1
|
Shimizu S, Ochiai Y, Kamijima K, Takai N, Watanabe S, Aihara M. Development and characterization of a chronic high intraocular pressure model in New Zealand white rabbits for glaucoma research. Exp Eye Res 2024; 245:109973. [PMID: 38880377 DOI: 10.1016/j.exer.2024.109973] [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: 03/01/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Glaucoma is a neurodegenerative disease characterized by visual field loss associated with optic nerve damage and ocular hypertension. The biological basis for the elevated intraocular pressure (IOP) is largely unknown, such that lowering the IOP is currently the only established treatment. Several animal models have been developed to elucidate the mechanism underlying the increased IOP and for use in drug discovery research, but their utility is often limited by the occurrence of severe intraocular inflammation and by technical challenges. In this study, we developed a rabbit glaucoma model that does not require experimental disease induction. Rabbits were chosen as the model because their eyeballs are similar in size to those of humans, and they are easy to breed. By crossing rabbit strains with inherited glaucoma, as indicated by obvious buphthalmos, we produced a strain that exhibits ocular hypertension. The IOP of the Ocular Hypertension (OH) rabbits was significantly higher than that of the wild type (WT; normal New Zealand white rabbits) from the age of 3 weeks to at least 22 weeks. The significantly larger corneal diameter of the OH rabbits indicated ocular enlargement, whereas there was no significant difference in corneal thickness compared with WT rabbits. Anterior segment ocular coherence tomography and gonioscopic observations revealed an open angle in the OH rabbits. Hematoxylin and eosin (HE) staining together with Masson's trichrome staining showed abnormal collagen accumulation in the angle of the OH rabbit's eyes. Furthermore, aqueous humor (AH) outflow imaging following an intravitreal injection of a fluorescent probe into the anterior chamber for tissue-section analysis revealed retention of the probe in the area of collagen deposition in the OH eyes. The OH rabbits also had a time-dependent increase in the cup/disc ratio. In conclusion, investigations using our newly developed rabbit model of open-angle ocular hypertension showed that abnormal accumulation of extracellular matrix at the angle increased AH outflow resistance in the conventional outflow pathway, leading to a high IOP. Furthermore, the OH rabbits exhibited glaucomatous optic disc cupping over time. These findings suggest the utility of the OH rabbits as a model for open-angle glaucoma (OAG).
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuichiro Ochiai
- Operation Department, Kitayama Labes Co., Ltd., Nagano, Japan
| | - Kazuki Kamijima
- Operation Department, Kitayama Labes Co., Ltd., Nagano, Japan
| | - Naofumi Takai
- Operation Department, Kitayama Labes Co., Ltd., Nagano, Japan
| | - Sumiko Watanabe
- Department of Retinal Biology and Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Shimizu S, Honjo M, Liu M, Aihara M. An Autotaxin-Induced Ocular Hypertension Mouse Model Reflecting Physiological Aqueous Biomarker. Invest Ophthalmol Vis Sci 2024; 65:32. [PMID: 38386333 PMCID: PMC10896239 DOI: 10.1167/iovs.65.2.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024] Open
Abstract
Purpose Animal models of ocular hypertension (OH) have been developed to understand the pathogenesis of glaucoma and facilitate drug discovery. However, many of these models are fraught with issues, including severe intraocular inflammation and technical challenges. Lysophosphatidic acid (LPA) is implicated in trabecular meshwork fibrosis and increased resistance of aqueous outflow, factors that contribute to high intraocular pressure (IOP) in human open-angle glaucoma. We aimed to elevate IOP by increasing expression of the LPA-producing enzyme autotaxin (ATX) in mouse eyes. Methods Tamoxifen-inducible ATX transgenic mice were developed. Tamoxifen was administered to six- to eight-week-old mice via eye drops to achieve ATX overexpression in the eye. IOP and retinal thickness were measured over time, and retinal flat-mount were evaluated to count retinal ganglion cells (RGCs) loss after three months. Results Persistent elevation of ATX expression in mouse eyes was confirmed through immunohistochemistry and LysoPLD activity measurement. ATX Tg mice exhibited significantly increased IOP for nearly two months following tamoxifen treatment, with no anterior segment changes or inflammation. Immunohistochemical analysis revealed enhanced expression of extracellular matrix near the angle after two weeks and three months of ATX induction. This correlated with reduced outflow facility, indicating that sustained ATX overexpression induces angle fibrosis, elevating IOP. Although inner retinal layer thickness remained stable, peripheral retina showed a notable reduction in RGC cell count. Conclusions These findings confirm the successful creation of an open-angle OH mouse model, in which ATX expression in the eye prompts fibrosis near the angle and maintains elevated IOP over extended periods.
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Senju Laboratory of Ocular Science, Senju Pharmaceutical Co., Ltd., Kobe, Hyogo, Japan
| | - Megumi Honjo
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Mengxuan Liu
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Senapati S, Youssef AAA, Sweeney C, Cai C, Dudhipala N, Majumdar S. Cannabidiol Loaded Topical Ophthalmic Nanoemulsion Lowers Intraocular Pressure in Normotensive Dutch-Belted Rabbits. Pharmaceutics 2022; 14:2585. [PMID: 36559077 PMCID: PMC9781840 DOI: 10.3390/pharmaceutics14122585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Cannabidiol (CBD) is the major non-psychoactive and most widely studied of the cannabinoid constituents and has great therapeutic potential in a variety of diseases. However, contradictory reports in the literature with respect to CBD's effect on intraocular pressure (IOP) have raised concerns and halted research exploring its use in ocular therapeutics. Therefore, the current investigation aimed to further evaluate CBD's impact on the IOP in the rabbit model. CBD nanoemulsions, containing Carbopol® 940 NF as a mucoadhesive agent (CBD-NEC), were prepared using hot-homogenization followed by probe sonication. The stability of the formulations post-moist-heat sterilization, in terms of physical and chemical characteristics, was studied for three different storage conditions. The effect of the formulation on the intraocular pressure (IOP) profile in normotensive Dutch Belted male rabbits was then examined. The lead CBD-NEC formulation (1% w/v CBD) exhibited a globule size of 259 ± 2.0 nm, 0.27 ± 0.01 PDI, and 23.2 ± 0.4 cP viscosity, and was physically and chemically stable for one month (last time point tested) at 4 °C, 25 °C, and 40 °C. CBD-NEC significantly lowered the IOP in the treated eyes for up to 360 min, with a peak drop in IOP of 4.5 mmHg observed at the 150 min time point, post-topical application. The IOP of the contralateral eye (untreated) was also observed to be lowered significantly, but the effect lasted up to the 180 min time point only. Overall, topically administered CBD, formulated in a mucoadhesive nanoemulsion formulation, reduced the IOP in the animal model studied. The results support further exploration of CBD as a therapeutic option for various inflammation-based ocular diseases.
Collapse
Affiliation(s)
- Samir Senapati
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Ahmed Adel Ali Youssef
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Corinne Sweeney
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Chuntian Cai
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Narendar Dudhipala
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Soumyajit Majumdar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi, Oxford, MS 38677, USA
| |
Collapse
|
4
|
Hongjamrassilp W, Zhang R, Natterson-Horowitz B, Blumstein DT. Glaucoma through Animal’s Eyes: Insights from the Evolution of Intraocular Pressure in Mammals and Birds. Animals (Basel) 2022; 12:ani12162027. [PMID: 36009617 PMCID: PMC9404445 DOI: 10.3390/ani12162027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/06/2022] [Accepted: 08/07/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Understanding how a disease evolved across the animal kingdom could help us better understand the disease and might lead to novel methods for treatment. Here, we studied the evolution of glaucoma, an irreversible eye disease, in mammals and birds, by studying the evolution of intraocular pressure (IOP), a central driver of glaucoma, and searching for associations between life history traits and IOP. Our results revealed that IOP is a taxa-specific trait that is higher in some species than in others. Higher IOPs appear to have evolved multiple times in mammals and birds. Higher IOPs were found in mammals with higher body mass and in aquatic birds. We also found that higher IOPs evolved through stabilizing selection, with the optimum IOP in mammals and birds being 17.67 and 14.31 mmHg, respectively. This supports the hypothesis that higher IOPs may be an adaptive trait for certain animals. Focusing on species with higher IOPs but no evidence of glaucoma may help identify glaucoma-resistant adaptations, which could be developed into human therapies. Abstract Glaucoma, an eye disorder caused by elevated intraocular pressure (IOP), is the leading cause of irreversible blindness in humans. Understanding how IOP levels have evolved across animal species could shed light on the nature of human vulnerability to glaucoma. Here, we studied the evolution of IOP in mammals and birds and explored its life history correlates. We conducted a systematic review, to create a dataset of species-specific IOP levels and reconstructed the ancestral states of IOP using three models of evolution (Brownian, Early burst, and Ornstein–Uhlenbeck (OU)) to understand the evolution of glaucoma. Furthermore, we tested the association between life history traits (e.g., body mass, blood pressure, diet, longevity, and habitat) and IOP using phylogenetic generalized least squares (PGLS). IOP in mammals and birds evolved under the OU model, suggesting stabilizing selection toward an optimal value. Larger mammals had higher IOPs and aquatic birds had higher IOPs; no other measured life history traits, the type of tonometer used, or whether the animal was sedated when measuring IOP explained the significant variation in IOP in this dataset. Elevated IOP, which could result from physiological and anatomical processes, evolved multiple times in mammals and birds. However, we do not understand how species with high IOP avoid glaucoma. While we found very few associations between life history traits and IOP, we suggest that more detailed studies may help identify mechanisms by which IOP is decoupled from glaucoma. Importantly, species with higher IOPs (cetaceans, pinnipeds, and rhinoceros) could be good model systems for studying glaucoma-resistant adaptations.
