Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (366)

Search Parameters:
Keywords = retina ganglion cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 10448 KiB  
Article
Ultrastructural and Immunohistochemical Study of Double and Combined Intravitreal Administration of Antifungal Agents in the Retina of New Zealand Albino Rabbits: An Experimental Protocol
by Sofia Karachrysafi, Maria Kourti, Sophia Tsokkou, Despoina Ioannou, Evangelia Kofidou, Georgios Delis, Sotiris Sotiriou, Athanasios Karamitsos, Maria Xioteli, Ioanna Dori, Penelope Anastasiadou, Ioannis Konstantinidis, Dimitrios Kavvadas, Fotios Chatzinikolaou, Anastasia Komnenou, Vasileios Karampatakis, Antonia Sioga and Theodora Papamitsou
J. Fungi 2025, 11(8), 564; https://doi.org/10.3390/jof11080564 - 29 Jul 2025
Viewed by 703
Abstract
Introduction: Fungal endophthalmitis (FE) is a rare but serious intraocular inflammatory disorder, resulting from an infection of the vitreous cavity from either endogenous or exogenous components that ultimately results in blindness. This current research study aims to elucidate the histological effects of the [...] Read more.
Introduction: Fungal endophthalmitis (FE) is a rare but serious intraocular inflammatory disorder, resulting from an infection of the vitreous cavity from either endogenous or exogenous components that ultimately results in blindness. This current research study aims to elucidate the histological effects of the intravitreal injection of the maximum safe dosage of voriconazole and micafungin on the retina and investigate potential histological alterations after the double and combined administration of voriconazole and micafungin. Methodology: Nine New Zealand Albino Rabbits were randomly assigned into three groups (V2, M2, and VM), and in each, voriconazole, micafungin, and a combination of the two medications were administered respectively. After the administration of the antifungal agents, the animals were sacrificed and their retinas were retrieved and studied under optical and electron microscopes. The immunohistochemical markers TNF-a and IL6 were also studied. Results: TNF-a was positive in the VM group, as it was found to be mildly positive in the presence of apoptotic cells in the ganglion cell layer. Conclusions: This study revealed voriconazole has a greater toxicity in a multi-dosage administration. However, micafungin revealed a greater toxicity than voriconazole from the extent of the lesions observed. Full article
Show Figures

Graphical abstract

18 pages, 6694 KiB  
Article
Effects of a ROCK Inhibitor on Retinal Ganglion Cells In Vivo and In Vitro
by Wanjing Chen, Yoko Iizuka, Fumihiko Mabuchi and Kenji Kashiwagi
J. Clin. Med. 2025, 14(15), 5344; https://doi.org/10.3390/jcm14155344 - 29 Jul 2025
Viewed by 237
Abstract
Objective: To investigate the neuroprotective effects of a Rho-associated kinase (ROCK) inhibitor on retinal ganglion cells (RGCs) in vitro and in vivo. Methods: For in vivo studies, a unilateral optic nerve crush mouse model was established. Then, 100 mM Y-27632 (a [...] Read more.
Objective: To investigate the neuroprotective effects of a Rho-associated kinase (ROCK) inhibitor on retinal ganglion cells (RGCs) in vitro and in vivo. Methods: For in vivo studies, a unilateral optic nerve crush mouse model was established. Then, 100 mM Y-27632 (a ROCK inhibitor) or saline was applied to the experimental eyes once a day for 14 days. The effects of the ROCK inhibitor were evaluated by counting the surviving RGCs in the enucleated flat retina tissues and measuring the inner retinal thickness using optical coherence tomography (OCT), the amplitude of the electroretinogram (ERG), and the change in intraocular pressure (IOP). For the in vitro study, RGCs were isolated from five-day-old mice using a modified immunopanning method with magnetic beads. The isolated RGCs were incubated for 72 h with various concentrations of Y-27632, after which TUNEL assays were performed to determine the number of surviving RGCs. Results: Y-27632 has neuroprotective effects, as it significantly increased the number of surviving RGCs by approximately 6.3%. OCT and ERG data also revealed that Y-27632 induced neuroprotective effects in vivo; furthermore, Y-27632 reduced IOP by approximately 18.3%. The in vitro study revealed the dose-dependent neuroprotective effects of Y-27632, with the highest dose of Y-27632 (1000 nM) increasing the RGC survival rate after 72 h of incubation compared with that of the control. Conclusions: The ROCK inhibitor Y-27632 may exert some neuroprotective effects on RGCs when it is used as an eye drop through an IOP-independent mechanism. Full article
(This article belongs to the Section Ophthalmology)
Show Figures

