1
|
Thompson JL, McCool S, Smith JC, Schaal V, Pendyala G, Yelamanchili S, Van Hook MJ. Microglia remodeling in the visual thalamus of the DBA/2J mouse model of glaucoma. PLoS One 2025; 20:e0323513. [PMID: 40373024 PMCID: PMC12080812 DOI: 10.1371/journal.pone.0323513] [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: 01/10/2025] [Accepted: 04/10/2025] [Indexed: 05/17/2025] Open
Abstract
Microglia are the resident immune cells of the central nervous system and mediate a broad array of adaptations during disease, injury, and development. Typically, microglia morphology is understood to provide a window into their function and microglia have the capacity to adopt a broad spectrum of functional phenotypes characterized by numerous morphologies and gene expression profiles. Glaucoma, which leads to blindness from retinal ganglion cell (RGC) degeneration, is commonly associated with elevated intraocular pressure (IOP) and triggers microglia responses within the retinal layers, at the optic nerve head, and in retinal projection targets in the brain. The goal of this study was to determine the relationship of microglia morphology to intraocular pressure and the loss of RGC output synapses in the dorsolateral geniculate nucleus (dLGN), a RGC projection target in the thalamus that conveys information to the primary visual cortex. We accomplished this by analyzing microglia morphologies in dLGN sections from DBA/2J mice, which develop a form of inherited glaucoma, at 4, 9, and 12 months of age, representing distinct time points in disease progression. Microglia morphology was analyzed using skeletonized Iba1 fluorescence images and fractal analyses of individually reconstructed microglia cells. We found that microglia in older DBA/2J mice adopted simplified morphologies, characterized by fewer endpoints and less total process length per microglia cell. There was an age-dependent shift in microglia morphology in tissue from control mice (DBA/2JGpnmb+) that was accelerated in DBA/2J mice. Measurements of microglia morphology correlated with cumulative IOP, immunofluorescence labeling for complement component C1q, and vGluT2-labeled RGC axon terminal density. Additionally, fractal analysis revealed a clear distinction between control and glaucomatous dLGN, with microglia from ocular hypertensive DBA/2J dLGN tissue showing an elongated rod-like morphology. RNA-sequencing of dLGN showed an upregulation of immune system-related genes. These results suggest that microglia in the dLGN alter their physiology to respond to RGC degeneration in glaucoma, potentially contributing to CNS adaptations to neurodegenerative vision loss.
Collapse
Affiliation(s)
- Jennifer L. Thompson
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska United States of America
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska United States of America
| | - Shaylah McCool
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska United States of America
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska United States of America
| | - Jennie C. Smith
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska United States of America
| | - Victoria Schaal
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska United States of America
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska United States of America
| | - Sowmya Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska United States of America
| | - Matthew J. Van Hook
- Department of Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska United States of America
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
2
|
Norouz Dolatabadi E, Akbarzadeh Zaky MR, Hashim Abbas F, Eftekhari Milani A, André H, Alizadeh E. Recent advances on modeling retinal disease: Towards efficient gene/drug therapy. Exp Eye Res 2025; 256:110416. [PMID: 40320033 DOI: 10.1016/j.exer.2025.110416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/22/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Advanced modeling biotechnologies are required to understand retinal diseases and develop effective treatments based on the patient's genetic background, lifestyle, and environment. In this work, recent advances in different types of study models that are used in the retinal disease area of research will be explored. The retinal models to be covered are: in vivo systems (human and animal), in vitro organisms (cell lines, primary cells, patient-derived stem cells, microfluidics, organoids, and spheroids), ex vivo models (explant cultures and retinal tissue preparations), and in silico models (computational and mathematical). Moreover, the unique comprehension of models of retinal disease, advantages, and disadvantages will be scrutinized. Finally, innovations/improvements derived from models towards gene and pharmacological therapy that display promise for treating retinal illnesses are elucidated.
Collapse
Affiliation(s)
- Elham Norouz Dolatabadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fatima Hashim Abbas
- Department of Aesthetic and Laser Techniques, College of Health and Medical Techniques, Al-Mustagbal University, Babylon, Iraq
| | | | - Helder André
- Department of Clinical Neuroscience, Karolinska Institute, Karolinska, Sweden
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Endocrin Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
İnam MG, İnam O, Yang X, Zeng Q, Tezel G. Integrating Retinal Segmentation Metrics with Machine Learning for Predictions from Mouse SD-OCT Scans. Curr Eye Res 2025; 50:502-511. [PMID: 39849306 PMCID: PMC12037304 DOI: 10.1080/02713683.2025.2456783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/14/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
PURPOSE This study aimed to initially test whether machine learning approaches could categorically predict two simple biological features, mouse age and mouse species, using the retinal segmentation metrics. METHODS The retinal layer thickness data obtained from C57BL/6 and DBA/2J mice were processed for machine learning after segmenting mouse retinal SD-OCT scans. Twenty-two models were trained to predict the mouse groups. The best neural network model was optimized for better outcomes. Prediction accuracy, the area under the curve, sensitivity, specificity, precision, and F-1 score values were obtained. RESULTS The Wilcoxon Signed-Rank test provided significantly higher validation accuracy for neural networks than decision trees, discriminant analysis, support vector machines, and k-nearest neighbor classifiers (p = 0.005 for all). For C57BL/6-DBA/2J classification, a mean validation accuracy of 88.11 ± 3.92% (95% CI: 86.99-89.22) was achieved for the neural network when the optimized neural network had 92.31% final test accuracy with an area under the curve value of 0.9762, 94.44% sensitivity, 90.48% specificity, 89.47% precision, and 0.92 F-1 score. The optimized neural network model for age group differentiation had a final test accuracy of 82.05% with a 0.9064 area under the curve value, 77.27% sensitivity, 88.24% specificity, 89.47% precision, and 0.83 F-1 score. CONCLUSIONS These findings validate that machine learning, using segmentation metrics instead of images, can effectively analyze retinal OCT scans in mice for categorical predictions in experimental models. Expanding this approach with additional features, including histopathological and functional correlations, is expected to improve the prediction power further, promising valuable applications to predict more complex outcomes in experimental and clinical studies.
Collapse
Affiliation(s)
- Maide Gözde İnam
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Onur İnam
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
- Department of Biophysics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Xiangjun Yang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Qun Zeng
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Gülgün Tezel
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
4
|
Huang J, Chang Z, Deng X, Cai S, Jiang B, Zeng W, Ke M. Identification of Sequential Molecular Mechanisms and Key Biomarkers in Early Glaucoma by Integrated Bioinformatics Analysis. Mol Neurobiol 2025; 62:4952-4970. [PMID: 39495230 DOI: 10.1007/s12035-024-04563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Glaucoma is a neurodegenerative disease characterized by progressive optic nerve degeneration and retinal ganglion cell (RGC) loss. In early glaucoma, before obvious axon loss, highly organized pathological processes in RGCs occur sequentially, involving axons, dendrites and synaptic terminals. The optic nerve head (ONH) is the critical structure of early glaucomatous neurodegeneration. Taking advantage of high-throughput data from the ONH and the weighted gene coexpression network analysis (WGCNA) method, the current study aims to gain insight into the full scope of pathological events in early glaucoma and define their chronological sequence. The expression profiles of GSE26299, GSE110019, and GSE139605, which measure ONH gene expression in different glaucoma models, were downloaded from the Gene Expression Omnibus (GEO) database. In GSE26299, which uses 10.5-month-old DBA/2 J mice, WGCNA was utilized to construct a gene coexpression network, and the most significant modules of early (NOE), moderate (MOD) and severe (SEV) glaucoma were identified. The differentially expressed genes (DEGs) of GSE110019 and GSE139605 significantly overlapped with the correlated module of the MOD group, so the 3 gene sets were analyzed together. Pathway enrichment analysis via the GO, KEGG, and Reactome pathways was subsequently performed, followed by protein‒protein interaction (PPI) analysis to screen key genes associated with each stage. Several hub gene expression patterns were identified in a glucocorticoid-induced glaucoma (GIG) model via quantitative PCR and immunostaining. The pink module was positively correlated with the NOE group (r = 0.48, p = 4e-04) and negatively correlated with the glaucoma stage (r = -0.88, p = 3e-17). The genes in the pink module were enriched in the synaptic transmission and axonal transport pathways. The tan module was negatively correlated with the NOE group (r = -0.43, p = 0.002) and positively correlated with the glaucoma stage (r = 0.77, p = 7e-11). The genes in the tan module were associated with pathways such as tight junctions, retinol metabolism, and linoleic acid metabolism. The purple module was positively correlated with the MOD group (r = 0.64, p = 5e-07). The common genes among the purple module and the DEGs of the two other datasets were enriched in pathways related to mitotic cell division, cytokine activity, and the extracellular matrix (ECM). The hub genes identified by PPI included Nrn1, Cplx1, Timp1, and Cdk1. Quantitative PCR and immunostaining confirmed that Limk1 expression was increased in the ONH of GIG mice. In early glaucomatous neuropathy, intrinsic changes in RGCs precede the activation of glial cells and ECM remodeling. These latter events are common pathological changes observed in the ONH in both cats and mice. Our study may provide new targets for the early detection and treatment of glaucoma.
Collapse
Affiliation(s)
- Jingqiu Huang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhaohui Chang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xizhi Deng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuncheng Cai
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bin Jiang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wen Zeng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
5
|
Bahrani Fard MR, Chan J, Read AT, Li G, Cheng L, Safa BN, Siadat SM, Jhunjhunwala A, Grossniklaus HE, Emelianov SY, Stamer WD, Kuehn MH, Ethier CR. MAGNETICALLY STEERED CELL THERAPY FOR REDUCTION OF INTRAOCULAR PRESSURE AS A TREATMENT STRATEGY FOR OPEN-ANGLE GLAUCOMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.13.593917. [PMID: 38798683 PMCID: PMC11118342 DOI: 10.1101/2024.05.13.593917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Trabecular meshwork (TM) cell therapy has been proposed as a next-generation treatment for elevated intraocular pressure (IOP) in glaucoma, the most common cause of irreversible blindness. Using a magnetic cell steering technique with excellent efficiency and tissue-specific targeting, we delivered two types of cells into a mouse model of glaucoma: either human adipose-derived mesenchymal stem cells (hAMSCs) or induced pluripotent cell derivatives (iPSC-TM cells). We observed a 4.5 [3.1, 6.0] mmHg or 27% reduction in intraocular pressure (IOP) for nine months after a single dose of only 1500 magnetically-steered hAMSCs, explained by increased outflow through the conventional pathway and associated with an higher TM cellularity. iPSC-TM cells were also effective, but less so, showing only a 1.9 [0.4, 3.3] mmHg or 13% IOP reduction and increased risk of tumorigenicity. In both cases, injected cells remained detectable in the iridocorneal angle three weeks post-transplantation. Based on the locations of the delivered cells, the mechanism of IOP lowering is most likely paracrine signaling. We conclude that magnetically-steered hAMSC cell therapy has potential for long-term treatment of ocular hypertension in glaucoma. One Sentence Summary A novel magnetic cell therapy provided effective intraocular pressure reduction in a mouse model, motivating future translational studies.
Collapse
|
6
|
Sarkis S, Chamard C, Johansen B, Daien V, Michon F. Challenging glaucoma with emerging therapies: an overview of advancements against the silent thief of sight. Front Med (Lausanne) 2025; 12:1527319. [PMID: 40206485 PMCID: PMC11979169 DOI: 10.3389/fmed.2025.1527319] [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: 11/13/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Glaucoma, a leading cause of irreversible blindness, represents a significant challenge in ophthalmology. This review examines recent advancements in glaucoma treatment, focusing on innovative medications and creative strategies. While new agents offer promising methods for lowering intraocular pressure (IOP), they also pose challenges related to efficacy and side effects. Alongside IOP reduction, emerging neuroprotective approaches are being explored to safeguard retinal ganglion cells (RGCs) from glaucoma-induced damage. The review also evaluates the potential of novel drug delivery systems, such as biodegradable implants and nanoparticles, to enhance treatment effectiveness and patient adherence. Additionally, it highlights the role of personalized medicine in identifying new biomarkers and customizing therapies based on individual genetic and environmental factors.