Collapse
Affiliation(s)
- Watcharapong Hongjamrassilp
- Department of Marine Science, Faculty of Science, Chulalongkorn University, Phayathai Road, Bangkok 10330, Thailand
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, 621 Young Drive South, Los Angeles, CA 90095, USA
- Correspondence: or (W.H.); (D.T.B.)
| | - Roger Zhang
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, 621 Young Drive South, Los Angeles, CA 90095, USA
| | - B. Natterson-Horowitz
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, 621 Young Drive South, Los Angeles, CA 90095, USA
- David Geffen School of Medicine at UCLA, Division of Cardiology, 650 Charles E. Young Drive South, A2-237, Los Angeles, CA 90095, USA
| | - Daniel T. Blumstein
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, 621 Young Drive South, Los Angeles, CA 90095, USA
- Correspondence: or (W.H.); (D.T.B.)
| |
Collapse
|
5
|
McDowell CM, Kizhatil K, Elliott MH, Overby DR, van Batenburg-Sherwood J, Millar JC, Kuehn MH, Zode G, Acott TS, Anderson MG, Bhattacharya SK, Bertrand JA, Borras T, Bovenkamp DE, Cheng L, Danias J, De Ieso ML, Du Y, Faralli JA, Fuchshofer R, Ganapathy PS, Gong H, Herberg S, Hernandez H, Humphries P, John SWM, Kaufman PL, Keller KE, Kelley MJ, Kelly RA, Krizaj D, Kumar A, Leonard BC, Lieberman RL, Liton P, Liu Y, Liu KC, Lopez NN, Mao W, Mavlyutov T, McDonnell F, McLellan GJ, Mzyk P, Nartey A, Pasquale LR, Patel GC, Pattabiraman PP, Peters DM, Raghunathan V, Rao PV, Rayana N, Raychaudhuri U, Reina-Torres E, Ren R, Rhee D, Chowdhury UR, Samples JR, Samples EG, Sharif N, Schuman JS, Sheffield VC, Stevenson CH, Soundararajan A, Subramanian P, Sugali CK, Sun Y, Toris CB, Torrejon KY, Vahabikashi A, Vranka JA, Wang T, Willoughby CE, Xin C, Yun H, Zhang HF, Fautsch MP, Tamm ER, Clark AF, Ethier CR, Stamer WD. Consensus Recommendation for Mouse Models of Ocular Hypertension to Study Aqueous Humor Outflow and Its Mechanisms. Invest Ophthalmol Vis Sci 2022; 63:12. [PMID: 35129590 PMCID: PMC8842499 DOI: 10.1167/iovs.63.2.12] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
Due to their similarities in anatomy, physiology, and pharmacology to humans, mice are a valuable model system to study the generation and mechanisms modulating conventional outflow resistance and thus intraocular pressure. In addition, mouse models are critical for understanding the complex nature of conventional outflow homeostasis and dysfunction that results in ocular hypertension. In this review, we describe a set of minimum acceptable standards for developing, characterizing, and utilizing mouse models of open-angle ocular hypertension. We expect that this set of standard practices will increase scientific rigor when using mouse models and will better enable researchers to replicate and build upon previous findings.
Collapse
Affiliation(s)
- Colleen M. McDowell
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | | - Michael H. Elliott
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Darryl R. Overby
- Department of Bioengineering, Imperial College London, United Kingdom
| | | | - J. Cameron Millar
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences and Institute for Vision Research, The University of Iowa; Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, Iowa, United States
| | - Gulab Zode
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Ted S. Acott
- Ophthalmology and Biochemistry and Molecular Biology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Michael G. Anderson
- Department of Molecular Physiology and Biophysics and Department of Ophthalmology and Visual Sciences, The University of Iowa; Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, Iowa, United States
| | | | - Jacques A. Bertrand
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Terete Borras
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | | | - Lin Cheng
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - John Danias
- SUNY Downstate Health Sciences University, Brooklyn, New York, United States
| | - Michael Lucio De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania, United States
| | - Jennifer A. Faralli
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | | | - Peter Humphries
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Simon W. M. John
- Department of Ophthalmology, Columbia University, New York, New York, United States
| | - Paul L. Kaufman
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Kate E. Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Mary J. Kelley
- Department of Ophthalmology and Department of Integrative Biosciences, Oregon Health & Science University, Portland, Oregon, United States
| | - Ruth A. Kelly
- Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - David Krizaj
- Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania, United States
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, University of California, Davis, Davis, California, United States
| | - Raquel L. Lieberman
- Department of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Paloma Liton
- Department of Ophthalmology and Department of Pathology, Duke University, Durham, North Carolina, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, James & Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Katy C. Liu
- Duke Eye Center, Duke Health, Durham, North Carolina, United States
| | - Navita N. Lopez
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, United States
| | - Weiming Mao
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Timur Mavlyutov
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Fiona McDonnell
- Duke Eye Center, Duke Health, Durham, North Carolina, United States
| | - Gillian J. McLellan
- Department of Surgical Sciences and Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Philip Mzyk
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Andrews Nartey
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Louis R. Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Gaurang C. Patel
- Ophthalmology Research, Regeneron Pharmaceuticals, Tarreytown, New York, United States
| | | | - Donna M. Peters
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Naga Rayana
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Urmimala Raychaudhuri
- Department of Neurobiology, University of California, Irvine, Irvine, California, United States
| | - Ester Reina-Torres
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Ruiyi Ren
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Douglas Rhee
- Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Uttio Roy Chowdhury
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - John R. Samples
- Washington State University, Floyd Elson College of Medicine, Spokane, Washington, United States
| | | | - Najam Sharif
- Santen Inc., Emeryville, California, United States
| | - Joel S. Schuman
- Department of Ophthalmology and Department of Physiology and Neuroscience, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States; Departments of Biomedical Engineering and Electrical and Computer Engineering, New York University Tandon School of Engineering, Brooklyn, New York, United States; Center for Neural Science, College of Arts and Science, New York University, New York, New York, United States
| | - Val C. Sheffield
- Department of Pediatrics and Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Cooper H. Stevenson
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Avinash Soundararajan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | | | - Chenna Kesavulu Sugali
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Yang Sun
- Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California, United States
| | - Carol B. Toris
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States; Department of Ophthalmology and Vision Sciences, The Ohio State University, Columbus, Ohio, United States
| | | | - Amir Vahabikashi
- Cell and Developmental Biology Department, Northwestern University, Chicago, Illinois, United States
| | - Janice A. Vranka
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ting Wang
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Colin E. Willoughby
- Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Chen Xin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hongmin Yun
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Hao F. Zhang
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States
| | - Michael P. Fautsch
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States
| | | | - Abbot F. Clark
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - C. Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology; Emory University School of Medicine, Emory University, Atlanta, Georgia, United States
