we summarize our and others’ recent findings showing how variations in the translational complex composition regulate selective (mRNA-specific) translation, thereby controlling CNS axon regrowth.
Collapse
|
46
|
Kwon HJ, Santhosh D, Huang Z. A novel monomeric amyloid β-activated signaling pathway regulates brain development via inhibition of microglia. eLife 2024; 13:RP100446. [PMID: 39635981 PMCID: PMC11620749 DOI: 10.7554/elife.100446] [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] [Indexed: 12/07/2024] Open
Abstract
Amyloid β (Aβ) forms aggregates in the Alzheimer's disease brain and is well known for its pathological roles. Recent studies show that it also regulates neuronal physiology in the healthy brain. Whether Aβ also regulates glial physiology in the normal brain, however, has remained unclear. In this article, we describe the discovery of a novel signaling pathway activated by the monomeric form of Aβ in vitro that plays essential roles in the regulation of microglial activity and the assembly of neocortex during mouse development in vivo. We find that activation of this pathway depends on the function of amyloid precursor and the heterotrimeric G protein regulator Ric8a in microglia and inhibits microglial immune activation at transcriptional and post-transcriptional levels. Genetic disruption of this pathway during neocortical development results in microglial dysregulation and excessive matrix proteinase activation, leading to basement membrane degradation, neuronal ectopia, and laminar disruption. These results uncover a previously unknown function of Aβ as a negative regulator of brain microglia and substantially elucidate the underlying molecular mechanisms. Considering the prominence of Aβ and neuroinflammation in the pathology of Alzheimer's disease, they also highlight a potentially overlooked role of Aβ monomer depletion in the development of the disease.
Collapse
Affiliation(s)
- Hyo Jun Kwon
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Devi Santhosh
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Zhen Huang
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
47
|
Masin L, Bergmans S, Van Dyck A, Farrow K, De Groef L, Moons L. Local glycolysis supports injury-induced axonal regeneration. J Cell Biol 2024; 223:e202402133. [PMID: 39352499 PMCID: PMC11451009 DOI: 10.1083/jcb.202402133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/09/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
Successful axonal regeneration following injury requires the effective allocation of energy. How axons withstand the initial disruption in mitochondrial energy production caused by the injury and subsequently initiate regrowth is poorly understood. Transcriptomic data showed increased expression of glycolytic genes after optic nerve crush in retinal ganglion cells with the co-deletion of Pten and Socs3. Using retinal cultures in a multicompartment microfluidic device, we observed increased regrowth and enhanced mitochondrial trafficking in the axons of Pten and Socs3 co-deleted neurons. While wild-type axons relied on mitochondrial metabolism, after injury, in the absence of Pten and Socs3, energy production was supported by local glycolysis. Specific inhibition of lactate production hindered injury survival and the initiation of regrowth while slowing down glycolysis upstream impaired regrowth initiation, axonal elongation, and energy production. Together, these observations reveal that glycolytic ATP, combined with sustained mitochondrial transport, is essential for injury-induced axonal regrowth, providing new insights into the metabolic underpinnings of axonal regeneration.