Collapse
Affiliation(s)
- Solange Sarkis
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Laboratoires Théa, Clermont-Ferrand, France
| | - Chloé Chamard
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| | | | - Vincent Daien
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
- Sydney Medical School, The Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Frederic Michon
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
7
|
Roddy GW, Kohli D, Niknam P, Omer ME, Chowdhury UR, Anderson KJ, Pacheco Marrero JM, Rinkoski TA, Fautsch MP. Subconjunctival Administration of an Adeno-Associated Virus Expressing Stanniocalcin-1 Provides Sustained Intraocular Pressure Reduction in Mice. OPHTHALMOLOGY SCIENCE 2025; 5:100590. [PMID: 39328825 PMCID: PMC11426120 DOI: 10.1016/j.xops.2024.100590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/28/2024]
Abstract
Purpose To investigate subconjunctival administration of a single-stranded, adeno-associated virus, serotype 2, engineered to express stanniocalcin-1 with a FLAG tag (ssAAV2-STC-1-FLAG) as a novel sustained (IOP) lowering agent with a reduced ocular surface side effect profile. Design In vivo preclinical investigation in mice. Subjects C57BL/6J, DBA/2J, prostaglandin F (FP) receptor knockout mice. Methods Normotensive C57BL/6J mice were treated with a subconjunctival injection of ssAAV2-STC-1-FLAG (2 μL; 6 × 109 viral genomes [VGs]) in 1 eye and the same volume and concentration of ssAAV2-green fluorescent protein (GFP) or the same volume of phosphate-buffered saline in the fellow eye. Ocular hypertensive DBA/2J mice were subconjunctivally injected with 6 × 109 VGs of ssAAV2-STC-1-FLAG or ssAAV2-GFP. Steroid-mediated ocular hypertension was induced in C57BL/6J mice with weekly injections of dexamethasone into the conjunctival fornix, and mice were then injected subconjunctivally with 6 × 109 VGs of ssAAV2-STC-1-FLAG or ssAAV2-GFP. Prostaglandin F receptor knockout mice were injected subconjunctivally with 6 × 109 VGs of ssAAV2-STC-1-FLAG or phosphate-buffered saline. An identical vector was constructed without the FLAG tag (ssAAV2-STC-1) and evaluated in normotensive C57BL/6J mice. Intraocular pressure was assessed using the Tonolab tonometer for all experiments. Tumor necrosis factor alpha (TNFα), a marker of ocular surface inflammation, was compared between subconjunctivally delivered ssAAV2-STC-1-FLAG and other treatments including daily topical latanoprost. Main Outcome Measures Intraocular pressure assessment. Results Subconjunctival delivery of ssAAV2-STC-1-FLAG significantly reduced IOP for 10 weeks post injection in normotensive mice. Maximal IOP reduction was seen at week 3 postinjection (17.4%; 17.1 ± 0.8 vs. 14.1 ± 0.8 mmHg, P < 0.001). After the IOP-lowering effect had waned, a second injection restored the ocular hypotensive effect. Subconjunctivally delivered ssAAV2-STC-1-FLAG lowered IOP in DBA/2J mice (16.9%; 17.8 ± 2.0 vs. 14.8 ± 0.9 mmHg, P < 0.001) and steroid-mediated ocular hypertensive mice (20.0%; 19.0 ± 0.6 vs. 15.2 ± 0.7 mmHg, P < 0.001) over the experimental period. This construct also reduced IOP to a similar extent in wild-type (15.9%) and FP receptor knockout (15.7%) mice compared with the fellow eye. A related construct also lowered IOP without the FLAG tag in a similar manner. Reduction in conjunctival TNFα was seen when comparing subconjunctivally delivered ssAAV2-STC-1-FLAG to daily topical latanoprost. Conclusions Subconjunctival delivery of the STC-1 transgene with a vector system may represent a novel treatment strategy for sustained IOP reduction and improved ocular tolerability that also avoids the daily dosing requirements of currently available medications. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Gavin W. Roddy
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, 55905
| | - Darrell Kohli
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, 55905
| | - Parvin Niknam
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, 55905
| | - Mohammed E. Omer
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, 55905
| | | | | | | | - Tommy A. Rinkoski
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, 55905
| | | |
Collapse
|
8
|
Standley A, Xie J, Lau AW, Grote L, Gifford AJ. Working with Miraculous Mice: Mus musculus as a Model Organism. Curr Protoc 2024; 4:e70021. [PMID: 39435766 DOI: 10.1002/cpz1.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The laboratory mouse has been described as a "miracle" model organism, providing a window by which we may gain an understanding of ourselves. Since the first recorded mouse experiment in 1664, the mouse has become the most used animal model in biomedical research. Mice are ideally suited as a model organism because of their small size, short gestation period, large litter size, and genetic similarity to humans. This article provides a broad overview of the laboratory mouse as a model organism and is intended for undergraduates and those new to working with mice. We delve into the history of the laboratory mouse and outline important terminology to accurately describe research mice. The types of laboratory mice available to researchers are reviewed, including outbred stocks, inbred strains, immunocompromised mice, and genetically engineered mice. The critical role mice have played in advancing knowledge in the areas of oncology, immunology, and pharmacology is highlighted by examining the significant contribution of mice to Nobel Prize winning research. International mouse mutagenesis programs and accurate phenotyping of mouse models are outlined. We also explain important considerations for working with mice, including animal ethics; the welfare principles of replacement, refinement, and reduction; and the choice of mouse model in experimental design. Finally, we present practical advice for maintaining a mouse colony, which involves adequate training of staff, the logistics of mouse housing, monitoring colony health, and breeding strategies. Useful resources for working with mice are also listed. The aim of this overview is to equip the reader with a broad appreciation of the enormous potential and some of the complexities of working with the laboratory mouse in a quest to improve human health. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anick Standley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jinhan Xie
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Angelica Wy Lau
- Garvan Institute of Medical Research, St Vincent's Clinical School, Darlinghurst, NSW, Australia
| | - Lauren Grote
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- Anatomical Pathology, NSW Heath Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Liu J, Zhou L, Wu X, Chen Z, Zheng X, Wang H, So KF, Ma L, Wang J, Chiu K. Lycium ruthenicum water extract preserves retinal ganglion cells in chronic ocular hypertension mouse models. Front Pharmacol 2024; 15:1404119. [PMID: 39021836 PMCID: PMC11252021 DOI: 10.3389/fphar.2024.1404119] [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: 03/20/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Lycium ruthenicum Murray (LR), known as "black goji berry" or "black wolfberry", is widely utilized in chinese herbal medicine. LR fruit showed its antioxidant and/or anti-inflammation activity in treating cardiac injury, experimental colitis, nonalcoholic fatty liver disease, fatigue, and aging. Glaucoma is the leading cause of irreversible blindness. Besides elevated intraocular pressure (IOP), oxidative stress and neuroinflammation were recognized to contribute to the pathogenesis of glaucoma. This study investigated the treatment effects of LR water extract (LRE) on retinal ganglion cells (RGCs) threatened by sustained IOP elevation in a laser-induced chronic ocular hypertension (COH) mouse model and the DBA/2J mouse strain. The antioxidation and anti-inflammation effects of LRE were further tested in the H2O2-challenged immortalized microglial (IMG) cell line in vitro. LRE oral feeding (2 g/kg) preserved the function of RGCs and promoted their survival in both models mimicking glaucoma. LRE decreased 8-hydroxyguanosine (oxidative stress marker) expression in the retina. LRE reduced the number of Iba-1+ microglia in the retina of COH mice, but not in the DBA/2J mice. At the mRNA level, LRE reversed the COH induced HO-1 and SOD-2 overexpressions in the retina of COH mice. Further in vitro study demonstrated that LRE pretreatment to IMG cells could significantly reduce H2O2 induced oxidative stress through upregulation of GPX-4, Prdx-5, HO-1, and SOD-2. Our work demonstrated that daily oral intake of LRE can be used as a preventative/treatment agent to protect RGCs under high IOP stress probably through reducing oxidative stress and inhibiting microglial activation in the retina.
Collapse
Affiliation(s)
- Jinfeng Liu
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, Shenzhen, China
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lina Zhou
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xueping Wu
- Jinzhou Medical University, Jinzhou, China
| | - Zihang Chen
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Kwok Fai So
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Psychology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Lan Ma
- Institute of Biopharmaceutical and Health Engineering, Tsinghua University Shenzhen Graduate School, Tsinghua University, Shenzhen, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Jiantao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, Shenzhen, China
| | - Kin Chiu
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Psychology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
10
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [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: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
11
|
Koike S, Keino-Masu K, Masu M. Enpp2 haploinsufficiency induces an eye-open-at-birth phenotype in the DBA/2 background. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001212. [PMID: 38725939 PMCID: PMC11079641 DOI: 10.17912/micropub.biology.001212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/12/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
Autotaxin, encoded by the Enpp2 gene, produces lysophosphatidic acid (LPA), which exerts numerous biological functions via its cognate receptors. Enpp2 null mutant mice die by embryonic day 9.5 owing to aberrant vascular development in the yolk sac, preventing analysis after that period. In this study, we found that Enpp2 heterozygous mice in the DBA/2 genetic background showed an eye-open-at-birth phenotype at high frequency, caused by failure of eyelid closure during the embryonic stage. Notably, wildtype pups from the Enpp2 heterozygous dam showed the phenotype, although at lower frequency, suggesting that maternal LPA affects the embryonic development.
Collapse
Affiliation(s)
- Seiichi Koike
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Neurobiology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Engineering for Research, University of Toyama, Toyama, Toyama, Japan
| | - Kazuko Keino-Masu
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Neurobiology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masayuki Masu
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Neurobiology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
12
|
Bitard J, Grellier EK, Lourdel S, Filipe HP, Hamon A, Fenaille F, Castelli FA, Chu-Van E, Roger JE, Locker M, Perron M. Uveitic glaucoma-like features in Yap conditional knockout mice. Cell Death Discov 2024; 10:48. [PMID: 38272861 PMCID: PMC10811226 DOI: 10.1038/s41420-023-01791-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Glaucoma is a multifactorial neurodegenerative disease characterized by the progressive and irreversible degeneration of the optic nerve and retinal ganglion cells. Despite medical advances aiming at slowing degeneration, around 40% of treated glaucomatous patients will undergo vision loss. It is thus of utmost importance to have a better understanding of the disease and to investigate more deeply its early causes. The transcriptional coactivator YAP, an important regulator of eye homeostasis, has recently drawn attention in the glaucoma research field. Here we show that Yap conditional knockout mice (Yap cKO), in which the deletion of Yap is induced in both Müller glia (i.e. the only retinal YAP-expressing cells) and the non-pigmented epithelial cells of the ciliary body, exhibit a breakdown of the aqueous-blood barrier, accompanied by a progressive collapse of the ciliary body. A similar phenotype is observed in human samples that we obtained from patients presenting with uveitis. In addition, aged Yap cKO mice harbor glaucoma-like features, including deregulation of key homeostatic Müller-derived proteins, retinal vascular defects, optic nerve degeneration and retinal ganglion cell death. Finally, transcriptomic analysis of Yap cKO retinas pointed to early-deregulated genes involved in extracellular matrix organization potentially underlying the onset and/or progression of the observed phenotype. Together, our findings reveal the essential role of YAP in preserving the integrity of the ciliary body and retinal ganglion cells, thereby preventing the onset of uveitic glaucoma-like features.
Collapse
Affiliation(s)
- Juliette Bitard
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France.
| | - Elodie-Kim Grellier
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France
| | - Sophie Lourdel
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France
| | - Helena Prior Filipe
- West Lisbon Hospitals Center, Hospital de Egas Moniz, Lisbon, Portugal
- Egas Moniz Center for Interdisciplinary Research, Lisbon, Portugal
| | - Annaïg Hamon
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif sur Yvette, France
| | - Florence Anne Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif sur Yvette, France
| | - Emeline Chu-Van
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif sur Yvette, France
| | - Jérôme E Roger
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France
| | - Morgane Locker
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France
| | - Muriel Perron
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, France.
| |
Collapse
|
13
|
Rong S, Yu X, Wiggs JL. Genetic Basis of Pigment Dispersion Syndrome and Pigmentary Glaucoma: An Update and Functional Insights. Genes (Basel) 2024; 15:142. [PMID: 38397132 PMCID: PMC10887877 DOI: 10.3390/genes15020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Pigment Dispersion Syndrome (PDS) and Pigmentary Glaucoma (PG) comprise a spectrum of ocular disorders characterized by iris pigment dispersion and trabecular meshwork changes, resulting in increased intraocular pressure and potential glaucomatous optic neuropathy. This review summarizes recent progress in PDS/PG genetics including rare pathogenic protein coding alterations (PMEL) and susceptibility loci identified from genome-wide association studies (GSAP and GRM5/TYR). Areas for future research are also identified, especially the development of efficient model systems. While substantial strides have been made in understanding the genetics of PDS/PG, our review identifies key gaps and outlines the future directions necessary for further advancing this important field of ocular genetics.