Figure 1

22 pages, 9284 KiB  
Article
Comparative Analysis of Tyrosine Hydroxylase Amacrine Cells in the Mammalian Retina: Distribution and Quantification in Mouse, Rat, Ground Squirrel and Macaque Retinas
by Kiyoharu J. Miyagishima, Xiaomin Lai, Amurta Nath, William N. Grimes, Xiyuan Ping, Jeffrey S. Diamond, Morven A. Cameron, Wei Li and Francisco M. Nadal-Nicolás
Int. J. Mol. Sci. 2025, 26(14), 6972; https://doi.org/10.3390/ijms26146972 - 20 Jul 2025
Viewed by 368
Abstract
Dopaminergic amacrine cells (DACs) are a subclass of amacrine cells that modulate retinal processing and light adaptation by releasing dopamine. Although the role of dopamine is largely conserved, their retinal distribution across mammals remains incompletely characterized. In mice, rats, thirteen-lined ground squirrels (TLGSs), [...] Read more.
Dopaminergic amacrine cells (DACs) are a subclass of amacrine cells that modulate retinal processing and light adaptation by releasing dopamine. Although the role of dopamine is largely conserved, their retinal distribution across mammals remains incompletely characterized. In mice, rats, thirteen-lined ground squirrels (TLGSs), and macaques, we systematically compared the localization, number, and topography of DACs by their expression of tyrosine hydroxylase (TH), a crucial enzyme in the biosynthesis of dopamine. In all species examined, TH+ cells were primarily located in the inner nuclear layer; however, there was a species-dependent influence on their number and distribution. Mice exhibited the highest density of TH+cells but completely lacked displaced TH+cells (dTH+cells) in the ganglion cell layer. Despite interspecies variation in the total number of TH+cells in the retina, the overall density in rats, TLGSs, and macaques was similar. Most species displayed a higher density of DACs toward central retinal regions. However, rats exhibited a distinctive dorsal concentration, particularly among dTH+cells. Although most species examined exhibited a similar ratio of TH+cells to Brn3a+ retinal ganglion cells, TLGSs showed a marked reduction, indicating a potentially diminished dopaminergic modulatory role. Species-specific DAC topographies aligned with specialized visual regions, such as the visual streak in TLGS and the macula in macaques. These results reveal both conserved and divergent features of retinal dopamine circuitry, reflecting evolutionary adaptations to visual processing demands. Full article
(This article belongs to the Section Molecular Nanoscience)
Show Figures

Figure 1

29 pages, 14985 KiB  
Article
Spatiotemporal Characterization of Changes in the Respiratory Tract and the Nervous System, Including the Eyes in SARS-CoV-2-Infected K18-hACE2 Mice
by Malgorzata Rosiak, Tom Schreiner, Georg Beythien, Eva Leitzen, Anastasiya Ulianytska, Lisa Allnoch, Kathrin Becker, Lukas M. Michaely, Sandra Lockow, Sabrina Clever, Christian Meyer zu Natrup, Asisa Volz, Wolfgang Baumgärtner, Malgorzata Ciurkiewicz, Kirsten Hülskötter and Katharina M. Gregor
Viruses 2025, 17(7), 963; https://doi.org/10.3390/v17070963 - 9 Jul 2025
Viewed by 554
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is known to affect multiple organ systems, including the respiratory tract and nervous and ocular systems. This retrospective study aimed to characterize the spatiotemporal distribution of viral antigen [...] Read more.
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is known to affect multiple organ systems, including the respiratory tract and nervous and ocular systems. This retrospective study aimed to characterize the spatiotemporal distribution of viral antigen and associated pathological changes in the nose, lungs, brain, and eyes of K18-hACE2 mice intranasally infected with SARS-CoV-2. Using histology and immunohistochemistry, tissues were examined at 3, 6, and 7/8 days post-infection (dpi). In addition, lung and brain tissues were analyzed by means of RT-qPCR to determine viral RNA titers. Viral antigen was most pronounced in the nose, brain, and lung at 3, 6, and 7/8 dpi, respectively, whereas viral antigen was detected at 6 and 7/8 dpi in the retina. Quantitative PCR confirmed increasing viral RNA levels in both lung and brain, peaking at 7/8 dpi. Nasal and lung inflammation mirrored viral antigen distribution and localization. In the brain, the predominantly basal viral spread correlated with lymphohistiocytic meningoencephalitis, neuronal vacuolation, and altered neurofilament immunoreactivity. Retinal ganglion cells showed viral antigen expression without associated lesions. Microglial activation was evident in both the optic chiasm and the brain. These findings highlight the K18-hACE2 model’s utility for studying extrapulmonary SARS-CoV-2 pathogenesis. Understanding the temporal and spatial dynamics of viral spread enhances insights into SARS-CoV-2 neurotropism and its clinical manifestations. Full article
(This article belongs to the Section Coronaviruses)
Show Figures