| | - W. Daniel Stamer
- Duke Ophthalmology, Duke University, Durham, North Carolina, United States
| |
Collapse
|
6
|
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: 3.0] [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
|
7
|
Sharif NA. Therapeutic Drugs and Devices for Tackling Ocular Hypertension and Glaucoma, and Need for Neuroprotection and Cytoprotective Therapies. Front Pharmacol 2021; 12:729249. [PMID: 34603044 PMCID: PMC8484316 DOI: 10.3389/fphar.2021.729249] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
Damage to the optic nerve and the death of associated retinal ganglion cells (RGCs) by elevated intraocular pressure (IOP), also known as glaucoma, is responsible for visual impairment and blindness in millions of people worldwide. The ocular hypertension (OHT) and the deleterious mechanical forces it exerts at the back of the eye, at the level of the optic nerve head/optic disc and lamina cribosa, is the only modifiable risk factor associated with glaucoma that can be treated. The elevated IOP occurs due to the inability of accumulated aqueous humor (AQH) to egress from the anterior chamber of the eye due to occlusion of the major outflow pathway, the trabecular meshwork (TM) and Schlemm’s canal (SC). Several different classes of pharmaceutical agents, surgical techniques and implantable devices have been developed to lower and control IOP. First-line drugs to promote AQH outflow via the uveoscleral outflow pathway include FP-receptor prostaglandin (PG) agonists (e.g., latanoprost, travoprost and tafluprost) and a novel non-PG EP2-receptor agonist (omidenepag isopropyl, Eybelis®). TM/SC outflow enhancing drugs are also effective ocular hypotensive agents (e.g., rho kinase inhibitors like ripasudil and netarsudil; and latanoprostene bunod, a conjugate of a nitric oxide donor and latanoprost). One of the most effective anterior chamber AQH microshunt devices is the Preserflo® microshunt which can lower IOP down to 10–13 mmHg. Other IOP-lowering drugs and devices on the horizon will be also discussed. Additionally, since elevated IOP is only one of many risk factors for development of glaucomatous optic neuropathy, a treatise of the role of inflammatory neurodegeneration of the optic nerve and retinal ganglion cells and appropriate neuroprotective strategies to mitigate this disease will also be reviewed and discussed.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| |
Collapse
|
8
|
van der Merwe Y, Murphy MC, Sims JR, Faiq MA, Yang XL, Ho LC, Conner IP, Yu Y, Leung CK, Wollstein G, Schuman JS, Chan KC. Citicoline Modulates Glaucomatous Neurodegeneration Through Intraocular Pressure-Independent Control. Neurotherapeutics 2021; 18:1339-1359. [PMID: 33846961 PMCID: PMC8423893 DOI: 10.1007/s13311-021-01033-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
Glaucoma is a neurodegenerative disease that causes progressive, irreversible vision loss. Currently, intraocular pressure (IOP) is the only modifiable risk factor for glaucoma. However, glaucomatous degeneration may continue despite adequate IOP control. Therefore, there exists a need for treatment that protects the visual system, independent of IOP. This study sought, first, to longitudinally examine the neurobehavioral effects of different magnitudes and durations of IOP elevation using multi-parametric magnetic resonance imaging (MRI), optokinetics and histology; and, second, to evaluate the effects of oral citicoline treatment as a neurotherapeutic in experimental glaucoma. Eighty-two adult Long Evans rats were divided into six groups: acute (mild or severe) IOP elevation, chronic (citicoline-treated or untreated) IOP elevation, and sham (acute or chronic) controls. We found that increasing magnitudes and durations of IOP elevation differentially altered structural and functional brain connectivity and visuomotor behavior, as indicated by decreases in fractional anisotropy in diffusion tensor MRI, magnetization transfer ratios in magnetization transfer MRI, T1-weighted MRI enhancement of anterograde manganese transport, resting-state functional connectivity, visual acuity, and neurofilament and myelin staining along the visual pathway. Furthermore, 3 weeks of oral citicoline treatment in the setting of chronic IOP elevation significantly reduced visual brain integrity loss and visual acuity decline without altering IOP. Such effects sustained after treatment was discontinued for another 3 weeks. These results not only illuminate the close interplay between eye, brain, and behavior in glaucomatous neurodegeneration, but also support a role for citicoline in protecting neural tissues and visual function in glaucoma beyond IOP control.
Collapse
Affiliation(s)
- Yolandi van der Merwe
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew C Murphy
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey R Sims
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
| | - Muneeb A Faiq
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
| | - Xiao-Ling Yang
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leon C Ho
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ian P Conner
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yu Yu
- Pleryon Therapeutics Limited, Shenzhen, China
| | - Christopher K Leung
- University Eye Center, Hong Kong Eye Hospital, Hong Kong, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gadi Wollstein
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
- Center for Neural Science, College of Arts and Science, New York University, New York, NY, USA
| | - Joel S Schuman
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
- Center for Neural Science, College of Arts and Science, New York University, New York, NY, USA
- Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
| | - Kevin C Chan
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA.
- Center for Neural Science, College of Arts and Science, New York University, New York, NY, USA.
- Department of Radiology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA.
- Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA.
| |
Collapse
|
9
|
Evangelho K, Mastronardi CA, de-la-Torre A. Experimental Models of Glaucoma: A Powerful Translational Tool for the Future Development of New Therapies for Glaucoma in Humans-A Review of the Literature. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E280. [PMID: 31212881 PMCID: PMC6630440 DOI: 10.3390/medicina55060280] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Abstract
Glaucoma is a common complex disease that leads to irreversible blindness worldwide. Even though preclinical studies showed that lowering intraocular pressure (IOP) could prevent retinal ganglion cells loss, clinical evidence suggests that lessening IOP does not prevent glaucoma progression in all patients. Glaucoma is also becoming more prevalent in the elderly population, showing that age is a recognized major risk factor. Indeed, recent findings suggest that age-related tissue alterations contribute to the development of glaucoma and have encouraged exploration for new treatment approaches. In this review, we provide information on the most frequently used experimental models of glaucoma and describe their advantages and limitations. Additionally, we describe diverse animal models of glaucoma that can be potentially used in translational medicine and aid an efficient shift to the clinic. Experimental animal models have helped to understand the mechanisms of formation and evacuation of aqueous humor, and the maintenance of homeostasis of intra-ocular pressure. However, the transfer of pre-clinical results obtained from animal studies into clinical trials may be difficult since the type of study does not only depend on the type of therapy to be performed, but also on a series of factors observed both in the experimental period and the period of transfer to clinical application. Conclusions: Knowing the exact characteristics of each glaucoma experimental model could help to diminish inconveniences related to the process of the translation of results into clinical application in humans.
Collapse
Affiliation(s)
- Karine Evangelho
- Doctorado en Ciencias Biomédicas y Biológicas, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá,11121, Colombia.
| | - Claudio A Mastronardi
- Neuroscience Research Group (NeurUROS), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, 11121, Colombia.
| | - Alejandra de-la-Torre
- Neuroscience Research Group (NeurUROS), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, 11121, Colombia.