Collapse
Affiliation(s)
- Shisong Rong
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA;
| | - Xinting Yu
- Department of Medicine, Brigham and Women’s Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02115, USA;
| | - Janey L. Wiggs
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA;
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
14
|
Agarwal R, Iezhitsa I. Genetic rodent models of glaucoma in representing disease phenotype and insights into the pathogenesis. Mol Aspects Med 2023; 94:101228. [PMID: 38016252 DOI: 10.1016/j.mam.2023.101228] [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: 07/20/2023] [Revised: 10/31/2023] [Accepted: 11/11/2023] [Indexed: 11/30/2023]
Abstract
Genetic rodent models are widely used in glaucoma related research. With vast amount of information revealed by human studies about genetic correlations with glaucoma, use of these models is relevant and required. In this review, we discuss the glaucoma endophenotypes and importance of their representation in an experimental animal model. Mice and rats are the most popular animal species used as genetic models due to ease of genetic manipulations in these animal species as well as the availability of their genomic information. With technological advances, induction of glaucoma related genetic mutations commonly observed in human is possible to achieve in rodents in a desirable manner. This approach helps to study the pathobiology of the disease process with the background of genetic abnormalities, reveals potential therapeutic targets and gives an opportunity to test newer therapeutic options. Various genetic manipulation leading to appearance of human relevant endophenotypes in rodents indicate their relevance in glaucoma pathology and the utility of these rodent models for exploring various aspects of the disease related to targeted mutation. The molecular pathways involved in the pathophysiology of glaucoma leading to elevated intraocular pressure and the disease hallmark, apoptosis of retinal ganglion cells and optic nerve degeneration, have been extensively explored in genetic rodent models. In this review, we discuss the consequences of various genetic manipulations based on the primary site of pathology in the anterior or the posterior segment. We discuss how these genetic manipulations produce features in rodents that can be considered a close representation of disease phenotype in human. We also highlight several molecular mechanisms revealed by using genetic rodent models of glaucoma including those involved in increased aqueous outflow resistance, loss of retinal ganglion cells and optic neuropathy. Lastly, we discuss the limitations of the use of genetic rodent models in glaucoma related research.
Collapse
Affiliation(s)
- Renu Agarwal
- School of Medicine, International Medical University, Malaysia.
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Malaysia
| |
Collapse
|
15
|
Lo J, Mehta K, Dhillon A, Huang YK, Luo Z, Nam MH, Al Diri I, Chang KC. Therapeutic strategies for glaucoma and optic neuropathies. Mol Aspects Med 2023; 94:101219. [PMID: 37839232 PMCID: PMC10841486 DOI: 10.1016/j.mam.2023.101219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Glaucoma is a neurodegenerative eye disease that causes permanent vision impairment. The main pathological characteristics of glaucoma are retinal ganglion cell (RGC) loss and optic nerve degeneration. Glaucoma can be caused by elevated intraocular pressure (IOP), although some cases are congenital or occur in patients with normal IOP. Current glaucoma treatments rely on medicine and surgery to lower IOP, which only delays disease progression. First-line glaucoma medicines are supported by pharmacotherapy advancements such as Rho kinase inhibitors and innovative drug delivery systems. Glaucoma surgery has shifted to safer minimally invasive (or microinvasive) glaucoma surgery, but further trials are needed to validate long-term efficacy. Further, growing evidence shows that adeno-associated virus gene transduction and stem cell-based RGC replacement therapy hold potential to treat optic nerve fiber degeneration and glaucoma. However, better understanding of the regulatory mechanisms of RGC development is needed to provide insight into RGC differentiation from stem cells and help choose target genes for viral therapy. In this review, we overview current progress in RGC development research, optic nerve fiber regeneration, and human stem cell-derived RGC differentiation and transplantation. We also provide an outlook on perspectives and challenges in the field.
Collapse
Affiliation(s)
- Jung Lo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Kamakshi Mehta
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Armaan Dhillon
- Sue Anschutz-Rodgers Eye Center and Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yu-Kai Huang
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Mi-Hyun Nam
- Sue Anschutz-Rodgers Eye Center and Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Issam Al Diri
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA; Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
16
|
De Sousa RAL, Cassilhas RC. Gestational Diabetes Induced by Low Vitamin B6. Endocrinology 2023; 164:bqad171. [PMID: 37951839 DOI: 10.1210/endocr/bqad171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Physical Education Department, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, 39100-000, Brazil
- Neuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício-GENE), UFVJM, Diamantina, MG, 39100-000, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, 39100-000, Brazil
| | - Ricardo Cardoso Cassilhas
- Physical Education Department, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, 39100-000, Brazil
- Neuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício-GENE), UFVJM, Diamantina, MG, 39100-000, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PMPGCF), UFVJM, Brazilian Society of Physiology, Diamantina, MG, 39100-000, Brazil
- Post Graduation Program in Health Sciences (PPGCS), UFVJM, Diamantina, MG, 39100-000, Brazil
| |
Collapse
|
17
|
Arcuri J, Elbaz A, Sharif NA, Bhattacharya SK. Ocular Treatments Targeting Separate Prostaglandin Receptors in Mice Exhibit Alterations in Intraocular Pressure and Optic Nerve Lipidome. J Ocul Pharmacol Ther 2023; 39:541-550. [PMID: 37267222 PMCID: PMC10616952 DOI: 10.1089/jop.2023.0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 06/04/2023] Open
Abstract
Background: Prostaglandin (PG) receptor agonists are the first-line eyedrop medication treatment for glaucoma. The pathophysiology of this disease is not completely known, and elevated intraocular pressure (IOP) is the key risk factor. The membranes of the axons (of the retinal ganglion cells) passing through the optic nerve (ON) head experience significant damage. Lipids are an essential component of the cell's membranes, and their profile changes owing to neurodegeneration. In this investigation, three agonists for distinct PG receptors were used to lower IOP and to determine their effect on the ON lipids. We utilized DBA/2J mice as a model of progressive IOP increase and C57BL/6J mice as a model of ON crush. Methods: DBA/2J and C57BL/6J mice were treated daily for 2 weeks with Latanoprost, PF-04217329, or Rivenprost. The IOP was measured every 2 days and pattern electroretinogram was conducted for DBA/2J throughout the study. Lipidomics of ONs were performed for each model and treatment group. Results: Of the tested compounds, Latanoprost and Rivenprost were the most effective agents decreasing IOP in DBA/2J mice. Triglyceride levels increased in the ONs of DBA/2J mouse model, but phosphatidylethanolamine levels underwent highest level changes in the C57BL/6J mouse model when treated with Latanoprost. Conclusions: Topical ocular FP- and EP4-receptor agonists appreciably lowered IOP in the DBA/2J mice representing pigmentary glaucoma. The observed changes in ON lipidomics in the different models of neurodegeneration suggest possible use of such measures in the development of more effective medicines for both IOP reduction and ON protection.
Collapse
Affiliation(s)
- Jennifer Arcuri
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, Florida, USA
- Miami Integrative Metabolomics Research Center, Miami, Florida, USA
- Molecular Cellular Pharmacology Graduate Program, University of Miami, Miami, Florida, USA
| | - Abdelrahman Elbaz
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, Florida, USA
- Miami Integrative Metabolomics Research Center, Miami, Florida, USA
| | - Najam A. Sharif
- Eye-APC Duke-National University of Singapore Medical School, Singapore
- Singapore Eye Research Institute, Singapore
- Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA
- Department of Pharmacy Sciences, Creighton University, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
- Imperial College of Science and Technology, St. Mary's Campus, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- Ophthalmology Innovation Center and Product Development Division, Santen Inc., Emeryville, California, USA
| | - Sanjoy K. Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, Florida, USA
- Miami Integrative Metabolomics Research Center, Miami, Florida, USA
| |
Collapse
|
18
|
Kugler E, Bravo I, Durmishi X, Marcotti S, Beqiri S, Carrington A, Stramer B, Mattar P, MacDonald RB. GliaMorph: a modular image analysis toolkit to quantify Müller glial cell morphology. Development 2023; 150:dev201008. [PMID: 36625162 PMCID: PMC10110500 DOI: 10.1242/dev.201008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Cell morphology is crucial for all cell functions. This is particularly true for glial cells as they rely on complex shape to contact and support neurons. However, methods to quantify complex glial cell shape accurately and reproducibly are lacking. To address this, we developed the image analysis pipeline 'GliaMorph'. GliaMorph is a modular analysis toolkit developed to perform (1) image pre-processing, (2) semi-automatic region-of-interest selection, (3) apicobasal texture analysis, (4) glia segmentation, and (5) cell feature quantification. Müller glia (MG) have a stereotypic shape linked to their maturation and physiological status. Here, we characterized MG on three levels: (1) global image-level, (2) apicobasal texture, and (3) regional apicobasal vertical-to-horizontal alignment. Using GliaMorph, we quantified MG development on a global and single-cell level, showing increased feature elaboration and subcellular morphological rearrangement in the zebrafish retina. As proof of principle, we analysed expression changes in a mouse glaucoma model, identifying subcellular protein localization changes in MG. Together, these data demonstrate that GliaMorph enables an in-depth understanding of MG morphology in the developing and diseased retina.
Collapse
Affiliation(s)
- Elisabeth Kugler
- Institute of Ophthalmology, University College London, 11-43 Bath St, Greater London EC1V 9EL, UK
| | - Isabel Bravo
- Institute of Ophthalmology, University College London, 11-43 Bath St, Greater London EC1V 9EL, UK
| | - Xhuljana Durmishi
- Institute of Ophthalmology, University College London, 11-43 Bath St, Greater London EC1V 9EL, UK
| | - Stefania Marcotti
- Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, London SE1 1UL, UK
| | - Sara Beqiri
- Institute of Ophthalmology, University College London, 11-43 Bath St, Greater London EC1V 9EL, UK
| | - Alicia Carrington
- Institute of Ophthalmology, University College London, 11-43 Bath St, Greater London EC1V 9EL, UK
| | - Brian Stramer
- Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, London SE1 1UL, UK
| | - Pierre Mattar
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Ottawa Hospital Research Institute (OHRI), Ottawa, ON, K1H 8L6, Canada
| | - Ryan B. MacDonald
- Institute of Ophthalmology, University College London, 11-43 Bath St, Greater London EC1V 9EL, UK
| |
Collapse
|
19
|
Loss of Retinogeniculate Synaptic Function in the DBA/2J Mouse Model of Glaucoma. eNeuro 2022; 9:ENEURO.0421-22.2022. [PMID: 36526366 PMCID: PMC9794376 DOI: 10.1523/eneuro.0421-22.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/22/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Retinal ganglion cell (RGC) axons comprise the optic nerve and carry information to the dorsolateral geniculate nucleus (dLGN), which is then relayed to the cortex for conscious vision. Glaucoma is a blinding neurodegenerative disease that commonly results from intraocular pressure (IOP)-associated injury leading to RGC axonal pathology, disruption of RGC outputs to the brain, and eventual apoptotic loss of RGC somata. The consequences of elevated IOP and glaucomatous pathology on RGC signaling to the dLGN are largely unknown yet are likely to contribute to vision loss. Here, we used anatomic and physiological approaches to study the structure and function of retinogeniculate (RG) synapses in male and female DBA/2J (D2) mice with inherited glaucoma before and after IOP elevation. D2 mice showed progressive loss of anterograde optic tract transport to the dLGN and vGlut2 labeling of RGC axon terminals while patch-clamp measurements of RG synaptic function showed that synaptic transmission was reduced in 9-month and 12-month D2 mice because of the loss of individual RGC axon inputs. TC neuron dendrites had reduced Sholl complexity at 12 months, suggestive of delayed reorganization following reduced synaptic input. There was no detectable change in RGC density in 11- to 12-month D2 retinas, quantified as the number of ganglion cell layer-residing somata immuno-positive for NeuN and immuno-negative for the amacrine marker choline acetyltransferase (ChAT). Thus, observed synaptic defects appear to precede RGC somatic loss. These findings identify glaucoma-associated and IOP-associated deficits in an important subcortical RGC projection target, shedding light on processes linking IOP to vision loss.