Graphical abstract

20 pages, 2060 KiB  
Article
Involvement of Microglia in Retinal Ganglion Cell Injury Induced by IOP Elevation in a Rat Ex Vivo Acute Glaucoma Model
by Taimu Sato, Makoto Ishikawa, Yukitoshi Izumi, Naoya Shibata, Kota Sato, Michiko Ohno-Oishi, Hiroshi Tawarayama, Hiroshi Kunikata, Charles F. Zorumski and Toru Nakazawa
Biomedicines 2025, 13(7), 1670; https://doi.org/10.3390/biomedicines13071670 - 8 Jul 2025
Viewed by 474
Abstract
Background: An acute angle-closure attack (AAC) is an ocular emergency that results from a rapid increase in intraocular pressure (IOP). Sustained IOP elevation induces severe degeneration of retinal ganglion cells (RGCs) without treatment. Overactivated microglia, key participants in innate immune responses, have [...] Read more.
Background: An acute angle-closure attack (AAC) is an ocular emergency that results from a rapid increase in intraocular pressure (IOP). Sustained IOP elevation induces severe degeneration of retinal ganglion cells (RGCs) without treatment. Overactivated microglia, key participants in innate immune responses, have critical roles in the pathogenesis of IOP-induced RGC death, although precise mechanisms remain unclear. In the present study, we used a rat ex vivo acute glaucoma model to investigate the role of microglial signaling in RGC death and examined whether pharmacological depletion of microglia using a CSF-1R inhibitor, PLX5622, exerts neuroprotection against pressure-induced retinal injury. Methods: Ex vivo rat retinas were exposed to hydrostatic pressure (10 mmHg or 75 mmHg) for 24 h. Pressure-dependent changes in retinal microglia and RGCs were detected by immunofluorescence. Morphological changes in the retina and RGC apoptosis were examined using light microscopy and TUNEL staining, respectively. The expression of NLRP3, active caspase-1, pro IL-1β, and IL-1β were examined using Western blotting. Effects of PLX5622, an agent that depletes microglia, were examined in morphology, apoptosis, and protein expression assays, while TAK-242, a TLR4 inhibitor, was examined against protein expression. Results: Pressure loading at 75 mmHg markedly increased activated microglia and apoptotic RGCs in the isolated retinas. Western blotting revealed increases in expression of NLRP3, active caspase-1, pro IL-1β, and IL-1β at 75 mmHg compared to 10 mmHg. Inhibition of pressure-induced increases in NLRP3 by TAK-242 indicates that pressure elevation induces RGC death via activation of the TLR4–NLRP3 inflammasome cascade. PLX5622 depleted microglia at 75 mmHg and significantly decreased expression of NLRP3, active caspase-1, pro IL-1β, and IL-1β at 75 mmHg, resulting in preservation of RGCs. Conclusions: These results indicate that pressure elevation induces proliferation of inflammatory microglia and promotes IL-1β production via activation of the TLR4–NLRP3 inflammasome cascade, resulting in RGC death. Pharmacological depletion of microglia with PLX5622 could be a potential neuroprotective approach to preserve RGCs from inflammatory cytokines in AAC eyes. Full article
(This article belongs to the Special Issue Glaucoma: New Diagnostic and Therapeutic Approaches, 2nd Edition)
Show Figures

Figure 1

17 pages, 932 KiB  
Review
Retinal Neurochemistry
by Dominic Man-Kit Lam and George Ayoub
Brain Sci. 2025, 15(7), 727; https://doi.org/10.3390/brainsci15070727 - 8 Jul 2025
Viewed by 331
Abstract
The vertebrate retina is a complex neural tissue composed of a repeating array of distinct cell types that communicate through specialized synaptic connections. The neurochemistry underlying these connections reveals the synaptic chemistry, including the neurotransmitters involved and their corresponding receptors. The basic pattern [...] Read more.
The vertebrate retina is a complex neural tissue composed of a repeating array of distinct cell types that communicate through specialized synaptic connections. The neurochemistry underlying these connections reveals the synaptic chemistry, including the neurotransmitters involved and their corresponding receptors. The basic pattern of communication is that the pathway from photoreceptors to bipolar cells to ganglion cells typically uses glutamate as the signaling transmitter, with three ionotropic and one metabotropic receptor types. In contrast, much of the lateral feedback, performed by horizontal cells and amacrine cells, uses the inhibitory neurotransmitter GABA, while other amacrine cells use glycine or dopamine. This review examines all of these neurotransmitter systems for each retinal cell type, along with how these systems process the visual signals transmitted to the lateral geniculate nucleus and the visual cortex. Full article
(This article belongs to the Special Issue Retinal Neurochemistry and Development)
Show Figures