Collapse
Affiliation(s)
- Luca Masin
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Steven Bergmans
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Annelies Van Dyck
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Karl Farrow
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Neuro-Electronics Research Flanders, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- imec, Leuven, Belgium
| | - Lies De Groef
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Lieve Moons
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
48
|
Latini L, De Araujo DSM, Amato R, Canovai A, Buccarello L, De Logu F, Novelli E, Vlasiuk A, Malerba F, Arisi I, Florio R, Asari H, Capsoni S, Strettoi E, Villetti G, Imbimbo BP, Dal Monte M, Nassini R, Geppetti P, Marinelli S, Cattaneo A. A p75 neurotrophin receptor-sparing nerve growth factor protects retinal ganglion cells from neurodegeneration by targeting microglia. Br J Pharmacol 2024; 181:4890-4919. [PMID: 39252503 DOI: 10.1111/bph.17316] [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: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Retinal ganglion cells (RGCs) are the output stage of retinal information processing, via their axons forming the optic nerve (ON). ON damage leads to axonal degeneration and death of RGCs, and results in vision impairment. Nerve growth factor (NGF) signalling is crucial for RGC operations and visual functions. Here, we investigate a new neuroprotective mechanism of a novel therapeutic candidate, a p75-less, TrkA-biased NGF agonist (hNGFp) in rat RGC degeneration, in comparison with wild type human NGF (hNGFwt). EXPERIMENTAL APPROACH Both neonate and adult rats, whether subjected or not to ON lesion, were treated with intravitreal injections or eye drops containing either hNGFp or hNGFwt. Different doses of the drugs were administered at days 1, 4 or 7 after injury for a maximum of 10 days, when immunofluorescence, electrophysiology, cellular morphology, cytokine array and behaviour studies were carried out. Pharmacokinetic evaluation was performed on rabbits treated with hNGFp ocular drops. RESULTS hNGFp exerted a potent RGC neuroprotection by acting on microglia cells, and outperformed hNGFwt in rescuing RGC degeneration and reducing inflammatory molecules. Delayed use of hNGFp after ON lesion resulted in better outcomes compared with treatment with hNGFwt. Moreover, hNGFp-based ocular drops were less algogenic than hNGFwt. Pharmacokinetic measurements revealed that biologically relevant quantities of hNGFp were found in the rabbit retina. CONCLUSIONS AND IMPLICATIONS Our data point to microglia as a new cell target through which NGF-induced TrkA signalling exerts neuroprotection of the RGC, emphasizing hNGFp as a powerful treatment to tackle retinal degeneration.
Collapse
Affiliation(s)
- Laura Latini
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | | | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Elena Novelli
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Anastasiia Vlasiuk
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rita Florio
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Hiroki Asari
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Simona Capsoni
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Enrica Strettoi
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Gino Villetti
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
49
|
Huang KC, Tawfik M, Samuel MA. Retinal ganglion cell circuits and glial interactions in humans and mice. Trends Neurosci 2024; 47:994-1013. [PMID: 39455342 PMCID: PMC11631666 DOI: 10.1016/j.tins.2024.09.010] [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: 05/17/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
Retinal ganglion cells (RGCs) are the brain's gateway for vision, and their degeneration underlies several blinding diseases. RGCs interact with other neuronal cell types, microglia, and astrocytes in the retina and in the brain. Much knowledge has been gained about RGCs and glia from mice and other model organisms, often with the assumption that certain aspects of their biology may be conserved in humans. However, RGCs vary considerably between species, which could affect how they interact with their neuronal and glial partners. This review details which RGC and glial features are conserved between mice, humans, and primates, and which differ. We also discuss experimental approaches for studying human and primate RGCs. These strategies will help to bridge the gap between rodent and human RGC studies and increase study translatability to guide future therapeutic strategies.
Collapse
Affiliation(s)
- Kang-Chieh Huang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030. USA.
| | - Mohamed Tawfik
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030. USA
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030. USA.
| |
Collapse
|
50
|
Kosior-Jarecka E, Grzybowski A. Retinal Ganglion Cell Replacement in Glaucoma Therapy: A Narrative Review. J Clin Med 2024; 13:7204. [PMID: 39685661 DOI: 10.3390/jcm13237204] [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: 06/11/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide. It leads to the progressive degeneration of retinal ganglion cells (RGCs), the axons of which form the optic nerve. Enormous RGC apoptosis causes a lack of transfer of visual information to the brain. The RGC loss typical of the central nervous system is irreversible, and when glaucoma progresses, the total amount of RGCs in the retina enormously diminishes. The successful treatment in glaucoma patients is a direct neuroprotection by decreasing the intraocular pressure, which enables RGC protection but does not revive the lost ones. The intriguing new therapy for advanced glaucoma is the possibility of RGC replacement with new healthy cells. In this review article, the strategies regarding RGC replacement therapy are presented with the latest advances in the technique and the obstacles that it meets.
Collapse
Affiliation(s)
- Ewa Kosior-Jarecka
- Department of Diagnostics and Microsurgery of Glaucoma, Medical University of Lublin, 20-079 Lublin, Poland
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, 60-836 Poznan, Poland
| |
Collapse
|