Collapse
|
20
|
Van Hook MJ. Influences of Glaucoma on the Structure and Function of Synapses in the Visual System. Antioxid Redox Signal 2022; 37:842-861. [PMID: 35044228 PMCID: PMC9587776 DOI: 10.1089/ars.2021.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/31/2021] [Indexed: 11/12/2022]
Abstract
Significance: Glaucoma is an age-related neurodegenerative disorder of the visual system associated with sensitivity to intraocular pressure (IOP). It is the leading irreversible cause of vision loss worldwide, and vision loss results from damage and dysfunction of the retinal output neurons known as retinal ganglion cells (RGCs). Recent Advances: Elevated IOP and optic nerve injury triggers pruning of RGC dendrites, altered morphology of excitatory inputs from presynaptic bipolar cells, and disrupted RGC synaptic function. Less is known about RGC outputs, although evidence to date indicates that glaucoma is associated with altered mitochondrial and synaptic structure and function in RGC-projection targets in the brain. These early functional changes likely contribute to vision loss and might be a window into early diagnosis and treatment. Critical Issues: Glaucoma affects different RGC populations to varying extents and along distinct time courses. The influence of glaucoma on RGC synaptic function as well as the mechanisms underlying these effects remain to be determined. Since RGCs are an especially energetically demanding population of neurons, altered intracellular axon transport of mitochondria and mitochondrial function might contribute to RGC synaptic dysfunction in the retina and brain as well as RGC vulnerability in glaucoma. Future Directions: The mechanisms underlying differential RGC vulnerability remain to be determined. Moreover, the timing and mechanisms of RGCs synaptic dysfunction and degeneration will provide valuable insight into the disease process in glaucoma. Future work will be able to capitalize on these findings to better design diagnostic and therapeutic approaches to detect disease and prevent vision loss. Antioxid. Redox Signal. 37, 842-861.
Collapse
Affiliation(s)
- Matthew J. Van Hook
- Department of Ophthalmology & Visual Science and Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Cellular & Integrative Physiology, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
21
|
Masuda S, Tanaka S, Shiraki H, Sotomaru Y, Harada K, Hide I, Kiuchi Y, Sakai N. GPR3 expression in retinal ganglion cells contributes to neuron survival and accelerates axonal regeneration after optic nerve crush in mice. Neurobiol Dis 2022; 172:105811. [PMID: 35809764 DOI: 10.1016/j.nbd.2022.105811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
Glaucoma is an optic neuropathy and is currently one of the most common diseases that leads to irreversible blindness. The axonal degeneration that occurs before retinal ganglion neuronal loss is suggested to be involved in the pathogenesis of glaucoma. G protein-coupled receptor 3 (GPR3) belongs to the class A rhodopsin-type GPCR family and is highly expressed in various neurons. GPR3 is unique in its ability to constitutively activate the Gαs protein without a ligand, which elevates the basal intracellular cAMP level. Our earlier reports suggested that GPR3 enhances both neurite outgrowth and neuronal survival. However, the potential role of GPR3 in axonal regeneration after neuronal injury has not been elucidated. Herein, we investigated retinal GPR3 expression and its possible involvement in axonal regeneration after retinal injury in mice. GPR3 was relatively highly expressed in retinal ganglion cells (RGCs). Surprisingly, RGCs in GPR3 knockout mice were vulnerable to neural death during aging without affecting high intraocular pressure (IOP) and under ischemic conditions. Primary cultured neurons from the retina showed that GPR3 expression was correlated with neurite outgrowth and neuronal survival. Evaluation of the effect of GPR3 on axonal regeneration using GPR3 knockout mice revealed that GPR3 in RGCs participates in axonal regeneration after optic nerve crush (ONC) under zymosan stimulation. In addition, regenerating axons were further stimulated when GPR3 was upregulated in RGCs, and the effect was further augmented when combined with zymosan treatment. These results suggest that GPR3 expression in RGCs helps maintain neuronal survival and accelerates axonal regeneration after ONC in mice.
Collapse
Affiliation(s)
- Shun Masuda
- Department of Molecular and Pharmacological Neuroscience, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan; Department of Ophthalmology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | - Hiroko Shiraki
- Department of Molecular and Pharmacological Neuroscience, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yusuke Sotomaru
- Natural Science Center for Basic Research and Development, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| |
Collapse
|
22
|
Noailles A, Kutsyr O, Mayordomo-Febrer A, Lax P, López-Murcia M, Sanz-González SM, Pinazo-Durán MD, Cuenca N. Sodium Hyaluronate-Induced Ocular Hypertension in Rats Damages the Direction-Selective Circuit and Inner/Outer Retinal Plexiform Layers. Invest Ophthalmol Vis Sci 2022; 63:2. [PMID: 35503230 PMCID: PMC9078050 DOI: 10.1167/iovs.63.5.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To assess the changes in retinal morphology in a rat model of chronic glaucoma induced by ocular hypertension. Methods Intraocular pressure (IOP) was surgically increased through weekly injections of sodium hyaluronate (HYA) in the anterior eye chamber of the left eye of male Wistar rats, whereas the right eyes were sham operated (salt solution). During the 10-week experimental period, IOP was measured weekly with a rebound tonometer. Retinal cryosections were prepared for histological/immunohistochemical analysis and morphometry. Results IOP was higher in HYA-treated eyes than in sham-operated eyes along the 10-week period, which was significant from the fourth to the nineth week. Ocular hypertension in HYA-treated eyes was associated with morphologic and morphometric changes in bipolar cells, ON-OFF direction-selective ganglion cells, ON/OFF starburst amacrine cells, and inner plexiform layer sublamina. Conclusions Serial HYA treatment in the rat anterior eye chamber results in mild-to-moderate elevated and sustained IOP and ganglion cell death, which mimics most human open-angle glaucoma hallmarks. The reduced number of direction-selective ganglion cells and starburst amacrine cells accompanied by a deteriorated ON/OFF plexus in this glaucoma model could lend insight to the abnormalities in motion perception observed in patients with glaucoma.
Collapse
Affiliation(s)
- Agustina Noailles
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| | - Oksana Kutsyr
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| | - Aloma Mayordomo-Febrer
- Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universidad CEU Cardenal Herrera, Valencia, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain.,Mixed Research Unit for Visual Health and Veterinary Ophthalmology CEU/FISABIO, Valencia, Spain
| | - Pedro Lax
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| | - María López-Murcia
- Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universidad CEU Cardenal Herrera, Valencia, Spain.,Mixed Research Unit for Visual Health and Veterinary Ophthalmology CEU/FISABIO, Valencia, Spain
| | - Silvia M Sanz-González
- OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain.,Cellular and Molecular Ophthalmo-biology Research Group, Department of Surgery, University of Valencia, Valencia, Spain.,Ophthalmic Research Unit "Santiago Grisolía"/FISABIO, Valencia, Spain
| | - María Dolores Pinazo-Durán
- OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain.,Cellular and Molecular Ophthalmo-biology Research Group, Department of Surgery, University of Valencia, Valencia, Spain.,Ophthalmic Research Unit "Santiago Grisolía"/FISABIO, Valencia, Spain
| | - Nicolás Cuenca
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| |
Collapse
|
23
|
Dias MS, Luo X, Ribas VT, Petrs-Silva H, Koch JC. The Role of Axonal Transport in Glaucoma. Int J Mol Sci 2022; 23:ijms23073935. [PMID: 35409291 PMCID: PMC8999615 DOI: 10.3390/ijms23073935] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a neurodegenerative disease that affects the retinal ganglion cells (RGCs) and leads to progressive vision loss. The first pathological signs can be seen at the optic nerve head (ONH), the structure where RGC axons leave the retina to compose the optic nerve. Besides damage of the axonal cytoskeleton, axonal transport deficits at the ONH have been described as an important feature of glaucoma. Axonal transport is essential for proper neuronal function, including transport of organelles, synaptic components, vesicles, and neurotrophic factors. Impairment of axonal transport has been related to several neurodegenerative conditions. Studies on axonal transport in glaucoma include analysis in different animal models and in humans, and indicate that its failure happens mainly in the ONH and early in disease progression, preceding axonal and somal degeneration. Thus, a better understanding of the role of axonal transport in glaucoma is not only pivotal to decipher disease mechanisms but could also enable early therapies that might prevent irreversible neuronal damage at an early time point. In this review we present the current evidence of axonal transport impairment in glaucomatous neurodegeneration and summarize the methods employed to evaluate transport in this disease.
Collapse
Affiliation(s)
- Mariana Santana Dias
- Intermediate Laboratory of Gene Therapy and Viral Vectors, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.S.D.); (H.P.-S.)
| | - Xiaoyue Luo
- Department of Neurology, University Medical Center Göttingen, 37077 Göttingen, Germany;
| | - Vinicius Toledo Ribas
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Hilda Petrs-Silva
- Intermediate Laboratory of Gene Therapy and Viral Vectors, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.S.D.); (H.P.-S.)
| | - Jan Christoph Koch
- Department of Neurology, University Medical Center Göttingen, 37077 Göttingen, Germany;
- Correspondence:
| |
Collapse
|
24
|
Bierlein ER, Smith JC, Van Hook MJ. Mechanism for altered dark-adapted electroretinogram responses in DBA/2J mice includes pupil dilation deficits. Curr Eye Res 2022; 47:897-907. [PMID: 35179406 DOI: 10.1080/02713683.2022.2044055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE The DBA/2J (D2) mouse is an established model of pigmentary glaucoma, a type of primary open angle glaucoma. Prior studies have documented defects in flash electroretinogram (ERG) responses in D2 mice, but the origin of those defects is not clear. The purpose of this study was to understand the origin of these A-wave and B-wave changes in D2 ERGs.Materials & Methods: To accomplish this, we analyzed the differences between 9-month-old DBA/2J-Gpnmb+ (D2-control) and D2 mouse eyes in relation to ERG responses, intraocular pressure (IOP), outer nuclear layer thickness, and pupil area. RESULTS D2 scotopic ERGs showed lower A-wave amplitude and longer implicit time as well as a significant rightward shift in the intensity-response curve. D2 IOP increased at approximately seven months of age and had a weak correlation with the ERG A-wave sensitivity. Outer nuclear layer thickness was not significantly different in D2s compared to D2-control retinas. D2 mouse pupils also showed abnormal pupillary shape and no dilation following treatment with tropicamide eye drops. The pupil size moderately correlated with the A-wave sensitivity and this was pharmacologically replicated in C57Bl/6J mice following administration of pilocarpine to constrict the pupils. However, pilocarpine treatment did not affect ERG amplitudes. CONCLUSIONS These data suggest that the smaller pupil sizes prevented light from reaching the photoreceptors and thus contributed to reduced ERG sensitivity in D2 mice. The reduced ERG A-wave amplitude in D2 mice likely results from dysfunctional photoreceptor responses.
Collapse
Affiliation(s)
- Elizabeth R Bierlein
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jennie C Smith
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
25
|
Zeng H, Dumitrescu AV, Wadkins D, Elwood BW, Gramlich OW, Kuehn MH. Systemic Treatment with Pioglitazone Reverses Vision Loss in Preclinical Glaucoma Models. Biomolecules 2022; 12:281. [PMID: 35204782 PMCID: PMC8961625 DOI: 10.3390/biom12020281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroinflammation significantly contributes to the pathophysiology of several neurodegenerative diseases. This is also the case in glaucoma and may be a reason why many patients suffer from progressive vision loss despite maximal reduction in intraocular pressure. Pioglitazone is an agonist of the peroxisome proliferator-activated receptor gamma (PPARγ) whose pleiotrophic activities include modulation of cellular energy metabolism and reduction in inflammation. In this study we employed the DBA2/J mouse model of glaucoma with chronically elevated intraocular pressure to investigate whether oral low-dose pioglitazone treatment preserves retinal ganglion cell (RGC) survival. We then used an inducible glaucoma model in C57BL/6J mice to determine visual function, pattern electroretinographs, and tracking of optokinetic reflex. Our findings demonstrate that pioglitazone treatment does significantly protect RGCs and prevents axonal degeneration in the glaucomatous retina. Furthermore, treatment preserves and partially reverses vision loss in spite of continuously elevated intraocular pressure. These data suggest that pioglitazone may provide treatment benefits for those glaucoma patients experiencing continued vision loss.