| |
Collapse
|
10
|
Reinhard J, Wiemann S, Joachim SC, Palmhof M, Woestmann J, Denecke B, Wang Y, Downey GP, Faissner A. Heterozygous Meg2 Ablation Causes Intraocular Pressure Elevation and Progressive Glaucomatous Neurodegeneration. Mol Neurobiol 2019; 56:4322-4345. [PMID: 30315478 DOI: 10.1007/s12035-018-1376-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
Glaucomatous neurodegeneration represents one of the major causes of irreversible blindness worldwide. Yet, the detailed molecular mechanisms that initiate optic nerve damage and retinal ganglion cell (RGC) loss are not fully understood. Members of the protein tyrosine phosphatase (PTP) superfamily are key players in numerous neurodegenerative diseases. In order to investigate the potential functional relevance of the PTP megakaryocyte 2 (Meg2) in retinal neurodegeneration, we analyzed Meg2 knockout (KO) and heterozygous (HET)-synonym protein-tyrosine phosphatase non-receptor type 9 (Ptpn9)-mice. Interestingly, via global microarray and quantitative real-time PCR (RT-qPCR) analyses of Meg2 KO and HET retinae, we observed a dysregulation of several candidate genes that are highly associated with retinal degeneration and intraocular pressure (IOP) elevation, the main risk factor for glaucoma. Subsequent IOP measurements in Meg2 HET mice verified progressive age-dependent IOP elevation. Ultrastructural analyses and immunohistochemistry showed severe optic nerve degeneration accompanied by a dramatic loss of RGCs. Additionally, HET mice displayed reactive micro-/macrogliosis and early activation of the classical complement cascade with pronounced deposition of the membrane attack complex (MAC) in the retina and optic nerve. When treated with latanoprost, significant IOP lowering prevented RGC loss and microglial invasion in HET mice. Finally, electroretinogram (ERG) recordings revealed reduced a- and b-wave amplitudes, indicating impaired retinal functionality in Meg2 HET mice. Collectively, our findings indicate that the heterozygous loss of Meg2 in mice is sufficient to cause IOP elevation and glaucomatous neurodegeneration. Thus, Meg2 HET mice may serve as a novel animal model to study the pathomechanism involved in the onset and progression of glaucoma.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Marina Palmhof
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Julia Woestmann
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Bernd Denecke
- Interdisciplinary Centre for Clinical Research, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Yingchun Wang
- Division of Respirology, Department of Medicine, University of Toronto and Toronto General Hospital Research Institute of the University Health Network, 610 University Avenue, Toronto, ON, M5S 1A8, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Departments of Medicine and Immunology and Microbiology, University of Colorado, Aurora, CO, 80045, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Departments of Medicine, Pediatrics and Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
11
|
Samuels BC, Siegwart JT, Zhan W, Hethcox L, Chimento M, Whitley R, Downs JC, Girkin CA. A Novel Tree Shrew (Tupaia belangeri) Model of Glaucoma. Invest Ophthalmol Vis Sci 2019; 59:3136-3143. [PMID: 30025140 PMCID: PMC6018453 DOI: 10.1167/iovs.18-24261] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Primates and rodents are used widely as animal models of glaucoma, but each has significant limitations. Researchers need additional animal models that closely resemble the relevant anatomy and pathologic features of the human disease to more quickly advance research. We validate a novel glaucoma animal model in tree shrews (Tupaia belangeri). Methods Experimental glaucoma was induced in adult tree shrews (n = 8) by injecting 50 μL of a 25 mg/mL ferromagnetic bead solution into the anterior chamber. Beads were directed into the iridocorneal angle with a magnet to impede aqueous outflow. Animals were followed for 3 months with weekly IOP measurements and biweekly spectral domain optical coherence tomography (SD-OCT) images of the optic nerve head. Histopathology of the optic nerve and optic nerve axon counts were completed at the end of the study. Results The 12-week average mean IOP was 22.7 ± 3.6 and 8.6 ± 2.9 mm Hg in the treated and control eyes, respectively. Longitudinal analysis showed significant retinal nerve fiber layer (RNFL) thinning throughout the study. Axon counts were significantly reduced (59.7%) in treated versus control eyes. SD-OCT imaging showed cupping and posterior displacement of the lamina cribrosa in glaucomatous eyes. RNFL thickness and optic nerve axon counts were reduced consistent with IOP elevation. Optic nerves demonstrated histopathology consistent with glaucomatous optic neuropathy. Conclusions Tree shrews with experimental glaucoma show key pathologic characteristics of the human disease. The tree shrew model of glaucoma has the potential to help researchers accelerate our understanding of glaucoma pathophysiology.
Collapse
Affiliation(s)
- Brian C Samuels
- Department of Ophthalmology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - John T Siegwart
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Wenjie Zhan
- Department of Ophthalmology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Lisa Hethcox
- Department of Ophthalmology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Melissa Chimento
- High Resolution Imaging Facility, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Ryan Whitley
- Department of Ophthalmology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - J Crawford Downs
- Department of Ophthalmology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Christopher A Girkin
- Department of Ophthalmology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| |
Collapse
|
12
|
Dey A, Manthey AL, Chiu K, Do CW. Methods to Induce Chronic Ocular Hypertension: Reliable Rodent Models as a Platform for Cell Transplantation and Other Therapies. Cell Transplant 2019; 27:213-229. [PMID: 29637819 PMCID: PMC5898687 DOI: 10.1177/0963689717724793] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glaucoma, a form of progressive optic neuropathy, is the second leading cause of blindness worldwide. Being a prominent disease affecting vision, substantial efforts are being made to better understand glaucoma pathogenesis and to develop novel treatment options including neuroprotective and neuroregenerative approaches. Cell transplantation has the potential to play a neuroprotective and/or neuroregenerative role for various ocular cell types (e.g., retinal cells, trabecular meshwork). Notably, glaucoma is often associated with elevated intraocular pressure, and over the past 2 decades, several rodent models of chronic ocular hypertension (COH) have been developed that reflect these changes in pressure. However, the underlying pathophysiology of glaucoma in these models and how they compare to the human condition remains unclear. This limitation is the primary barrier for using rodent models to develop novel therapies to manage glaucoma and glaucoma-related blindness. Here, we review the current techniques used to induce COH-related glaucoma in various rodent models, focusing on the strengths and weaknesses of the each, in order to provide a more complete understanding of how these models can be best utilized. To so do, we have separated them based on the target tissue (pre-trabecular, trabecular, and post-trabecular) in order to provide the reader with an encompassing reference describing the most appropriate rodent COH models for their research. We begin with an initial overview of the current use of these models in the evaluation of cell transplantation therapies.
Collapse
Affiliation(s)
- Ashim Dey
- 1 School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Abby L Manthey
- 2 Laboratory of Retina Brain Research, Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kin Chiu
- 2 Laboratory of Retina Brain Research, Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,3 Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong, China.,4 State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Chi-Wai Do
- 1 School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
13
|
Toris CB, Fan S, Johnson TV, Camras LJ, Hays CL, Liu H, Ishimoto BM. Aqueous Flow Measured by Fluorophotometry in the Mouse. Invest Ophthalmol Vis Sci 2017; 57:3844-52. [PMID: 27447085 PMCID: PMC4968429 DOI: 10.1167/iovs.14-15144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE A fluorophotometer designed to measure aqueous flow in murine eyes was tested with artificial fluorescein chambers and in live mice with different anesthesia regimens, aqueous flow suppressants, and an anterior chamber cannulation method. METHODS Two hours following topical fluorescein application, one group of CD-1 mice was anesthetized with ketamine/xylazine, 2,2,2-tribromoethanol, or ketamine alone. Cornea and anterior chamber fluorescein concentrations were measured periodically for 60 to 90 minutes by fluorophotometric scans to calculate aqueous flow. Later, a subgroup of mice underwent aqueous flow measurement by anterior chamber cannulation. A third group was treated with timolol, dorzolamide, and vehicle in a crossover manner 1 hour prior to fluorophotometric scans. RESULTS Aqueous flow with ketamine/xylazine anesthesia (0.09 ± 0.05 μL/min, mean ± SD, n = 24) was slower than with tribromoethanol or ketamine alone (P < 0.001). Timolol reduced aqueous flow from 0.20 ± 0.07 μL/min to 0.07 ± 0.03 μL/min (P = 0.001) under tribromoethanol anesthesia and from 0.14 ± 0.03 μL/min to 0.10 ± 0.02 μL/min (P = 0.004) under ketamine anesthesia but not under ketamine/xylazine anesthesia. Dorzolamide reduced aqueous flow from 0.09 ± 0.03 to 0.06 ± 0.03 μL/min (P = 0.04) under ketamine/xylazine anesthesia. Aqueous flow by anterior chamber cannulation (0.20 ± 0.13 μL/min) was greater (P = 0.05) than by fluorophotometry (0.09 ± 0.07 μL/min). CONCLUSIONS A new noninvasive fluorophotometric method detected effects of general anesthesia and known aqueous suppressants on aqueous flow in mice. Aqueous flow measured by fluorophotometry was slower than by cannulation, and was technically easier with less variability. The mouse fluorophotometer is useful for repeated measurements of aqueous flow in the murine eye making crossover and longitudinal studies possible.