Figure 1

22 pages, 4242 KiB  
Review
Extracellular Vesicle Metabolomics Holds Promise for Adult Axon Regeneration
by Maria D. Cabrera Gonzalez, Jackson Watson, Laura Leal, Isabella Moceri, Camille Plummer, Biraj Mahato, Abdelrahman Y. Fouda and Sanjoy K. Bhattacharya
Metabolites 2025, 15(7), 454; https://doi.org/10.3390/metabo15070454 - 4 Jul 2025
Viewed by 788
Abstract
Extracellular vesicles (EVs) are bilayer lipid membrane particles that are released by every cell type. These secretions are further classified as exosomes, ectosomes, and microvesicles. They contain biomolecules (RNAs, proteins, metabolites, and lipids) with the ability to modulate various biological processes and have [...] Read more.
Extracellular vesicles (EVs) are bilayer lipid membrane particles that are released by every cell type. These secretions are further classified as exosomes, ectosomes, and microvesicles. They contain biomolecules (RNAs, proteins, metabolites, and lipids) with the ability to modulate various biological processes and have been shown to play a role in intercellular communication and cellular rejuvenation. Various studies suggest exosomes and/or microvesicles as a potential platform for drug delivery. EVs may deliver lipids and nucleotides directly to an injury site in an axon, promoting growth cone stabilization and membrane expansion as well as repair, thus positively modulating adult axon regeneration. In this review, we will provide a perspective on the metabolite composition of EVs in adult axonal regeneration relevant to the central nervous system (CNS), specifically that pertaining to the optic nerve. We will present an overview of the methods for isolation, enrichment, omics data analysis and quantification of extracellular vesicles with the goal of providing direction for future studies relevant to axon regeneration. We will also include current resources for multi-omics data integration relevant to extracellular vesicles from diverse cell types. Full article
Show Figures

Graphical abstract

14 pages, 822 KiB  
Article
Optical Coherence Tomography (OCT) Findings in Post-COVID-19 Healthcare Workers
by Sanela Sanja Burgić, Mirko Resan, Milka Mavija, Saša Smoljanović Skočić, Sanja Grgić, Daliborka Tadić and Bojan Pajic
J. Imaging 2025, 11(6), 195; https://doi.org/10.3390/jimaging11060195 - 12 Jun 2025
Viewed by 1022
Abstract
Recent evidence suggests that SARS-CoV-2 may induce subtle anatomical changes in the retina, detectable through advanced imaging techniques. This retrospective case–control study utilized optical coherence tomography (OCT) to assess medium-term retinal alterations in 55 healthcare workers, including 25 individuals with PCR-confirmed COVID-19 and [...] Read more.
Recent evidence suggests that SARS-CoV-2 may induce subtle anatomical changes in the retina, detectable through advanced imaging techniques. This retrospective case–control study utilized optical coherence tomography (OCT) to assess medium-term retinal alterations in 55 healthcare workers, including 25 individuals with PCR-confirmed COVID-19 and 30 non-COVID-19 controls, all of whom had worked in COVID-19 clinical settings. Comprehensive ophthalmological examinations, including OCT imaging, were conducted six months after infection. The analysis considered demographic variables, comorbidities, COVID-19 severity, risk factors, and treatments received. Central macular thickness (CMT) was significantly increased in the post-COVID-19 group (p < 0.05), with a weak but statistically significant positive correlation between CMT and disease severity (r = 0.245, p < 0.05), suggesting potential post-inflammatory retinal responses. No significant differences were observed in retinal nerve fiber layer (RNFL) or ganglion cell complex (GCL + IPL) thickness. However, mild negative trends in inferior RNFL and average GCL+IPL thickness may indicate early neurodegenerative changes. Notably, patients with comorbidities exhibited a significant reduction in superior and inferior RNFL thickness, pointing to possible long-term neurovascular impairment. These findings underscore the value of OCT imaging in identifying subclinical retinal alterations following COVID-19 and highlight the need for continued surveillance in recovered patients, particularly those with pre-existing systemic conditions. Full article
(This article belongs to the Special Issue Learning and Optimization for Medical Imaging)
Show Figures