Collapse
Affiliation(s)
- Huilan Zeng
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
- Human Clinical Research Center of Ophthalmic Disease, Changsha 410011, China
| | - Alina V. Dumitrescu
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
| | - David Wadkins
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| | - Benjamin W. Elwood
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| | - Oliver W. Gramlich
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| | - Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| |
Collapse
|
26
|
Rohowetz LJ, Mardelli ME, Duncan RS, Riordan SM, Koulen P. The Contribution of Anterior Segment Abnormalities to Changes in Intraocular Pressure in the DBA/2J Mouse Model of Glaucoma: DBA/2J-Gpnmb+/SjJ Mice as Critical Controls. Front Neurosci 2022; 15:801184. [PMID: 35185449 PMCID: PMC8850401 DOI: 10.3389/fnins.2021.801184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
The contributions of anterior segment abnormalities to the development of ocular hypertension was determined in the DBA/2J mouse model of glaucoma. Intraocular pressure (IOP) was measured non-invasively. Iris pigment dispersion (IPD) and corneal calcification were measured weekly starting at 20 weeks of age in DBA/2J and DBA/2J-Gpnmb+/SjJ mice. Thickness, surface area, auto-fluorescence intensity, and perimeter length of calcified regions were measured in postmortem corneas using confocal microscopy. DBA/2J mice developed elevated IOP between 9 and 12 months of age, but DBA/2J-Gpnmb+/SjJ mice did not. Corneal calcification was found at all ages observed and at similar frequencies in both strains with 83.3% of DBA/2J eyes and 60.0% of DBA/2J-Gpnmb+/SjJ eyes affected at 12 months (P = 0.11). Calcification increased with age in both DBA/2J (P = 0.049) and DBA/2J-Gpnmb+/SjJ mice (P = 0.04) when assessed qualitatively and based on mixed-effects analysis. No differences in the four objective measures of calcification were observed between strains or ages. At 12 months of age, DBA/2J mice with corneal calcification had greater mean IOP than DBA/2J mice without corneal calcification. IOP was not correlated with the qualitatively assessed measures of calcification. For the subset of eyes with ocular hypertension, which were only found in DBA/2J mice, IOP was negatively correlated with the qualitative degree of calcification, but was not correlated with the four quantitative measures of calcification. Differences in IOP were not observed between DBA/2J-Gpnmb+/SjJ mice with and without calcification at any age. IPD increased with age and demonstrated a moderate correlation with IOP in DBA/2J mice, but was not observed in DBA/2J-Gpnmb+/SjJ mice. In the DBA/2J mouse model of glaucoma, increased IPD is positively correlated with an increase in IOP and corneal calcification is present in the majority of eyes at and after age 9 months. However, while IPD causes ocular hypertension, corneal calcification does not appear to contribute to the elevation of IOP, as the control strain DBA/2J-Gpnmb+/SjJ exhibits corneal calcification similar to DBA/2J mice, but does not develop ocular hypertension. Corneal calcification, therefore, does not appear to be a contributing factor to the development of elevated IOP in DBA/2J mice.
Collapse
Affiliation(s)
- Landon J. Rohowetz
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Marc E. Mardelli
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
| | - R. Scott Duncan
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Sean M. Riordan
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Peter Koulen
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri—Kansas City, Kansas City, MO, United States
- *Correspondence: Peter Koulen,
| |
Collapse
|
27
|
van Mechelen RJS, Wolters JE, Bertens CJF, Webers CAB, van den Biggelaar FJHM, Gorgels TGMF, Beckers HJM. Animal models and drug candidates for use in glaucoma filtration surgery: A systematic review. Exp Eye Res 2022; 217:108972. [PMID: 35114212 DOI: 10.1016/j.exer.2022.108972] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/14/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
Glaucoma, a degenerative disease of the optic nerve, is the leading cause of irreversible blindness worldwide. Currently, there is no curative treatment. The only proven treatment is lowering intraocular pressure (IOP), the most important risk factor. Glaucoma filtration surgery (GFS) can effectively lower IOP. However, approximately 10% of all surgeries fail yearly due to excessive wound healing, leading to fibrosis. GFS animal models are commonly used for the development of novel treatment modalities. The aim of the present review was to provide an overview of available animal models and anti-fibrotic drug candidates. MEDLINE and Embase were systematically searched. Manuscripts until September 1st, 2021 were included. Studies that used animal models of GFS were included in this review. Additionally, the snowball method was used to identify other publications which had not been identified through the systematic search. Two hundred articles were included in this manuscript. Small rodents (e.g. mice and rats) are often used to study the fibrotic response after GFS and to test drug candidates. Due to their larger eyes, rabbits are better suited to develop medical devices. Novel drugs aim to inhibit specific pathways, e.g. through the use of modulators, monoclonal antibodies, aqueous suppressants or gene therapy. Although most newly studied drugs offer a higher safety profile compared to antimetabolites, their efficacy is in most cases lower when compared to MMC. Current literature on animal models and potential drug candidates for GFS were summarized in this review. Future research should focus on refining current animal models (for example through the induction of glaucoma prior to undertaking GFS) and standardizing animal research to ensure a higher reproducibility and reliability across different research groups. Lastly, novel therapies need to be further optimized, e.g. by conducting more research on the dosage, administration route, application frequency, the option of creating combination therapies, or the development of drug delivery systems for sustained release of anti-fibrotic medication.
Collapse
Affiliation(s)
- Ralph J S van Mechelen
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands; School for Mental Health and Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands; Chemelot Institute for Science and Technology (InSciTe), 6229 GS, Maastricht, the Netherlands.
| | - Jarno Ej Wolters
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands; School for Mental Health and Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands; Chemelot Institute for Science and Technology (InSciTe), 6229 GS, Maastricht, the Netherlands
| | - Christian J F Bertens
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands; School for Mental Health and Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands; Chemelot Institute for Science and Technology (InSciTe), 6229 GS, Maastricht, the Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands
| | - Frank J H M van den Biggelaar
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands
| | - Henny J M Beckers
- University Eye Clinic Maastricht, Maastricht University Medical Center+ (MUMC+), 6202 AZ, Maastricht, the Netherlands
| |
Collapse
|
28
|
Di Pierdomenico J, Henderson DCM, Giammaria S, Smith VL, Jamet AJ, Smith CA, Hooper ML, Chauhan BC. Age and intraocular pressure in murine experimental glaucoma. Prog Retin Eye Res 2021; 88:101021. [PMID: 34801667 DOI: 10.1016/j.preteyeres.2021.101021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
Age and intraocular pressure (IOP) are the two most important risk factors for the development and progression of open-angle glaucoma. While IOP is commonly considered in models of experimental glaucoma (EG), most studies use juvenile or adult animals and seldom older animals which are representative of the human disease. This paper provides a concise review of how retinal ganglion cell (RGC) loss, the hallmark of glaucoma, can be evaluated in EG with a special emphasis on serial in vivo imaging, a parallel approach used in clinical practice. It appraises the suitability of EG models for the purpose of in vivo imaging and argues for the use of models that provide a sustained elevation of IOP, without compromise of the ocular media. In a study with parallel cohorts of adult (3-month-old, equivalent to 20 human years) and old (2-year-old, equivalent to 70 human years) mice, we compare the effects of elevated IOP on serial ganglion cell complex thickness and individual RGC dendritic morphology changes obtained in vivo. We also evaluate how age modulates the impact of elevated IOP on RGC somal and axonal density in histological analysis as well the density of melanopsin RGCs. We discuss the challenges of using old animals and emphasize the potential of single RGC imaging for understanding the pathobiology of RGC loss and evaluating new therapeutic avenues.
Collapse
Affiliation(s)
- Johnny Di Pierdomenico
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Delaney C M Henderson
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sara Giammaria
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Victoria L Smith
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Aliénor J Jamet
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Corey A Smith
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michele L Hooper
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Balwantray C Chauhan
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
29
|
Kusano Y, Yamaguchi T, Nishisako S, Matsumura T, Fukui M, Higa K, Inoue T, Shimazaki J. Elevated Cytokine Levels in Aqueous Humor Are Associated with Peripheral Anterior Synechiae after Penetrating Keratoplasty. Int J Mol Sci 2021; 22:12268. [PMID: 34830147 PMCID: PMC8618311 DOI: 10.3390/ijms222212268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/06/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Peripheral anterior synechiae (PAS) after corneal transplantation leads to refractory glaucoma and permanent loss of vision. However, the exact mechanism remains elusive. This study aimed to evaluate the association between cytokine levels in the aqueous humor (AqH) and the progression of PAS after penetrating keratoplasty (PKP). We measured 20 cytokine levels in AqH and assessed the correlation with PAS progression after PKP in 85 consecutive patients who underwent PKP. We also evaluated age-dependent alterations in PAS and cytokine levels in DBA2J mice. PAS developed in 38 (44.7%) of 85 eyes after PKP. The incidence of intraocular pressure increase after PKP was significantly greater in eyes with PAS (26.3%) than in those without PAS (2%, p = 0.0009). The PAS area at 12 months after PKP was significantly positively correlated with the preoperative levels of interleukin (IL)-6, interferon (IFN)-γ and monocyte chemotactic protein (MCP)-1 (p ≤ 0.049). In the DBA2J mice, an experimental glaucoma model that developed PAS at 50 weeks, the AqH levels of IL-2, IL-6, IL-10, IFN-γ, tumor necrosis factor-α, MCP-1 and granulocyte-macrophage colony-stimulating factor (GM-CSF) significantly increased at 50 weeks compared to 8 weeks (p ≤ 0.021). In conclusion, inflammatory alterations in the AqH microenvironment, such as high preoperative specific cytokine levels, can lead to PAS formation and glaucoma.
Collapse
Affiliation(s)
- Yuki Kusano
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
- Department of Ophthalmology, Kumamoto University, Kumamoto 8608-556, Japan;
| | - Takefumi Yamaguchi
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
| | - Sota Nishisako
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
- Cornea Center Eye Bank, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan
| | - Takehiro Matsumura
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
| | - Masaki Fukui
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
| | - Kazunari Higa
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
- Cornea Center Eye Bank, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan
| | - Toshihiro Inoue
- Department of Ophthalmology, Kumamoto University, Kumamoto 8608-556, Japan;
| | - Jun Shimazaki
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan; (Y.K.); (S.N.); (T.M.); (M.F.); (K.H.); (J.S.)
- Cornea Center Eye Bank, Ichikawa General Hospital, Tokyo Dental College, Chiba 2728-513, Japan
| |
Collapse
|
30
|
Roddy GW, Chowdhury UR, Monson KJ, Fautsch MP. Stanniocalcin-1 Reduced Intraocular Pressure in Two Models of Ocular Hypertension. Curr Eye Res 2021; 46:1525-1530. [PMID: 33757401 PMCID: PMC10505966 DOI: 10.1080/02713683.2021.1899246] [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: 09/23/2020] [Revised: 01/11/2021] [Accepted: 02/23/2021] [Indexed: 10/21/2022]
Abstract
Purpose/Aim: Glaucomatous optic neuropathy (GON) remains the world's leading cause of irreversible blindness. Treatments including topical medications are directed at reducing intraocular pressure (IOP), the most significant risk factor for GON. Current medications, while generally effective, are limited by insufficient response and side-effects in some patients. In search of a more targeted therapy that acts downstream of existing medications that has a potential for a lower side effect profile, our laboratory has identified Stanniocalcin-1 (STC-1), a multifunctional hormone, as an effector molecule in latanoprost-mediated IOP reduction with similar IOP-lowering efficacy as latanoprost in normotensive mice.Materials and methods: To investigate whether STC-1 can also reduce IOP in ocular hypertensive mice, we used a steroid-induced ocular hypertensive mouse model characterized by trabecular meshwork dysfunction as well as the DBA/2J mouse as an inherited model of pigment dispersion and secondary angle closure. Steroid-induced ocular hypertension was induced by weekly injections of dexamethasone into the conjunctival fornix of wild-type C57BL/6J mice (6-8 months old). After confirmation of the steroid response, mice were administered STC-1 or phosphate buffered saline (PBS) topically once daily for six weeks. For DBA/2J mice (14 months old), after baseline IOP measurements, mice were treated topically once daily with STC-1 or PBS for 5 days and IOP was assessed twice daily.Results: In steroid-induced ocular hypertensive mice, STC-1 lowered IOP by 26% (P < .001, week three) and maintained this level of IOP reduction throughout the remainder of the treatment period (P < .001, week six). In DBA/2J mice, STC-1 lowered IOP by 37% (P < .001).Conclusions: Together, these data show that STC-1 reduced IOP in two models of ocular hypertension with different mechanisms of outflow obstruction.
Collapse
Affiliation(s)
- Gavin W Roddy
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
31
|
Abyadeh M, Gupta V, Paulo JA, Gupta V, Chitranshi N, Godinez A, Saks D, Hasan M, Amirkhani A, McKay M, Salekdeh GH, Haynes PA, Graham SL, Mirzaei M. A Proteomic View of Cellular and Molecular Effects of Cannabis. Biomolecules 2021; 11:1411. [PMID: 34680044 PMCID: PMC8533448 DOI: 10.3390/biom11101411] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/17/2022] Open
Abstract
Cannabis (Cannabis sativa), popularly known as marijuana, is the most commonly used psychoactive substance and is considered illicit in most countries worldwide. However, a growing body of research has provided evidence of the therapeutic properties of chemical components of cannabis known as cannabinoids against several diseases including Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease, schizophrenia and glaucoma; these have prompted changes in medicinal cannabis legislation. The relaxation of legal restrictions and increased socio-cultural acceptance has led to its increase in both medicinal and recreational usage. Several biochemically active components of cannabis have a range of effects on the biological system. There is an urgent need for more research to better understand the molecular and biochemical effects of cannabis at a cellular level, to understand fully its implications as a pharmaceutical drug. Proteomics technology is an efficient tool to rigorously elucidate the mechanistic effects of cannabis on the human body in a cell and tissue-specific manner, drawing conclusions associated with its toxicity as well as therapeutic benefits, safety and efficacy profiles. This review provides a comprehensive overview of both in vitro and in vivo proteomic studies involving the cellular and molecular effects of cannabis and cannabis-derived compounds.