Collapse
Affiliation(s)
- Carol B Toris
- Department of Ophthalmology and Visual Sciences University of Nebraska Medical Center, Omaha, Nebraska, United States 2Case Western Reserve University, Cleveland, Ohio, United States
| | - Shan Fan
- Department of Ophthalmology and Visual Sciences University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Thomas V Johnson
- Department of Ophthalmology and Visual Sciences University of Nebraska Medical Center, Omaha, Nebraska, United States 3Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lucinda J Camras
- Department of Ophthalmology and Visual Sciences University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Cassandra L Hays
- Department of Ophthalmology and Visual Sciences University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Hong Liu
- Department of Ophthalmology and Visual Sciences University of Nebraska Medical Center, Omaha, Nebraska, United States
| | | |
Collapse
|
14
|
Augustin M, Fialová S, Himmel T, Glösmann M, Lengheimer T, Harper DJ, Plasenzotti R, Pircher M, Hitzenberger CK, Baumann B. Multi-Functional OCT Enables Longitudinal Study of Retinal Changes in a VLDLR Knockout Mouse Model. PLoS One 2016; 11:e0164419. [PMID: 27711217 PMCID: PMC5053493 DOI: 10.1371/journal.pone.0164419] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/23/2016] [Indexed: 12/24/2022] Open
Abstract
We present a multi-functional optical coherence tomography (OCT) imaging approach to study retinal changes in the very-low-density-lipoprotein-receptor (VLDLR) knockout mouse model with a threefold contrast. In the retinas of VLDLR knockout mice spontaneous retinal-chorodoidal neovascularizations form, having an appearance similar to choroidal and retinal neovascularizations (CNV and RNV) in neovascular age-related macular degeneration (AMD) or retinal angiomatous proliferation (RAP). For this longitudinal study, the mice were imaged every 4 to 6 weeks starting with an age of 4 weeks and following up to the age of 11 months. Significant retinal changes were identified by the multi-functional imaging approach offering a threefold contrast: reflectivity, polarization sensitivity (PS) and motion contrast based OCT angiography (OCTA). By use of this intrinsic contrast, the long-term development of neovascularizations was studied and associated processes, such as the migration of melanin pigments or retinal-choroidal anastomosis, were assessed in vivo. Furthermore, the in vivo imaging results were validated with histological sections at the endpoint of the experiment. Multi-functional OCT proves as a powerful tool for longitudinal retinal studies in preclinical research of ophthalmic diseases. Intrinsic contrast offered by the functional extensions of OCT might help to describe regulative processes in genetic animal models and potentially deepen the understanding of the pathogenesis of retinal diseases such as wet AMD.
Collapse
Affiliation(s)
- Marco Augustin
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Stanislava Fialová
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Tanja Himmel
- Core Facility for Research and Technology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martin Glösmann
- Core Facility for Research and Technology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Theresia Lengheimer
- Division of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Danielle J. Harper
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Roberto Plasenzotti
- Division of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Michael Pircher
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Christoph K. Hitzenberger
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Kuehn MH, Lipsett KA, Menotti-Raymond M, Whitmore SS, Scheetz TE, David VA, O'Brien SJ, Zhao Z, Jens JK, Snella EM, Ellinwood NM, McLellan GJ. A Mutation in LTBP2 Causes Congenital Glaucoma in Domestic Cats (Felis catus). PLoS One 2016; 11:e0154412. [PMID: 27149523 PMCID: PMC4858209 DOI: 10.1371/journal.pone.0154412] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/31/2016] [Indexed: 01/18/2023] Open
Abstract
The glaucomas are a group of diseases characterized by optic nerve damage that together represent a leading cause of blindness in the human population and in domestic animals. Here we report a mutation in LTBP2 that causes primary congenital glaucoma (PCG) in domestic cats. We identified a spontaneous form of PCG in cats and established a breeding colony segregating for PCG consistent with fully penetrant, autosomal recessive inheritance of the trait. Elevated intraocular pressure, globe enlargement and elongated ciliary processes were consistently observed in all affected cats by 8 weeks of age. Varying degrees of optic nerve damage resulted by 6 months of age. Although subtle lens zonular instability was a common feature in this cohort, pronounced ectopia lentis was identified in less than 10% of cats examined. Thus, glaucoma in this pedigree is attributed to histologically confirmed arrest in the early post-natal development of the aqueous humor outflow pathways in the anterior segment of the eyes of affected animals. Using a candidate gene approach, significant linkage was established on cat chromosome B3 (LOD 18.38, θ = 0.00) using tightly linked short tandem repeat (STR) loci to the candidate gene, LTBP2. A 4 base-pair insertion was identified in exon 8 of LTBP2 in affected individuals that generates a frame shift that completely alters the downstream open reading frame and eliminates functional domains. Thus, we describe the first spontaneous and highly penetrant non-rodent model of PCG identifying a valuable animal model for primary glaucoma that closely resembles the human disease, providing valuable insights into mechanisms underlying the disease and a valuable animal model for testing therapies.
Collapse
Affiliation(s)
- Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Koren A. Lipsett
- Laboratory of Genomic Diversity, National Cancer Institute, Frederick, Maryland, United States of America
- Department of Chemistry, Gettysburg College, Gettysburg, Pennsylvania, United States of America
| | - Marilyn Menotti-Raymond
- Laboratory of Genomic Diversity, National Cancer Institute, Frederick, Maryland, United States of America
| | - S. Scott Whitmore
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Todd E. Scheetz
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Victor A. David
- Laboratory of Genomic Diversity, National Cancer Institute, Frederick, Maryland, United States of America
- Basic Research Laboratory, National Cancer Institute, Frederick, Maryland, United States of America
| | - Stephen J. O'Brien
- Laboratory of Genomic Diversity, National Cancer Institute, Frederick, Maryland, United States of America
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
- Oceanographic Center, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - Zhongyuan Zhao
- Department of Chemistry, Gettysburg College, Gettysburg, Pennsylvania, United States of America
| | - Jackie K. Jens
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Elizabeth M. Snella
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - N. Matthew Ellinwood
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Gillian J. McLellan
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Surgical Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- McPherson Eye Research Institute, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
16
|
Donegan RK, Lieberman RL. Discovery of Molecular Therapeutics for Glaucoma: Challenges, Successes, and Promising Directions. J Med Chem 2016; 59:788-809. [PMID: 26356532 PMCID: PMC5547565 DOI: 10.1021/acs.jmedchem.5b00828] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glaucoma, a heterogeneous ocular disorder affecting ∼60 million people worldwide, is characterized by painless neurodegeneration of retinal ganglion cells (RGCs), resulting in irreversible vision loss. Available therapies, which decrease the common causal risk factor of elevated intraocular pressure, delay, but cannot prevent, RGC death and blindness. Notably, it is changes in the anterior segment of the eye, particularly in the drainage of aqueous humor fluid, which are believed to bring about changes in pressure. Thus, it is primarily this region whose properties are manipulated in current and emerging therapies for glaucoma. Here, we focus on the challenges associated with developing treatments, review the available experimental methods to evaluate the therapeutic potential of new drugs, describe the development and evaluation of emerging Rho-kinase inhibitors and adenosine receptor ligands that offer the potential to improve aqueous humor outflow and protect RGCs simultaneously, and present new targets and approaches on the horizon.
Collapse
Affiliation(s)
- Rebecca K Donegan
- School of Chemistry and Biochemistry, Georgia Institute of Technology , 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| | - Raquel L Lieberman
- School of Chemistry and Biochemistry, Georgia Institute of Technology , 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| |
Collapse
|
17
|
Green DI, Ou Y. Towards the development of a human glaucoma disease-in-a-dish model using stem cells. EXPERT REVIEW OF OPHTHALMOLOGY 2015. [DOI: 10.1586/17469899.2015.1026329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Abbott CJ, Choe TE, Lusardi TA, Burgoyne CF, Wang L, Fortune B. Evaluation of retinal nerve fiber layer thickness and axonal transport 1 and 2 weeks after 8 hours of acute intraocular pressure elevation in rats. Invest Ophthalmol Vis Sci 2014; 55:674-87. [PMID: 24398096 DOI: 10.1167/iovs.13-12811] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE To compare in vivo retinal nerve fiber layer thickness (RNFLT) and axonal transport at 1 and 2 weeks after an 8-hour acute IOP elevation in rats. METHODS Forty-seven adult male Brown Norway rats were used. Procedures were performed under anesthesia. The IOP was manometrically elevated to 50 mm Hg or held at 15 mm Hg (sham) for 8 hours unilaterally. The RNFLT was measured by spectral-domain optical coherence tomography. Anterograde and retrograde axonal transport was assessed from confocal scanning laser ophthalmoscopy imaging 24 hours after bilateral injections of 2 μL 1% cholera toxin B-subunit conjugated to AlexaFluor 488 into the vitreous or superior colliculi, respectively. Retinal ganglion cell (RGC) and microglial densities were determined using antibodies against Brn3a and Iba-1. RESULTS The RNFLT in experimental eyes increased from baseline by 11% at 1 day (P < 0.001), peaked at 19% at 1 week (P < 0.0001), remained 11% thicker at 2 weeks (P < 0.001), recovered at 3 weeks (P > 0.05), and showed no sign of thinning at 6 weeks (P > 0.05). There was no disruption of anterograde transport at 1 week (superior colliculi fluorescence intensity, 75.3 ± 7.9 arbitrary units [AU] for the experimental eyes and 77.1 ± 6.7 AU for the control eyes) (P = 0.438) or 2 weeks (P = 0.188). There was no obstruction of retrograde transport at 1 week (RCG density, 1651 ± 153 per mm(2) for the experimental eyes and 1615 ± 135 per mm(2) for the control eyes) (P = 0.63) or 2 weeks (P = 0.25). There was no loss of Brn3a-positive RGC density at 6 weeks (P = 0.74) and no increase in microglial density (P = 0.92). CONCLUSIONS Acute IOP elevation to 50 mm Hg for 8 hours does not cause a persisting axonal transport deficit at 1 or 2 weeks or a detectable RNFLT or RGC loss by 6 weeks but does lead to transient RNFL thickening that resolves by 3 weeks.