Figure 1

18 pages, 5301 KiB  
Article
The Profile of Retinal Ganglion Cell Death and Cellular Senescence in Mice with Aging
by Wen-Ying Wang, Xin Bin, Yanxuan Xu, Si Chen, Shuyi Zhou, Shaowan Chen, Yingjie Cao, Kunliang Qiu and Tsz Kin Ng
Int. J. Mol. Sci. 2025, 26(12), 5436; https://doi.org/10.3390/ijms26125436 - 6 Jun 2025
Viewed by 2854
Abstract
Older age is a risk factor for glaucoma, in which progressive retinal ganglion cell (RGC) loss leads to visual field defects and irreversible visual impairment and even blindness. We recently identified the involvement of cellular senescence in RGC cell death post-optic nerve injury. [...] Read more.
Older age is a risk factor for glaucoma, in which progressive retinal ganglion cell (RGC) loss leads to visual field defects and irreversible visual impairment and even blindness. We recently identified the involvement of cellular senescence in RGC cell death post-optic nerve injury. Here we further aimed to delineate the profile of RGC survival in mice with aging, a physiological process with increasing cellular senescence. The numbers of senescent cells in the ganglion cell layer (GCL) significantly and progressively increased starting at 8 months of age. Yet, significant reduction of ganglion cell complex layer thickness began in the 10-month-old mice, and significant reduction in the number of RGCs began in the 12-month-old mice as compared to the 2-month-old mice. Meanwhile, pyroptosis and ferroptosis markers as well as cellular senescence-related cell cycle arrest proteins p15Ink4b, p16Ink4a, p21Cip1, and p53 were significantly and progressively increased in GCL. In contrast, there were no significant changes in dendritic field, complexity, and branches with increasing ages. Comparing between the 2- and 16-month-old mouse retinas, the differentially expressed genes were involved in the pathways of neurodegeneration, innate immunity, and mitochondrial ATP synthesis. In summary, this study revealed the gradual increase in senescent cells as well as pyroptosis and ferroptosis with progressive RGC reduction in mice with aging. Cellular senescence and the related cell death pathways are potential targets for age-related RGC reduction. Full article
Show Figures

Figure 1

21 pages, 813 KiB  
Review
Light, Sound, and Melatonin: Investigating Multisensory Pathways for Visual Restoration
by Dario Rusciano
Medicina 2025, 61(6), 1009; https://doi.org/10.3390/medicina61061009 - 28 May 2025
Cited by 1 | Viewed by 882
Abstract
Multisensory integration is fundamental for coherent perception and interaction with the environment. While cortical mechanisms of multisensory convergence are well studied, emerging evidence implicates specialized retinal ganglion cells—particularly melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs)—in crossmodal processing. This review explores how hierarchical brain [...] Read more.
Multisensory integration is fundamental for coherent perception and interaction with the environment. While cortical mechanisms of multisensory convergence are well studied, emerging evidence implicates specialized retinal ganglion cells—particularly melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs)—in crossmodal processing. This review explores how hierarchical brain networks (e.g., superior colliculus, parietal cortex) and ipRGCs jointly shape perception and behavior, focusing on their convergence in multisensory plasticity. We highlight ipRGCs as gatekeepers of environmental light cues. Their anatomical projections to multisensory areas like the superior colliculus are well established, although direct evidence for their role in human audiovisual integration remains limited. Through melanopsin signaling and subcortical projections, they may modulate downstream multisensory processing, potentially enhancing the salience of crossmodal inputs. A key theme is the spatiotemporal synergy between melanopsin and melatonin: melanopsin encodes light, while melatonin fine-tunes ipRGC activity and synaptic plasticity, potentially creating time-sensitive rehabilitation windows. However, direct evidence linking ipRGCs to audiovisual rehabilitation remains limited, with their role primarily inferred from anatomical and functional studies. Future implementations should prioritize quantitative optical metrics (e.g., melanopic irradiance, spectral composition) to standardize light-based interventions and enhance reproducibility. Nonetheless, we propose a translational framework combining multisensory stimuli (e.g., audiovisual cues) with circadian-timed melatonin to enhance recovery in visual disorders like hemianopia and spatial neglect. By bridging retinal biology with systems neuroscience, this review redefines the retina’s role in multisensory processing and offers novel, mechanistically grounded strategies for neurorehabilitation. Full article
(This article belongs to the Section Ophthalmology)
Show Figures