Collapse
Affiliation(s)
- Morteza Abyadeh
- ProGene Technologies Pty Ltd., Macquarie Park, Sydney, NSW 2113, Australia;
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia; (N.C.); (A.G.); (D.S.); (S.L.G.)
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA;
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC 2600, Australia;
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia; (N.C.); (A.G.); (D.S.); (S.L.G.)
| | - Angela Godinez
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia; (N.C.); (A.G.); (D.S.); (S.L.G.)
| | - Danit Saks
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia; (N.C.); (A.G.); (D.S.); (S.L.G.)
| | - Mafruha Hasan
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia;
| | - Ardeshir Amirkhani
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia;
| | - Matthew McKay
- Bowel Cancer and Biomarker Laboratory, Kolling Institute, Northern Clinical School, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Ghasem H. Salekdeh
- Department of Molecular Sciences, Macquarie University, Macquarie Park, Sydney, NSW 2109, Australia; (G.H.S.); (P.A.H.)
| | - Paul A. Haynes
- Department of Molecular Sciences, Macquarie University, Macquarie Park, Sydney, NSW 2109, Australia; (G.H.S.); (P.A.H.)
| | - Stuart L. Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia; (N.C.); (A.G.); (D.S.); (S.L.G.)
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia; (N.C.); (A.G.); (D.S.); (S.L.G.)
| |
Collapse
|
32
|
Taiyab A, Saraco A, Akula M, Deschamps P, Ball AK, Williams T, West-Mays JA. Progressive Loss of Retinal Ganglion Cells in Activating Protein-2β Neural Crest Cell Knockout Mice. Curr Eye Res 2021; 46:1509-1515. [PMID: 33689532 DOI: 10.1080/02713683.2021.1901939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Purpose: Our lab has shown that conditionally disrupting the transcription factor activating protein 2β (Tfap2b) gene, responsible for the activating protein-2β (AP-2β) transcription factor, exclusively in cranial neural crest cells (AP-2β NCC KO), leads to anterior segment dysgenesis and a closed angle phenotype. The purpose of the current study is to determine if there is a progressive loss of retinal ganglion cells (RGCs) in the mutant over time and whether this loss was associated with macroglial activity changes and elevated intraocular pressure (IOP).Methods: Using the Cre-loxP system, we generated a conditional knockout of Tfap2b exclusively in cranial NCC (AP-2β NCC KO). Immunohistochemistry was performed using anti-Brn3a, anti-GFAP and anti-Vimentin antibodies. IOP was measured using a tonometer and the data was analyzed using GraphPad Prism software. Brn3a and DAPI positive cells were counted using Image-J and statistical analysis was performed with GraphPad Prism software.Results: Our findings revealed that while no statistical difference in Brn3a expression was observed between wild-type and mutant mice at postnatal day (P) 4 or P10, at P40 (p < .01) and P42 (p < .0001) Brn3a expression was significantly reduced in the mutant retina at the region of the ONH. There was also increased expression of glial fibrillary acidic protein (GFAP) by Müller cells in the AP-2β NCC KO mice at P35 and P40, indicating the presence of neuroinflammation. Moreover, increased IOP was observed starting at P35 and continuing at P40 and P42 (p < .0001 for all three ages examined).Conclusions: Together, these findings suggest that the retinal damage observed in the KO mouse becomes apparent by P40 after increased IOP was observed at P35 and progressed over time. The AP-2β NCC KO mouse may therefore be a novel experimental model for glaucoma.
Collapse
Affiliation(s)
- Aftab Taiyab
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Anthony Saraco
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Monica Akula
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Paula Deschamps
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Alexander K Ball
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Trevor Williams
- Craniofacial Biology, University of Colorado, Aurora, Colorado, USA
| | | |
Collapse
|
33
|
Chen J, Sun J, Yu H, Huang P, Zhong Y. Evaluation of the Effectiveness of a Chronic Ocular Hypertension Mouse Model Induced by Intracameral Injection of Cross-Linking Hydrogel. Front Med (Lausanne) 2021; 8:643402. [PMID: 33829024 PMCID: PMC8019751 DOI: 10.3389/fmed.2021.643402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Glaucoma is an irreversible and blinding neurodegenerative disease that is characterized by progressive loss of retinal ganglion cells. The current animal models of glaucoma fail to provide a chronic elevated intraocular pressure and cannot maintain the optical media clarity for a long time, which brings some difficulties to the study of glaucoma. Here, we developed a new chronic ocular hypertension model of mice induced by cross-linking hydrogel intracameral injection. Methods: C57BL/6J mice aged 6–8 weeks were randomly divided into the control group and the operation group. The mice of the operation group were injected with cross-linking hydrogel to induce ocular hypertension. Intraocular pressure was measured preoperatively, 3 days after surgery, and weekly until the end of the study. Flash visual evoked potential (F-VEP) was used to observe optic nerve function at different times (preoperatively and 2, 4, and 6 weeks) after chronic ocular hypertension (COH). Retinal TNF-α, IL-1β, and IL-17A protein expression were measured by western blotting in the control group and in mice at 2, 4, and 6 weeks after COH. Microglial cell activation was evaluated by immunofluorescence staining and western blotting. Apoptosis and loss of retinal ganglion cells after 2, 4, and 6 weeks of intracameral injection of cross-linking hydrogel were observed by the TUNEL assay and Brn3a protein labeling. The loss of optic nerve axons in COH mice was evaluated by neurofilament heavy polypeptide protein labeling. Results: Intracameral injection of the cross-linking hydrogel induces increased intraocular pressure (IOP) to a mean value of 19.3 ± 4.1 mmHg, which was sustained for at least 8 weeks. A significant difference in IOP was noted between COH mice and sham-operation mice (p < 0.0001). The success rate was 75%. The average amplitude of F-VEP in mice with COH was reduced (p = 0.0149, 0.0012, and 0.0009 at 2, 4, and 6 weeks after COH vs. the control group, respectively), and the average latent period in mice with COH was longer (p = 0.0290, <0.0001, and <0.0001 at 2, 4, and 6 weeks after COH vs. the control group, respectively) compared with that in the control group. TNF-α, IL-1β, IL-17A, Iba-1, and CD68 protein expression increased in COH mice. During the processing of COH, the number of microglial cells increased along with cellular morphological changes of rounder bodies and thicker processes compared with the control group. Apoptosis of retinal ganglion cells (RGCs) was clearly observed in mice at 2, 4, and 6 weeks after COH (p = 0.0061, 0.0012, <0.0001, and 0.0371 at 2, 4, and 6 weeks after COH vs. the control group, respectively). The RGC density decreased significantly in the COH mice compared with the control group (p = 0.0042, 0.0036, and <0.0001 at 2, 4, and 6 weeks after COH vs. the control group, respectively). There was a significant loss of optic nerve axons in mice after intracameral injection of cross-linking hydrogel (p = 0.0095, 0.0002, and <0.0001 at 2, 4, and 6 weeks after COH vs. the control group, respectively). Conclusions: A single intracameral injection of cross-linking hydrogel can effectively induce chronic ocular hypertension in mice, which causes progressive loss of retinal ganglion cells, increased expression levels of inflammatory cytokines and microglial cell activation, and deterioration of optic nerve function.
Collapse
Affiliation(s)
- Junjue Chen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Sun
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Yu
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Huang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, Shanghai, China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Ocular phenotypes in a mouse model of impaired glucocerebrosidase activity. Sci Rep 2021; 11:6079. [PMID: 33727605 PMCID: PMC7971029 DOI: 10.1038/s41598-021-85528-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/02/2021] [Indexed: 01/08/2023] Open
Abstract
Mutations in the GBA1 gene encoding glucocerebrosidase (GCase) are linked to Gaucher (GD) and Parkinson's Disease (PD). Since some GD and PD patients develop ocular phenotypes, we determined whether ocular phenotypes might result from impaired GCase activity and the corresponding accumulation of glucosylceramide (GluCer) and glucosylsphingosine (GluSph) in the Gba1D409V/D409V knock-in (Gba KI/KI; "KI") mouse. Gba KI mice developed age-dependent pupil dilation deficits to an anti-muscarinic agent; histologically, the iris covered the anterior part of the lens with adhesions between the iris and the anterior surface of the lens (posterior synechia). This may prevent pupil dilation in general, beyond an un-responsiveness of the iris to anti-muscarinics. Gba KI mice displayed atrophy and pigment dispersion of the iris, and occlusion of the iridocorneal angle by pigment-laden cells, reminiscent of secondary open angle glaucoma. Gba KI mice showed progressive thinning of the retina consistent with retinal degeneration. GluSph levels were increased in the anterior and posterior segments of the eye, suggesting that accumulation of lipids in the eye may contribute to degeneration in this compartment. We conclude that the Gba KI model provides robust and reproducible eye phenotypes which may be used to test for efficacy and establish biomarkers for GBA1-related therapies.
Collapse
|
35
|
Schnichels S, Hurst J, de Vries JW, Ullah S, Frößl K, Gruszka A, Löscher M, Bartz-Schmidt KU, Spitzer MS, Herrmann A. Improved Treatment Options for Glaucoma with Brimonidine-Loaded Lipid DNA Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9445-9456. [PMID: 33528240 DOI: 10.1021/acsami.0c18626] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Glaucoma is the second leading cause of irreversible blindness worldwide. Among others, elevated intraocular pressure (IOP) is one of the hallmarks of the disease. Antiglaucoma drugs such as brimonidine can lower the IOP but their adherence to the ocular surface is low, leading to a low drug uptake. This results in a frequent dropping regime causing low compliance by the patients. Lipid DNA nanoparticles (NPs) have the intrinsic ability to bind to the ocular surface and can be loaded with different drugs. Here, we report DNA NPs functionalized for loading of brimonidine through specific aptamers and via hydrophobic interactions with double stranded micelles. Both NP systems exhibited improved affinity toward the cornea and retained release of the drug as compared to controls both in vitro and in vivo. Both NP types were able to lower the IOP in living animals significantly more than pristine brimonidine. Importantly, the brimonidine-loaded NPs showed no toxicity and improved efficacy and hence should improve compliance. In conclusion, this drug-delivery system offers high chances of an improved treatment for glaucoma and thus preserving vision in the aging population.
Collapse
Affiliation(s)
- Sven Schnichels
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - José Hurst
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Jan Willem de Vries
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Sami Ullah
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Katharina Frößl
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Agnieszka Gruszka
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Marina Löscher
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Karl-Ulrich Bartz-Schmidt
- Centre for Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, Tübingen D-72076, Germany
| | - Martin S Spitzer
- Clinic for Ophthalmology, University Medical Center Hamburg-Eppendorf (UKE), Martinistrasse 52, Hamburg D-20246, Germany
| | - Andreas Herrmann
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstrasse 50, Aachen 52056, Germany
- Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, Aachen 52074, Germany
| |
Collapse
|
36
|
Eastlake K, Jayaram H, Luis J, Hayes M, Khaw PT, Limb GA. Strain Specific Responses in a Microbead Rat Model of Experimental Glaucoma. Curr Eye Res 2021; 46:387-397. [PMID: 32842792 PMCID: PMC8025805 DOI: 10.1080/02713683.2020.1805472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/13/2020] [Accepted: 07/23/2020] [Indexed: 11/15/2022]
Abstract
PURPOSE A major challenge in glaucoma research is the lack of reproducible animal models of RGC and optic nerve damage, the characteristic features of this condition. We therefore examined the glaucomatous responses of two different rat strains, the Brown Norway (BN) and Lister Hooded (LH) rats, to high intraocular pressure (IOP) induced by injection of magnetic beads into the anterior chamber. METHODS Magnetic microsphere suspensions (20 µl of 5-20 mg/ml) were injected into the anterior chamber of BN (n = 9) or LH (N = 15) rats. Animals from each strain were divided into three groups, each receiving a different dose of microspheres. IOP was measured over 4 weeks using a rebound tonometer. Retinal ganglion cell (RGC) damage and function were assessed using scotopic electroretinograms (ERGs), retinal flatmounts and optic nerve histology. ANOVA and Student's t-tests were used to analyse the data. RESULTS A significant elevation in IOP was observed in BN rats receiving injections of 20 mg (37.18 ± 12.28 mmHg) or 10 mg microspheres/ml (36.95 ± 13.63 mmHg) when compared with controls (19.63 ± 4.29 mmHg) (p < .001) over 2 weeks. This correlated with a significant impairment of RGC function, as determined by scotopic ERG (p < .001), reduction in axon number (p < .05) and lower RGC density (P < .05) in animals receiving 20 mg or 10 mg microspheres/ml as compared with controls. LH rats receiving similar microsphere doses showed reduced scotopic ERG function (p < .001) after 2 weeks. No changes in IOP was seen in this strain, although a reduction in axon density was observed in optic nerve cross-sections (p < .05). Initial changes in IOP and ERG responses observed in BN rats remained unchanged for a duration of 7 weeks. In LH animals, ERG responses were decreased at 1-2 weeks and returned to control levels after 5 weeks. CONCLUSIONS Although this model was easily reproducible in BN rats, the phenotype of injury observed in LH rats was very different from that observed in BN animals. We suggest that differences in the glaucomatous response observed in these two strains may be ascribed to anatomical and physiological differences and merits further investigation.