Collapse
Affiliation(s)
- Carla J Abbott
- Discoveries in Sight Research Laboratories, Legacy Good Samaritan Devers Eye Institute, and Legacy Research Institute, Legacy Health, Portland, Oregon
| | | | | | | | | | | |
Collapse
|
19
|
Soluble guanylate cyclase α1-deficient mice: a novel murine model for primary open angle glaucoma. PLoS One 2013; 8:e60156. [PMID: 23527308 PMCID: PMC3603933 DOI: 10.1371/journal.pone.0060156] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/21/2013] [Indexed: 12/29/2022] Open
Abstract
Primary open angle glaucoma (POAG) is a leading cause of blindness worldwide. The molecular signaling involved in the pathogenesis of POAG remains unknown. Here, we report that mice lacking the α1 subunit of the nitric oxide receptor soluble guanylate cyclase represent a novel and translatable animal model of POAG, characterized by thinning of the retinal nerve fiber layer and loss of optic nerve axons in the context of an open iridocorneal angle. The optic neuropathy associated with soluble guanylate cyclase α1-deficiency was accompanied by modestly increased intraocular pressure and retinal vascular dysfunction. Moreover, data from a candidate gene association study suggests that a variant in the locus containing the genes encoding for the α1 and β1 subunits of soluble guanylate cyclase is associated with POAG in patients presenting with initial paracentral vision loss, a disease subtype thought to be associated with vascular dysregulation. These findings provide new insights into the pathogenesis and genetics of POAG and suggest new therapeutic strategies for POAG.
Collapse
|
20
|
Gilger BC, Abarca E, Salmon JH. Selection of Appropriate Animal Models in Ocular Research: Ocular Anatomy and Physiology of Common Animal Models. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2013. [DOI: 10.1007/7653_2013_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Casson RJ, Chidlow G, Wood JPM, Crowston JG, Goldberg I. Definition of glaucoma: clinical and experimental concepts. Clin Exp Ophthalmol 2012; 40:341-9. [PMID: 22356435 DOI: 10.1111/j.1442-9071.2012.02773.x] [Citation(s) in RCA: 300] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Glaucoma is a term describing a group of ocular disorders with multi-factorial etiology united by a clinically characteristic intraocular pressure-associated optic neuropathy. It is not a single entity and is sometimes referred to in the plural as the glaucomas. All forms are potentially progressive and can lead to blindness. The diverse conditions that comprise glaucoma are united by a clinically characteristic optic neuropathy: glaucomatous optic neuropathy (GON). Evidence suggests that the primary site of neurological injury is at the optic nerve head. This fact enables the conditions to be grouped, irrespective of the causal mechanism(s). The term experimental glaucoma implies model resemblance to the human condition. We propose that 'experimental glaucoma' be restricted to animal models with demonstrable features of GON and/or evidence of a primary axonopathy at the optic nerve head. A fundamental inadequacy in this framework is any reference to the pathogenesis of GON, which remains unclear.
Collapse
Affiliation(s)
- Robert J Casson
- South Australian Institute of Ophthalmology, Hanson Institute and Adelaide University, Adelaide, South Australia, Australia.
| | | | | | | | | |
Collapse
|
22
|
Tam ALC, Gupta N, Zhang Z, Yücel YH. Quantum dots trace lymphatic drainage from the mouse eye. NANOTECHNOLOGY 2011; 22:425101. [PMID: 21934199 DOI: 10.1088/0957-4484/22/42/425101] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Glaucoma is a leading cause of blindness in the world, often associated with elevated eye pressure. Currently, all glaucoma treatments aim to lower eye pressure by improving fluid exit from the eye. We recently reported the presence of lymphatics in the human eye. The lymphatic circulation is known to drain fluid from organ tissues and, as such, lymphatics may also play a role in draining fluid from the eye. We investigated whether lymphatic drainage from the eye is present in mice by visualizing the trajectory of quantum dots once injected into the eye. Whole-body hyperspectral fluorescence imaging was performed in 17 live mice. In vivo imaging was conducted prior to injection, and 5, 20, 40 and 70 min, and 2, 6 and 24 h after injection. A quantum dot signal was observed in the left neck region at 6 h after tracer injection into the eye. Examination of immunofluorescence-labelled sections using confocal microscopy showed the presence of a quantum dot signal in the left submandibular lymph node. This is the first direct evidence of lymphatic drainage from the mouse eye. The use of quantum dots to image this lymphatic pathway in vivo is a novel tool to stimulate new treatments to reduce eye pressure and prevent blindness from glaucoma.
Collapse
Affiliation(s)
- Alex L C Tam
- Department of Ophthalmology and Vision Sciences, University of Toronto, M5T 2S8, Canada
| | | | | | | |
Collapse
|
23
|
Chou TH, Kocaoglu OP, Borja D, Ruggeri M, Uhlhorn SR, Manns F, Porciatti V. Postnatal elongation of eye size in DBA/2J mice compared with C57BL/6J mice: in vivo analysis with whole-eye OCT. Invest Ophthalmol Vis Sci 2011; 52:3604-12. [PMID: 21372015 DOI: 10.1167/iovs.10-6340] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE To characterize postnatal changes in eye size in glaucomatous DBA/2J (D2) mice and in nonglaucomatous C57BL/6J mice (B6) in vivo by means of whole-eye optical coherence tomography (OCT). METHODS D2 (n = 32) and B6 (n = 36) mice were tested between 2 and 20 months of age in eight age bins. A custom time-domain OCT system with a center wavelength of 825 nm and an axial scan length of 7.1 mm produced axial A-scan interferograms at a rate of 20 A-lines/s with a resolution of 8 μm. Axial length (AL), corneal thickness (CT), anterior chamber depth (ACD), lens thickness (LT), vitreous chamber depth (VCD), and retinal thickness (RT) were measured in the optical axis and adjusted with corresponding refractive indices. Corneal curvature (CC) and IOP were also measured. RESULTS AL increased (P < 0.001) more in the D2 (21%) than in the B6 (9%) mice. There was an interaction effect (two-way ANOVA, P < 0.001) between age and strain for AL, CT, ACD, and VCD. In the D2 mice, the lens became dislocated posteriorly. Multiple regression analysis in the D2 mice revealed an independent effect of age and IOP (P ≤ 0.01) on axial length. CC steepened in the older D2 mice, whereas it flattened in the B6 mice. CONCLUSIONS In D2 mice, postnatal elongation of AL is larger than that in B6 mice and is associated with a greater increase in ACD and IOP, which seems to be a causal factor. The ease of use, short acquisition time, and noninvasiveness of whole-eye OCT make it suitable for routine use in longitudinal studies of mouse models.