Figure 1

44 pages, 18795 KiB  
Article
Citicoline and Coenzyme Q10: Therapeutic Agents for Glial Activation Reduction in Ocular Hypertension
by José A. Matamoros, Sara Rubio-Casado, José A. Fernández-Albarral, Miguel A. Martínez-López, Ana I. Ramírez, Elena Salobrar-García, Eva M. Marco, Victor Paleo-García, Rosa de Hoz, Inés López-Cuenca, Lorena Elvira-Hurtado, Lidia Sánchez-Puebla, José M. Ramírez, Meritxell López-Gallardo and Juan J. Salazar
Pharmaceuticals 2025, 18(5), 694; https://doi.org/10.3390/ph18050694 - 8 May 2025
Viewed by 2579
Abstract
Background/Objectives: The loss of retinal ganglion cells (RGCs) is a hallmark of glaucoma, a major cause of blindness. Glial cell activation due to increased intraocular pressure (IOP) significantly contributes to RGC death. Therefore, substances with anti-inflammatory properties could help prevent that process. [...] Read more.
Background/Objectives: The loss of retinal ganglion cells (RGCs) is a hallmark of glaucoma, a major cause of blindness. Glial cell activation due to increased intraocular pressure (IOP) significantly contributes to RGC death. Therefore, substances with anti-inflammatory properties could help prevent that process. This study investigated whether combining Citicoline and Coenzyme Q10 (CoQ10) can reduce glial activation in the retina and the rest of the visual pathway, potentially preventing neurodegeneration in a mouse model of unilateral laser-induced ocular hypertension (OHT). Methods: Four groups of mice were used: vehicle (n = 12), CitiQ10 (n = 12), OHT–vehicle (n = 18), and OHT–CitiQ10 (n = 18). The administration of Citicoline and CoQ10 was performed orally once a day, initiated 15 days prior to the laser treatment and maintained post-treatment until sacrifice (3 days for retina or 7 days for the rest of the visual pathway). The retina, dorsolateral geniculate nucleus, superior colliculus, and visual cortex (V1) were immunohistochemically stained and analyzed. Results: In the laser–CitiQ10 group, the Citicoline + CoQ10 compound revealed (1) an IOP decrease at 24 h and 3 days post-laser; and (2) reduced signs of macroglial (decreased GFAP area) and microglial (soma size, arbor area, microglia number, P2RY12 expression) activation in the retina and in the rest of the visual pathway (reduced activated microglial phenotypes and lower GFAP expression). Conclusions: This study shows that oral administration of Citicoline and CoQ10 can reduce glial activation caused by increased IOP in retina and visual pathway in a mouse model of OHT, potentially protecting RGCs from OHT-induced inflammation. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

18 pages, 4154 KiB  
Article
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
by Yue Hu, Lynn Michelle Grodzki and Udo Bartsch
Cells 2025, 14(9), 643; https://doi.org/10.3390/cells14090643 - 28 Apr 2025
Viewed by 830
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 [...] Read more.
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. Full article
Show Figures