Collapse
Affiliation(s)
- Karen Eastlake
- NIHR Biomedical Research Centre at Moorfields, Eye Hospitaland UCL Institute of Ophthalmology, London, UK
| | - Hari Jayaram
- NIHR Biomedical Research Centre at Moorfields, Eye Hospitaland UCL Institute of Ophthalmology, London, UK
| | - Joshua Luis
- NIHR Biomedical Research Centre at Moorfields, Eye Hospitaland UCL Institute of Ophthalmology, London, UK
| | - Matthew Hayes
- NIHR Biomedical Research Centre at Moorfields, Eye Hospitaland UCL Institute of Ophthalmology, London, UK
| | - Peng T. Khaw
- NIHR Biomedical Research Centre at Moorfields, Eye Hospitaland UCL Institute of Ophthalmology, London, UK
| | - G. Astrid Limb
- NIHR Biomedical Research Centre at Moorfields, Eye Hospitaland UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
37
|
Fiedorowicz M, Choragiewicz T, Turski WA, Kocki T, Nowakowska D, Wertejuk K, Kamińska A, Avitabile T, Wełniak-Kaminska M, Grieb P, Zweifel S, Rejdak R, Toro MD. Tryptophan Pathway Abnormalities in a Murine Model of Hereditary Glaucoma. Int J Mol Sci 2021; 22:1039. [PMID: 33494373 PMCID: PMC7865582 DOI: 10.3390/ijms22031039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND It has been shown that a possible pathogenetic mechanism of neurodegeneration in the mouse model of glaucoma (DBA/2J) may be an alteration of kynurenic acid (KYNA) in the retina. This study aimed to verify the hypothesis that alterations of tryptophan (TRP) metabolism in DBA/2J mice is not limited to the retina. METHODS Samples of the retinal tissue and serum were collected from DBA/2J mice (6 and 10 months old) and control C57Bl/6 mice of the same age. The concentration of TRP, KYNA, kynurenine (KYN), and 3-hydroxykynurenine (3OH-K) was measured by HPLC. The activity of indoleamine 2,3-dioxygenase (IDO) was also determined as a KYN/TRP ratio. RESULTS TRP, KYNA, L-KYN, and 3OH-K concentration were significantly lower in the retinas of DBA/2J mice than in C57Bl/6 mice. 3OH-K concentration was higher in older mice in both strains. Serum TRP, L-KYN, and KYNA concentrations were lower in DBA/2J than in age-matched controls. However, serum IDO activity did not differ significantly between compared groups and strains. CONCLUSIONS Alterations of the TRP pathway seem not to be limited to the retina in the murine model of hereditary glaucoma.
Collapse
Affiliation(s)
- Michal Fiedorowicz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.F.); (M.W.-K.); (P.G.)
| | - Tomasz Choragiewicz
- Department of General Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (T.C.); (D.N.); (K.W.); (R.R.)
| | - Waldemar A. Turski
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-079 Lublin, Poland; (W.A.T.); (T.K.)
| | - Tomasz Kocki
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-079 Lublin, Poland; (W.A.T.); (T.K.)
| | - Dominika Nowakowska
- Department of General Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (T.C.); (D.N.); (K.W.); (R.R.)
| | - Kamila Wertejuk
- Department of General Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (T.C.); (D.N.); (K.W.); (R.R.)
| | - Agnieszka Kamińska
- Faculty of Medical Sciences, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-815 Warsaw, Poland;
| | - Teresio Avitabile
- Department of Ophthalmology, University of Catania, 95123 Catania, Italy;
| | - Marlena Wełniak-Kaminska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.F.); (M.W.-K.); (P.G.)
| | - Pawel Grieb
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.F.); (M.W.-K.); (P.G.)
| | - Sandrine Zweifel
- Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (T.C.); (D.N.); (K.W.); (R.R.)
| | - Mario Damiano Toro
- Department of General Ophthalmology, Medical University of Lublin, 20-079 Lublin, Poland; (T.C.); (D.N.); (K.W.); (R.R.)
- Faculty of Medical Sciences, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-815 Warsaw, Poland;
- Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| |
Collapse
|
38
|
Tirendi S, Saccà SC, Vernazza S, Traverso C, Bassi AM, Izzotti A. A 3D Model of Human Trabecular Meshwork for the Research Study of Glaucoma. Front Neurol 2020; 11:591776. [PMID: 33335510 PMCID: PMC7736413 DOI: 10.3389/fneur.2020.591776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Glaucoma is a multifactorial syndrome in which the development of pro-apoptotic signals are the causes for retinal ganglion cell (RGC) loss. Most of the research progress in the glaucoma field have been based on experimentally inducible glaucoma animal models, which provided results about RGC loss after either the crash of the optic nerve or IOP elevation. In addition, there are genetically modified mouse models (DBA/2J), which make the study of hereditary forms of glaucoma possible. However, these approaches have not been able to identify all the molecular mechanisms characterizing glaucoma, possibly due to the disadvantages and limits related to the use of animals. In fact, the results obtained with small animals (i.e., rodents), which are the most commonly used, are often not aligned with human conditions due to their low degree of similarity with the human eye anatomy. Although the results obtained from non-human primates are in line with human conditions, they are little used for the study of glaucoma and its outcomes at cellular level due to their costs and their poor ease of handling. In this regard, according to at least two of the 3Rs principles, there is a need for reliable human-based in vitro models to better clarify the mechanisms involved in disease progression, and possibly to broaden the scope of the results so far obtained with animal models. The proper selection of an in vitro model with a "closer to in vivo" microenvironment and structure, for instance, allows for the identification of the biomarkers involved in the early stages of glaucoma and contributes to the development of new therapeutic approaches. This review summarizes the most recent findings in the glaucoma field through the use of human two- and three-dimensional cultures. In particular, it focuses on the role of the scaffold and the use of bioreactors in preserving the physiological relevance of in vivo conditions of the human trabecular meshwork cells in three-dimensional cultures. Moreover, data from these studies also highlight the pivotal role of oxidative stress in promoting the production of trabecular meshwork-derived pro-apoptotic signals, which are one of the first marks of trabecular meshwork damage. The resulting loss of barrier function, increase of intraocular pressure, as well the promotion of neuroinflammation and neurodegeneration are listed as the main features of glaucoma. Therefore, a better understanding of the first molecular events, which trigger the glaucoma cascade, allows the identification of new targets for an early neuroprotective therapeutic approach.
Collapse
Affiliation(s)
- Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Pisa, Italy
| | - Sergio Claudio Saccà
- Ophthalmology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania Vernazza
- Istituto di Ricovero e Cura a Carattere Scientifico, Fondazione Bietti, Rome, Italy
| | - Carlo Traverso
- Clinica Oculistica, Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno Infantili, University of Genoa and Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Pisa, Italy
| | - Alberto Izzotti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- Mutagenesis Unit, IST National Institute for Cancer Research, Istituto di Ricovero e Cura a Carattere Scientifico San Martino University Hospital, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
39
|
Fiedorowicz M, Wełniak-Kamińska M, Świątkiewicz M, Orzeł J, Chorągiewicz T, Toro MD, Rejdak R, Bogorodzki P, Grieb P. Changes of Ocular Dimensions as a Marker of Disease Progression in a Murine Model of Pigmentary Glaucoma. Front Pharmacol 2020; 11:573238. [PMID: 33013417 PMCID: PMC7500411 DOI: 10.3389/fphar.2020.573238] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose The elevation of intraocular pressure (IOP), a major risk factor in glaucoma, is an important parameter tracked in experimental models of this disease. However, IOP measurement in laboratory rodents is challenging and may not correlate with some key pathological events that occur in the development of glaucoma. The aims of this study were to quantify changes in ocular morphology in DBA/2J mice that develop spontaneous, age-dependent, pigmentary glaucoma and to check the possible correlation of these parameters with IOP. Method Eye morphology was evaluated with MRI in DBA/2J, DBA/2J-Gpnmb+/SjJ, and C57BL/6J female mice ages 3, 6, 9, 12, and 15 months. The animals were anesthetized with isoflurane. A planar receive-only surface coil (inner diameter = 10 mm) was placed over each animal’s left eye and the image was acquired with a 7T small animal-dedicated magnetic resonance tomograph and T2-weighted TurboRARE sequence. Ocular dimensions were manually quantitated using OsiriX software. IOP was measured with rebound tonometry. Results In the control animals, no age-related changes in the ocular morphology were noted. Since 6 months of age, the anterior chamber deepening and elongation of the eyeballs of DBA/2J mice was detectable. We found a significant, positive correlation between IOP and axial length, anterior chamber area, or anterior chamber width in C57BL/6J mice but not in DBA/2J mice. However, after excluding the measurements performed in the oldest DBA/2J mice (i.e. analyzing only the animals ages 3 to 12 months), we demonstrated a significant positive correlation between IOP and anterior chamber width. Conclusion High-resolution magnetic resonance imaging of the eye area in mice enables reproducible and consistent measures of key dimensions of the eyeball. We observed age-dependent alterations in the eye morphology of DBA/2J mice that mostly affected the anterior chamber. We also demonstrated a correlation between some of the ocular dimensions and the IOP of C57Bl/6J mice and DBA/2J mice with moderately advanced glaucomatous pathology.
Collapse
Affiliation(s)
- Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Świątkiewicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Jarosław Orzeł
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Faculty of Electronics and Information Technology, Warsaw University of Technology, Warsaw, Poland
| | - Tomasz Chorągiewicz
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Mario Damiano Toro
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland.,Faculty of Medical Sciences, Collegium Medicum, Cardinal Stefan Wyszyński University, Warsaw, Poland
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Piotr Bogorodzki
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Faculty of Electronics and Information Technology, Warsaw University of Technology, Warsaw, Poland
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
40
|
Schnichels S, Paquet-Durand F, Löscher M, Tsai T, Hurst J, Joachim SC, Klettner A. Retina in a dish: Cell cultures, retinal explants and animal models for common diseases of the retina. Prog Retin Eye Res 2020; 81:100880. [PMID: 32721458 DOI: 10.1016/j.preteyeres.2020.100880] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022]
Abstract
For many retinal diseases, including age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), the exact pathogenesis is still unclear. Moreover, the currently available therapeutic options are often unsatisfactory. Research designed to remedy this situation heavily relies on experimental animals. However, animal models often do not faithfully reproduce human disease and, currently, there is strong pressure from society to reduce animal research. Overall, this creates a need for improved disease models to understand pathologies and develop treatment options that, at the same time, require fewer or no experimental animals. Here, we review recent advances in the field of in vitro and ex vivo models for AMD, glaucoma, and DR. We highlight the difficulties associated with studies on complex diseases, in which both the initial trigger and the ensuing pathomechanisms are unclear, and then delineate which model systems are optimal for disease modelling. To this end, we present a variety of model systems, ranging from primary cell cultures, over organotypic cultures and whole eye cultures, to animal models. Specific advantages and disadvantages of such models are discussed, with a special focus on their relevance to putative in vivo disease mechanisms. In many cases, a replacement of in vivo research will mean that several different in vitro models are used in conjunction, for instance to analyze and validate causative molecular pathways. Finally, we argue that the analytical decomposition into appropriate cell and tissue model systems will allow making significant progress in our understanding of complex retinal diseases and may furthermore advance the treatment testing.