Collapse
Affiliation(s)
- Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Gunn DJ, Gole GA, Barnett NL. Specific amacrine cell changes in an induced mouse model of glaucoma. Clin Exp Ophthalmol 2011; 39:555-63. [PMID: 21176046 DOI: 10.1111/j.1442-9071.2010.02488.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND To investigate retinal cell population changes under chronic elevated intraocular pressure in an inducible mouse model of glaucoma. METHODS Chronic unilateral ocular hypertension was induced in 40 C57BL6/J mice by ablation of the limbal episcleral veins. After 5, 20, 40 and 60 days of elevated intraocular pressure, specific retinal cell types were identified and/or quantified by immunohistochemistry for protein kinase C α, glial fibrillary acidic protein, parvalbumin and calretinin. Apoptotic cells were identified by TUNEL and cleaved caspase-3 immunohistochemistry. RESULTS Elevations in intraocular pressure in the range 22-30 mmHg were developed and sustained in mice for up to 60 days. Protein kinase C α immunoreactivity localized to bipolar cells was unchanged. We observed a rapid increase in glial fibrillary acidic protein expression in Müller cells and a progressive loss of parvalbumin-labelled ganglion cells. After 60 days of elevated intraocular pressure, calretinin-immunoreactive cell counts declined by 55.4% and 46.4% in the inner nuclear and ganglion cell layers, respectively. However, at all time points examined, the markers of cell death were only observed in the ganglion cell layer, not in the inner nuclear layer. CONCLUSIONS In addition to ganglion cell death and reactive Müller cell changes, chronic experimental elevation of intraocular pressure alters calcium-binding protein immunohistochemistry in amacrine cells. However, these changes are not indicative of amacrine cell loss but may represent early indicators of cellular distress that precede physiological dysfunction or cell death.
Collapse
Affiliation(s)
- David J Gunn
- The University of Queensland, Perinatal Research Centre, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
25
|
Elsmo EJ, Kiland JA, Kaufman PL, McLellan GJ. Evaluation of rebound tonometry in non-human primates. Exp Eye Res 2011; 92:268-73. [PMID: 21315069 DOI: 10.1016/j.exer.2011.01.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 01/10/2011] [Accepted: 01/28/2011] [Indexed: 11/30/2022]
Abstract
To determine the accuracy and reproducibility of intraocular pressure (IOP) measurements obtained with the TonoVet® rebound tonometer in cynomolgus macaques and to determine the effects of corneal thickness on measurements obtained by the TonoVet®. The anterior chambers of both eyes of anesthetized monkeys were cannulated with branched 23-G needles; one branch was connected to a vertically adjustable reservoir and the other to a pressure transducer. IOP was increased by 5 mmHg increments and then decreased by 10 mmHg decrements. IOP was measured using the TonoVet® at each increment and decrement by 2 independent observers and at every other increment and every decrement by a single observer using 'minified' Goldmann applanation tonometry. Central corneal thickness was measured with a PachPen(TM) ultrasonic pachymeter. TonoVet® readings correlated well with manometric IOP (slope = 0.972, r(2) coefficient = 0.955). No significant differences were observed when comparing eyes or operators; however there was a non-significant trend for TonoVet® readings taken in right eyes to be closer to manometric IOP than those taken in left eyes. The TonoVet® had a non-significant tendency to underestimate manometric IOP. TonoVet® readings obtained during the decremental phase of the experiment were significantly closer (p < 0.004) to manometric IOP than those obtained during the incremental phase. Central corneal thickness significantly increased (p < 0.0001) over the course of the experiment. The TonoVet® rebound tonometer is a reliable and accurate tool for the measurement of IOP in cynomolgus macaques. This tonometer has several advantages, including portability, ease of use, and brief contact with the corneal surface making topical anesthetics unnecessary.
Collapse
Affiliation(s)
- Elizabeth J Elsmo
- Dept. of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI 53792, USA
| | | | | | | |
Collapse
|
26
|
Johnson EC, Doser TA, Cepurna WO, Dyck JA, Jia L, Guo Y, Lambert WS, Morrison JC. Cell proliferation and interleukin-6-type cytokine signaling are implicated by gene expression responses in early optic nerve head injury in rat glaucoma. Invest Ophthalmol Vis Sci 2011; 52:504-18. [PMID: 20847120 DOI: 10.1167/iovs.10-5317] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE In glaucoma, the optic nerve head (ONH) is the principal site of initial axonal injury, and elevated intraocular pressure (IOP) is the predominant risk factor. However, the initial responses of the ONH to elevated IOP are unknown. Here the authors use a rat glaucoma model to characterize ONH gene expression changes associated with early optic nerve injury. METHODS Unilateral IOP elevation was produced in rats by episcleral vein injection of hypertonic saline. ONH mRNA was extracted, and retrobulbar optic nerve cross-sections were graded for axonal degeneration. Gene expression was determined by microarray and quantitative PCR (QPCR) analysis. Significantly altered gene expression was determined by multiclass analysis and ANOVA. DAVID gene ontology determined the functional categories of significantly affected genes. RESULTS The Early Injury group consisted of ONH from eyes with <15% axon degeneration. By array analysis, 877 genes were significantly regulated in this group. The most significant upregulated gene categories were cell cycle, cytoskeleton, and immune system process, whereas the downregulated categories included glucose and lipid metabolism. QPCR confirmed the upregulation of cell cycle-associated genes and leukemia inhibitory factor (Lif) and revealed alterations in expression of other IL-6-type cytokines and Jak-Stat signaling pathway components, including increased expression of IL-6 (1553%). In contrast, astrocytic glial fibrillary acidic protein (Gfap) message levels were unaltered, and other astrocytic markers were significantly downregulated. Microglial activation and vascular-associated gene responses were identified. CONCLUSIONS Cell proliferation and IL-6-type cytokine gene expression, rather than astrocyte hypertrophy, characterize early pressure-induced ONH injury.
Collapse
Affiliation(s)
- Elaine C Johnson
- Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon 97201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ren L, Danias J. A Role for Complement in Glaucoma? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 703:95-104. [DOI: 10.1007/978-1-4419-5635-4_7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
28
|
Dai Y, Lindsey JD, Duong-Polk X, Nguyen D, Hofer A, Weinreb RN. Outflow facility in mice with a targeted type I collagen mutation. Invest Ophthalmol Vis Sci 2009; 50:5749-53. [PMID: 19797236 PMCID: PMC6335031 DOI: 10.1167/iovs.08-3367] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Transgenic Col1a1(r/r) mice develop elevated intraocular pressure (IOP) with an open angle and progressive optic nerve axon loss. The present study was undertaken to evaluate aqueous outflow facility and its age dependence in these mice. METHODS Homozygous B6;129S4-Col1a1(tm1Jae) mice and corresponding wild-type Col1a1(+/+) mice from 12 to 56 weeks of age were anesthetized, and IOP was measured with a microneedle. Outflow facility was determined by a two-level, constant-pressure infusion METHOD Type I collagen, subunit alpha1 was assessed in sclera and choroid by Western blot analysis. RESULTS The mean IOP in 12- to 36-week-old transgenic Col1a1(r/r) mice was 25.1% higher than in control Col1a1(+/+) mice (P < 0.01), whereas the mean outflow facility was 25.4% lower than in control mice (P < 0.01). After this period, the mean IOP in 42- to 56-week-old transgenic mice returned to normal levels, whereas outflow facility increased by 36.0%. Over the 12- to 56-week study period, IOP and outflow facility in the transgenic mice were inversely correlated (r(2) = -0.702, P < 0.01). Collagen I alpha1 content was greater in 37- and 43-week-old transgenic mice than in age-matched wild-type control mice. CONCLUSIONS Outflow facility is reduced in transgenic Col1a1(r/r) mice with IOP elevation. The inverse correlation of IOP elevation to facility reduction indicates that increased resistance in the aqueous outflow pathway contributes to ocular hypertension in Col1a1(r/r) mice. These mice may be useful as a model for open-angle glaucoma, as well as for assessing the relationship between collagen type I metabolism and aqueous outflow.