Figure 1

26 pages, 2831 KiB  
Article
Catalpol Protects Against Retinal Ischemia Through Antioxidation, Anti-Ischemia, Downregulation of β-Catenin, VEGF, and Angiopoietin-2: In Vitro and In Vivo Studies
by Howard Wen-Haur Chao, Windsor Wen-Jin Chao and Hsiao-Ming Chao
Int. J. Mol. Sci. 2025, 26(9), 4019; https://doi.org/10.3390/ijms26094019 - 24 Apr 2025
Viewed by 592
Abstract
Retinal ischemic disorders present significant threats to vision, characterized by inadequate blood supply oxygen–glucose deprivation (OGD), oxidative stress, and cellular injury, often resulting in irreversible injury. Catalpol, an iridoid glycoside derived from Rehmannia glutinosa, has demonstrated antioxidative and neuroprotective effects. This study [...] Read more.
Retinal ischemic disorders present significant threats to vision, characterized by inadequate blood supply oxygen–glucose deprivation (OGD), oxidative stress, and cellular injury, often resulting in irreversible injury. Catalpol, an iridoid glycoside derived from Rehmannia glutinosa, has demonstrated antioxidative and neuroprotective effects. This study aimed at investigating the protective effects and mechanisms of catalpol against oxidative stress or OGD in vitro and retinal ischemia in vivo, focusing on the modulation of key biomarkers of retinal ischemia, including HIF-1α, vascular endothelial growth factor (VEGF), angiopoietin-2, MCP-1, and the Wnt/β-catenin pathway. Cellular viability was assessed using retinal ganglion cell-5 (RGC-5) cells cultured in DMEM; a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed. H2O2 (1 mM)/OGD was utilized. Vehicle or different catalpol concentrations were administered 15 min before the ischemic-like insults. The Wistar rat eyes’ intraocular pressure was increased to 120 mmHg for 60 min to induce retinal ischemia. Intravitreous injections of catalpol (0.5 or 0.25 mM), Wnt inhibitor DKK1 (1 μg/4 μL), anti-VEGF Lucentis (40 μg/4 μL), or anti-VEGF Eylea (160 μg/4 μL) were administered to the rats’ eyes 15 min before or after retinal ischemia. Electroretinogram (ERG), fluorogold retrograde labeling RGC, Western blotting, ELISA, RT-PCR, and TUNEL were utilized. In vitro, both H2O2 and OGD models significantly (p < 0.001/p < 0.001; H2O2 and OGD) induced oxidative stress/ischemic-like insults, decreasing RGC-5 cell viability (from 100% to 55.14 ± 2.19%/60.84 ± 4.57%). These injuries were insignificantly (53.85 ± 1.28% at 0.25 mM)/(63.46 ± 3.30% at 0.25 mM) and significantly (p = 0.003/p = 0.012; 64.15 ± 2.41%/77.63 ± 8.59% at 0.5 mM) altered by the pre-administration of catalpol, indicating a possible antioxidative and anti-ischemic effect of 0.5 mM catalpol. In vivo, catalpol had less effect at 0.25 mM for ERG amplitude ratio (median [Q1, Q3] 14.75% [12.64%, 20.48%]) and RGC viability (mean ± SE 63.74 ± 5.13%), whereas (p < 0.05 and p < 0.05) at 0.5 mM ERG’s ratio (35.43% [24.35%, 43.08%]) and RGC’s density (74.34 ± 5.10%) blunted the ischemia-associated significant (p < 0.05 and p < 0.01) reduction in ERG b-wave amplitude (6.89% [4.24%, 10.40%]) and RGC cell viability (45.64 ± 3.02%). Catalpol 0.5 mM also significantly protected against retinal ischemia supported by the increased amplitude ratio of ERG a-wave and oscillatory potential, along with recovering a delayed a-/b-wave response time ratio. When contrasted with DKK1 or Lucentis, catalpol exhibited similar protective effects against retinal ischemia via significantly (p < 0.05) blunting the ischemia-induced overexpression of β-catenin, VEGF, or angiopoietin-2. Moreover, ischemia-associated significant increases in apoptotic cells in the inner retina, inflammatory biomarker MCP-1, and ischemic indicator HIF-1α were significantly nullified by catalpol. Catalpol demonstrated antiapoptotic, anti-inflammatory, anti-ischemic (in vivo retinal ischemia or in vitro OGD), and antioxidative (in vitro) properties, counteracting retinal ischemia via suppressing upstream Wnt/β-catenin and inhibiting downstream HIF-1α, VEGF, and angiopoietin-2, together with its decreasing TUNEL apoptotic cell number and inflammatory MCP-1 concentration. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