Collapse
Affiliation(s)
- Sven Schnichels
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Germany.
| | - François Paquet-Durand
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Germany
| | - Marina Löscher
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Germany
| | - Teresa Tsai
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Germany
| | - José Hurst
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Germany
| | - Alexa Klettner
- Department of Ophthalmology, University Medical Center, University of Kiel, Kiel, Germany
| |
Collapse
|
41
|
Schnichels S, Hurst J, de Vries JW, Ullah S, Gruszka A, Kwak M, Löscher M, Dammeier S, Bartz-Schmidt KU, Spitzer MS, Herrmann A. Self-assembled DNA nanoparticles loaded with travoprost for glaucoma-treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102260. [PMID: 32629041 DOI: 10.1016/j.nano.2020.102260] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/15/2020] [Accepted: 06/20/2020] [Indexed: 11/29/2022]
Abstract
Lipid DNA nanoparticles (NPs) exhibit an intrinsic affinity to the ocular surface and can be loaded by hybridization with fluorophore-DNA conjugates or with the anti-glaucoma drug travoprost by hybridizing an aptamer that binds the medication. In the travoprost-loaded NPs (Trav-NPs), the drug is bound by specific, non-covalent interactions, not requiring any chemical modification of the active pharmaceutical ingredient. Fluorescently labeled Trav-NPs show a long-lasting adherence to the eye, up to sixty minutes after eye drop instillation. Biosafety of the Trav-NPs was proved and in vivo. Ex vivo and in vivo quantification of travoprost via LC-MS revealed that Trav-NPs deliver at least twice the amount of the drug at every time-point investigated compared to the pristine drug. The data successfully show the applicability of a DNA-based drug delivery system in the field of ophthalmology for the treatment of a major retinal eye disease, i.e. glaucoma.
Collapse
Affiliation(s)
- Sven Schnichels
- Centre for Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany.
| | - José Hurst
- Centre for Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany
| | - Jan Willem de Vries
- Centre for Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany
| | - Sami Ullah
- Centre for Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany
| | - Agnieszka Gruszka
- Centre for Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan, Republic of Korea
| | - Marina Löscher
- Centre for Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany
| | - Sascha Dammeier
- Institute for Ophthalmic Research Tübingen, Tübingen, Germany
| | | | - Martin S Spitzer
- Clinic for Ophthalmology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Andreas Herrmann
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands; DWI - Leibniz Institute for Interactive Materials, Aachen, Germany; Institute for Technical and Macromolecular Chemistry, Aachen, Germany.
| |
Collapse
|
42
|
Thomson BR, Grannonico M, Liu F, Liu M, Mendapara P, Xu Y, Liu X, Quaggin SE. Angiopoietin-1 Knockout Mice as a Genetic Model of Open-Angle Glaucoma. Transl Vis Sci Technol 2020; 9:16. [PMID: 32818103 PMCID: PMC7396191 DOI: 10.1167/tvst.9.4.16] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose A leading cause of blindness worldwide, glaucoma is often caused by elevated intraocular pressure (IOP) due to impaired aqueous humor outflow from the anterior chamber through Schlemm's canal (SC) and the trabecular meshwork. Despite the large clinical burden, glaucoma research and drug development are hindered by a limited selection of preclinical models that accurately recapitulate human disease. Here, we propose that Angpt1 conditional knockout mice may provide one such model. Angiopoietin/TEK (ANGPT/TEK) signaling is crucial for SC formation and integrity in mice and humans, and mice lacking TEK or its ligand ANGPT1 develop a hypomorphic SC insufficient for normal aqueous humor outflow. Methods We used a comprehensive histology and physiology approach to characterize the glaucoma phenotype of Angpt1 inducible knockout mice, especially focusing on retina morphology and function. Results Angpt1 deletion resulted in persistent ocular hypertension beginning in the first month after birth and leading to decreased visual acuity with age due to glaucomatous neuropathy. In the neural retina, we identified marked and specific loss of the retinal ganglion cells, whereas other retinal neurons exhibited largely normal morphology and patterning. Electroretinogram recordings demonstrated reduced scotopic threshold response, further indicating loss of retinal ganglion cell function. Conclusions These findings highlight the potential of Angpt1 conditional knockout mice as a valuable new glaucoma model. Translational Relevance Currently, few reliable, rapid-onset genetic glaucoma models are available, and Angpt1 knockout mice will provide an additional tool for studies of IOP-induced neural damage, mechanisms of disease progression, and novel treatment strategies.
Collapse
Affiliation(s)
- Benjamin R. Thomson
- Feinberg Cardiovascular and Renal Research Institute and Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marta Grannonico
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Feng Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Mingna Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Parrykumar Mendapara
- Feinberg Cardiovascular and Renal Research Institute and Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ying Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Susan E. Quaggin
- Feinberg Cardiovascular and Renal Research Institute and Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
43
|
Lani R, Dias MS, Abreu CA, Araújo VG, Gonçalo T, Nascimento-Dos-Santos G, Dantas AM, Allodi S, Fiorani M, Petrs-Silva H, Linden R. A subacute model of glaucoma based on limbal plexus cautery in pigmented rats. Sci Rep 2019; 9:16286. [PMID: 31705136 PMCID: PMC6841973 DOI: 10.1038/s41598-019-52500-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022] Open
Abstract
Glaucoma is a neurodegenerative disorder characterized by the progressive functional impairment and degeneration of the retinal ganglion cells (RGCs) and their axons, and is the leading cause of irreversible blindness worldwide. Current management of glaucoma is based on reduction of high intraocular pressure (IOP), one of its most consistent risk factors, but the disease proceeds in almost half of the patients despite such treatments. Several experimental models of glaucoma have been developed in rodents, most of which present shortcomings such as high surgical invasiveness, slow learning curves, damage to the transparency of the optic media which prevents adequate functional assessment, and variable results. Here we describe a novel and simple method to induce ocular hypertension in pigmented rats, based on low-temperature cauterization of the whole circumference of the limbal vascular plexus, a major component of aqueous humor drainage and easily accessible for surgical procedures. This simple, low-cost and efficient method produced a reproducible subacute ocular hypertension with full clinical recovery, followed by a steady loss of retinal ganglion cells and optic axons, accompanied by functional changes detected both by electrophysiological and behavioral methods.
Collapse
Affiliation(s)
- Rafael Lani
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Mariana S Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla Andreia Abreu
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor G Araújo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thais Gonçalo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Silvana Allodi
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mario Fiorani
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hilda Petrs-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Rafael Linden
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
44
|
Mead B, Ahmed Z, Tomarev S. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Promote Neuroprotection in a Genetic DBA/2J Mouse Model of Glaucoma. Invest Ophthalmol Vis Sci 2019; 59:5473-5480. [PMID: 30452601 PMCID: PMC6735616 DOI: 10.1167/iovs.18-25310] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose To determine if bone marrow-derived stem cell (BMSC) small extracellular vesicles (sEV) promote retinal ganglion cell (RGC) neuroprotection in the genetic DBA/2J mouse model of glaucoma for 12 months. Methods BMSC sEV and control fibroblast-derived sEV were intravitreally injected into 3-month-old DBA/2J mice once a month for 9 months. IOP and positive scotopic threshold responses were measured from 3 months: IOP was measured monthly and positive scotopic threshold responses were measured every 3 months. RGC neuroprotection was determined in wholemounts stained with RNA binding protein with multiple splicing (RBPMS), whereas axonal damage was assessed using paraphenylenediamine staining. Results As expected, DBA/2J mice developed chronic ocular hypertension beginning at 6 months. The delivery of BMSC sEV, but not fibroblast sEV, provided significant neuroprotective effects for RBPMS+ RGC while significantly reducing the number of degenerating axons seen in the optic nerve. BMSC sEV significantly preserved RGC function in 6-month-old mice, but provided no benefit at 9 and 12 months. Conclusions BMSC sEV are an effective neuroprotective treatment in a chronic model of ocular hypertension for 1 year, preserving RGC numbers and protecting against axonal degeneration.
Collapse
Affiliation(s)
- Ben Mead
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States.,Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stanislav Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
45
|
Matrix-bound nanovesicles prevent ischemia-induced retinal ganglion cell axon degeneration and death and preserve visual function. Sci Rep 2019; 9:3482. [PMID: 30837658 PMCID: PMC6400956 DOI: 10.1038/s41598-019-39861-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/25/2019] [Indexed: 01/07/2023] Open
Abstract
Injury to retinal ganglion cells (RGC), central nervous system neurons that relay visual information to the brain, often leads to RGC axon degeneration and permanently lost visual function. Herein this study shows matrix-bound nanovesicles (MBV), a distinct class of extracellular nanovesicle localized specifically to the extracellular matrix (ECM) of healthy tissues, can neuroprotect RGCs and preserve visual function after severe, intraocular pressure (IOP) induced ischemia in rat. Intravitreal MBV injections attenuated IOP-induced RGC axon degeneration and death, protected RGC axon connectivity to visual nuclei in the brain, and prevented loss in retinal function as shown by histology, anterograde axon tracing, manganese-enhanced magnetic resonance imaging, and electroretinography. In the optic nerve, MBV also prevented IOP-induced decreases in growth associated protein-43 and IOP-induced increases in glial fibrillary acidic protein. In vitro studies showed MBV suppressed pro-inflammatory signaling by activated microglia and astrocytes, stimulated RGC neurite growth, and neuroprotected RGCs from neurotoxic media conditioned by pro-inflammatory astrocytes. Thus, MBV can positively modulate distinct signaling pathways (e.g., inflammation, cell death, and axon growth) in diverse cell types. Since MBV are naturally derived, bioactive factors present in numerous FDA approved devices, MBV may be readily useful, not only experimentally, but also clinically as immunomodulatory, neuroprotective factors for treating trauma or disease in the retina as well as other CNS tissues.
Collapse
|
46
|
Harada C, Kimura A, Guo X, Namekata K, Harada T. Recent advances in genetically modified animal models of glaucoma and their roles in drug repositioning. Br J Ophthalmol 2018; 103:161-166. [PMID: 30366949 PMCID: PMC6362806 DOI: 10.1136/bjophthalmol-2018-312724] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/21/2018] [Accepted: 08/25/2018] [Indexed: 12/18/2022]
Abstract
Glaucoma is one of the leading causes of vision loss in the world. Currently, pharmacological intervention for glaucoma therapy is limited to eye drops that reduce intraocular pressure (IOP). Recent studies have shown that various factors as well as IOP are involved in the pathogenesis of glaucoma, especially in the subtype of normal tension glaucoma. To date, various animal models of glaucoma have been established, including glutamate/aspartate transporter knockout (KO) mice, excitatory amino acid carrier 1 KO mice, optineurin E50K knock-in mice, DBA/2J mice and experimentally induced models. These animal models are very useful for elucidating the pathogenesis of glaucoma and for identifying potential therapeutic targets. However, each model represents only some aspects of glaucoma, never the whole disease. This review will summarise the benefits and limitations of using disease models of glaucoma and recent basic research in retinal protection using existing drugs.
Collapse
Affiliation(s)
- Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
47
|
Guymer C, Wood JPM, Chidlow G, Casson RJ. Neuroprotection in glaucoma: recent advances and clinical translation. Clin Exp Ophthalmol 2018; 47:88-105. [DOI: 10.1111/ceo.13336] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/21/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Chelsea Guymer
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| | - John PM Wood
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| | - Glyn Chidlow
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| | - Robert J Casson
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|
48
|
Molecular Genetics of Pigment Dispersion Syndrome and Pigmentary Glaucoma: New Insights into Mechanisms. J Ophthalmol 2018; 2018:5926906. [PMID: 29780638 PMCID: PMC5892222 DOI: 10.1155/2018/5926906] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/22/2018] [Indexed: 12/20/2022] Open
Abstract
We explore the ideas and advances surrounding the genetic basis of pigment dispersion syndrome (PDS) and pigmentary glaucoma (PG). As PG is the leading cause of nontraumatic blindness in young adults and current tailored interventions have proven ineffective, a better understanding of the underlying causes of PDS, PG, and their relationship is essential. Despite PDS being a subclinical disease, a large proportion of patients progress to PG with associated vision loss. Decades of research have supported a genetic component both for PDS and conversion to PG. We review the body of evidence supporting a genetic basis in humans and animal models and reevaluate classical mechanisms of PDS/PG considering this new evidence.
Collapse
|
49
|
Bui BV. Understanding glaucoma pathogenesis. Clin Exp Ophthalmol 2017; 45:853. [DOI: 10.1111/ceo.13072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Bang V Bui
- Department of Optometry and Vision Sciences; The University of Melbourne; Melbourne Victoria Australia
| |
Collapse
|