16 pages, 4152 KiB  
Article
Tauroursodeoxycholic Acid Protects Retinal Ganglion Cells and Reduces Inflammation in Mice Following Optic Nerve Crush
by Nan Zhang, Ying Li, Xian Zhang, Micah A. Chrenek, Jiaxing Wang, Preston E. Girardot, Jana T. Sellers, Eldon E. Geisert, John M. Nickerson and Jeffrey H. Boatright
Pharmaceuticals 2025, 18(4), 569; https://doi.org/10.3390/ph18040569 - 14 Apr 2025
Viewed by 873
Abstract
Purpose: The aim of this study was to investigate the protective effects of systemically administered tauroursodeoxycholic acid (TUDCA) in an optic nerve crush (ONC) mouse model of retinal ganglion cell (RGC) death. Methods: C57BL/6J mice were injected intraperitoneally (i.p.) three times per week [...] Read more.
Purpose: The aim of this study was to investigate the protective effects of systemically administered tauroursodeoxycholic acid (TUDCA) in an optic nerve crush (ONC) mouse model of retinal ganglion cell (RGC) death. Methods: C57BL/6J mice were injected intraperitoneally (i.p.) three times per week with TUDCA (500 mg/kg) for two weeks, after which unilateral ONC was performed. A control cohort was identically treated with a drug vehicle (phosphate buffered saline; PBS). A separate cohort did not undergo any injections or surgeries (this was termed the “Naïve” group). Pattern electroretinography (PERG) was recorded 3 days after ONC. Retinas were harvested for whole-mount immunofluorescence staining with an antibody against RGC marker Brn3a and imaged by fluorescent confocal microscopy. Apoptotic cells in the ganglion cell layer (GCL) were detected by Terminal Deoxynucleotidyl Transferase-Mediated dUTP Nick End Labeling (TUNEL) performed on fixed retina sections. Glial fibrillary acidic protein (GFAP) immunostaining on fixed retina sections was conducted to detect the activation of Müller cells. Total RNA was extracted from retinas and expression of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and IL-10 was determined by digital droplet PCR (ddPCR). Results: TUDCA treatment preserved visual function as assessed by PERG. P1 and N2 amplitudes from the PBS-treated ONC group were significantly diminished compared to those of the Naïve group (p < 0.001). TUDCA treatment prevented this diminution. The amplitudes of P1 and N2 in the TUDCA-treated ONC group were statistically indistinguishable from those of the Naïve group and were higher than the PBS-treated ONC group (TUDCA+ONC vs. PBS+ONC, P1: 6.99 ± 0.89 µV vs. 3.60 ± 0.69 µV, p < 0.01; N2: −9.30 (IQR: −13.43–−6.44) µV vs. −4.47 (IQR: −10.26–−2.17) µV). TUDCA treatment preserved RGCs. The ONC-vehicle-only group had 25% fewer RGCs (Brn3a-positive cells) than Naïve eyes (p < 0.0001). TUDCA treatment nearly completely prevented this loss, preserving all but 7.7% of the RGCs, and the number of RGCs in the TUDCA-treated ONC group was significantly higher than in the PBS-treated ONC group (TUDCA+ONC vs. PBS+ONC, 1738.00 ± 14.43 cells per field vs. 1454.00 ± 6.55 cells per field, p < 0.0001). The number of TUNEL-positive cells in the GCL (Naïve vs. PBS+ONC group: 1.00 (IQR: 0.00–2.00) % vs. 37.00 (IQR: 8.50–48.50) %, p < 0.05) and GFAP-positive fibers transversing retina sections (Naïve vs. PBS+ONC group: 33.00 ± 1.15 vs. 185.70 ± 42.37 fibers/retina, p < 0.05), and the expression of IL-6, TNF-α were significantly greater in the PBS-treated ONC group compared to that of the Naïve group (Naïve vs. PBS+ONC group, IL-6: 0.07 (IQR: 0.06–0.31) vs. 0.99 (IQR: 0.56–1.47), p < 0.05, TNF-α: 0.19 ± 0.069 vs. 1.39 ± 0.23; p < 0.01), an increase not observed with TUDCA treatment. Conclusions: Systemic TUDCA treatment significantly preserved RGC function and survival in the mouse ONC model of RGC damage. TUDCA treatment prevented RGC apoptosis, Müller glial cell activation, and retinal expression of several inflammatory cytokines. These data suggest that TUDCA is a promising therapeutic candidate for preserving RGC numbers and function. Full article
Show Figures

Graphical abstract

16 pages, 3851 KiB  
Article
Long–Term Impairment of Retinal Ganglion Cell Function After Oxygen–Induced Retinopathy
by Adam M. Schmitz, Stephanie M. Bumbaru, Laith S. Fakhouri and Dao-Qi Zhang
Cells 2025, 14(7), 512; https://doi.org/10.3390/cells14070512 - 29 Mar 2025
Viewed by 589
Abstract
Premature infants with retinopathy of prematurity (ROP) have neovascularization of the retina, potentially resulting in low vision and even blindness. Some of these infants still have visual impairment, even if ROP resolves as they age. However, the mechanisms underlying the visual problems post–ROP [...] Read more.
Premature infants with retinopathy of prematurity (ROP) have neovascularization of the retina, potentially resulting in low vision and even blindness. Some of these infants still have visual impairment, even if ROP resolves as they age. However, the mechanisms underlying the visual problems post–ROP are poorly understood. Because the pathological neovascularization in ROP infants can be mimicked in a mouse model with oxygen–induced retinopathy (OIR), we recapitulated post–ROP with post–OIR mice a few months after spontaneous regression of retinal neovascularization. Our pattern electroretinogram test demonstrates that post–OIR mice exhibit reduced P1–N2 responses, suggesting the impairment of retinal ganglion cells, the retina’s output neurons. However, immunohistochemistry reveals that the density of retinal ganglion cells remains unchanged in post–OIR mice, indicating that the aforementioned pattern electroretinogram changes are functional. Our data further demonstrate that both light–adapted ex vivo electroretinogram a–waves (cone responses) and in vivo electroretinogram b–waves (ON cone bipolar cell responses) were significantly impaired in post–OIR mice. These results suggest that post–OIR impairment of the retinal cone pathway appears to result in the dysfunction of retinal ganglion cells, contributing to visual problems. A similar cellular mechanism could occur in post–ROP children, which is responsible for their visual impairment. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

Back to TopTop