Collapse
Affiliation(s)
- Yi Dai
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, California
- Department of Ophthalmology and Vision Science, EYE & ENT Hospital, Fudan University, Shanghai, China
| | - James D. Lindsey
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, California
| | - Xuandao Duong-Polk
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, California
| | - Duy Nguyen
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, California
| | - Anthony Hofer
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, California
| | - Robert N. Weinreb
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, California
| |
Collapse
|
29
|
Fu CT, Sretavan D. Laser-induced ocular hypertension in albino CD-1 mice. Invest Ophthalmol Vis Sci 2009; 51:980-90. [PMID: 19815738 DOI: 10.1167/iovs.09-4324] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To establish a laser-induced model of ocular hypertension (LIOH) in albino CD-1 mice and to characterize the sequence of pathologic events triggered by intraocular pressure (IOP) elevation. METHODS LIOH was induced unilaterally in CD-1 mice by laser photocoagulation of limbal and episcleral veins 270 degrees to 300 degrees circumferentially, sparing the nasal aspect and the long ciliary arteries. IOP was measured with a rebound tonometer. Hematoxylin and eosin-stained plastic sections were used for morphometric analysis of retinal layers, and retinal whole-mounts were immunostained with anti-Brn-3b to quantify retinal ganglion cell (RGC) gene expression ion and density. Axonal and myelin morphologies were characterized using appropriate antibodies, and axon counts were obtained from paraphenylenediamine-stained optic nerve sections. RESULTS LIOH resulted in IOP doubling within 4 hours after laser treatment, which returned to normal by 7 days. Axon degenerative changes, reactive plasticity, and aberrant regrowth were detected at the optic nerve head (ONH) as early as 4 days after treatment. By 7 days, axon number was significantly reduced in the myelinated optic nerve, with concurrent signs of myelin degradation. At 14 days, Brn-3b(+) RGC density was reduced, with neuronal loss confined to the RGC layer and no apparent effects on other retinal layers. CONCLUSIONS Laser photocoagulation of limbal and episcleral veins induces transient ocular hypertension in albino CD-1 mice. The ensuing retinal and optic nerve pathologic events recapitulated key features of glaucoma and placed ONH RGC axon responses as an early manifestation of damage. LIOH in albino mice may be useful as a mouse model to examine mechanisms of RGC and axon glaucomatous injury.
Collapse
Affiliation(s)
- Christine T Fu
- Neuroscience Graduate Program, Department of Ophthalmology, University of California, San Francisco, San Francisco, California 94143, USA.
| | | |
Collapse
|
30
|
Howell GR, Libby RT, John SWM. Mouse genetic models: an ideal system for understanding glaucomatous neurodegeneration and neuroprotection. PROGRESS IN BRAIN RESEARCH 2009; 173:303-21. [PMID: 18929118 DOI: 10.1016/s0079-6123(08)01122-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Here we review how mouse studies are contributing to understanding glaucoma. We include discussion of aqueous humor drainage and intraocular pressure elevation, because new treatments to avoid exposure to high pressure will indirectly protect neurons from glaucoma, and complement direct neuroprotective strategies. We describe how mouse models are adding to both the understanding of glaucomatous neurodegeneration and the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Gareth R Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | | |
Collapse
|
31
|
Kompass KS, Agapova OA, Li W, Kaufman PL, Rasmussen CA, Hernandez MR. Bioinformatic and statistical analysis of the optic nerve head in a primate model of ocular hypertension. BMC Neurosci 2008; 9:93. [PMID: 18822132 PMCID: PMC2567987 DOI: 10.1186/1471-2202-9-93] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Accepted: 09/26/2008] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The nonhuman primate model of glaucomatous optic neuropathy most faithfully reproduces the human disease. We used high-density oligonucleotide arrays to investigate whole genome transcriptional changes occurring at the optic nerve head during primate experimental glaucoma. RESULTS Laser scarification of the trabecular meshwork of cynomolgus macaques produced elevated intraocular pressure that was monitored over time and led to varying degrees of damage in different samples. The macaques were examined clinically before enucleation and the myelinated optic nerves were processed post-mortem to determine the degree of neuronal loss. Global gene expression was examined in dissected optic nerve heads with Affymetrix GeneChip microarrays. We validated a subset of differentially expressed genes using qRT-PCR, immunohistochemistry, and immuno-enriched astrocytes from healthy and glaucomatous human donors. These genes have previously defined roles in axonal outgrowth, immune response, cell motility, neuroprotection, and extracellular matrix remodeling. CONCLUSION Our findings show that glaucoma is associated with increased expression of genes that mediate axonal outgrowth, immune response, cell motility, neuroprotection, and ECM remodeling. These studies also reveal that, as glaucoma progresses, retinal ganglion cell axons may make a regenerative attempt to restore lost nerve cell contact.
Collapse
Affiliation(s)
- Kenneth S Kompass
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Olga A Agapova
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenjun Li
- Department of Ophthalmology, Northwestern University, Chicago, IL 60611, USA
| | - Paul L Kaufman
- Department of Ophthalmology and Visual Sciences, University of Wisconsin Medical School, Madison, WI 53792, USA
| | - Carol A Rasmussen
- Department of Ophthalmology and Visual Sciences, University of Wisconsin Medical School, Madison, WI 53792, USA
| | - M Rosario Hernandez
- Department of Ophthalmology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
32
|
Kasper K, Kremling C, Geerling G. [Toxicity of a new moistening agent and preservative in vitro]. Ophthalmologe 2008; 105:557-62. [PMID: 18214492 DOI: 10.1007/s00347-007-1640-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE The use of preservatives such as benzalkonium chloride (BAC) usually increases the toxicity of pharmaceutical tear substitutes. HP-guar has been recently introduced as a new artificial tear substitute and includes the preservative Polyquad (0.001%), which is considered to be non-toxic. We therefore examined the effect of preserved (cetrimide 0.01%) and unpreserved HPMC (hydroxypropylmethyl cellulose) and HP-guar in dose and time-response experiments in a human corneal and conjunctival epithelial cell culture model. METHODS Immortalized human conjunctival and corneal epithelial cells were cultured in 96-well plates at 37 degrees C with 5% CO(2) and exposed to the test solutions. The ATP content was quantified by means of a luminescence-based ATP assay, intracellular esterase activity by double fluorescent viability staining (calcein AM/ethidium homodimer D-1) and cell migration by a colony dispersion assay. All experiments were performed in triplicate and repeated at least once. The significance of differences was determined with an unpaired two-sided t-test. RESULTS HPMC with preservative severely reduced the ATP content at all concentrations tested. Unpreserved HPMC, however, showed an inhibition of ATP production only at 100% and good esterase activity. HP-guar with and without preservative were found to reduce ATP activity more than unpreserved HPMC, but the unpreserved solution was found to reduce cellular ATP levels significantly more than the preserved solution. CONCLUSIONS The new preservative Polyquad induced significantly less cytotoxicity than cetrimide. However, even unpreserved HP-guar can induce cytotoxicity in vitro, while unpreserved HPMC remains a good alternative tear substitute with low cytotoxicity.
Collapse
Affiliation(s)
- K Kasper
- Augen- und Poliklinik, Universität Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Deutschland
| | | | | |
Collapse
|
33
|
Anderson MG, Nair KS, Amonoo LA, Mehalow A, Trantow CM, Masli S, John SWM. GpnmbR150X allele must be present in bone marrow derived cells to mediate DBA/2J glaucoma. BMC Genet 2008; 9:30. [PMID: 18402690 PMCID: PMC2373794 DOI: 10.1186/1471-2156-9-30] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Accepted: 04/10/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Gpnmb gene encodes a transmembrane protein whose function(s) remain largely unknown. Here, we assess if a mutant allele of Gpnmb confers susceptibility to glaucoma by altering immune functions. DBA/2J mice have a mutant Gpnmb gene and they develop a form of glaucoma preceded by a pigment dispersing iris disease and abnormalities of the immunosuppressive ocular microenvironment. RESULTS We find that the Gpnmb genotype of bone-marrow derived cell lineages significantly influences the iris disease and the elevation of intraocular pressure. GPNMB localizes to multiple cell types, including pigment producing cells, bone marrow derived F4/80 positive antigen-presenting cells (APCs) of the iris and dendritic cells. We show that APCs of DBA/2J mice fail to induce antigen induced immune deviation (a form of tolerance) when treated with TGFbeta2. This demonstrates that some of the immune abnormalities previously identified in DBA/2J mice result from intrinsic defects in APCs. However, the tested APC defects are not dependent on a mutant Gpnmb gene. Finally, we show that the Gpnmb mediated iris disease does not require elevated IL18 or mature B or T lymphocytes. CONCLUSION These results establish a role for Gpnmb in bone marrow derived lineages. They suggest that affects of Gpnmb on innate immunity influence susceptibility to glaucoma in DBA/2J mice.
Collapse
Affiliation(s)
- Michael G Anderson
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Nickells RW, Semaan SJ, Schlamp CL. Involvement of the Bcl2 gene family in the signaling and control of retinal ganglion cell death. PROGRESS IN BRAIN RESEARCH 2008; 173:423-35. [DOI: 10.1016/s0079-6123(08)01129-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
35
|
Glaucoma of the brain: a disease model for the study of transsynaptic neural degeneration. PROGRESS IN BRAIN RESEARCH 2008; 173:465-78. [PMID: 18929128 DOI: 10.1016/s0079-6123(08)01132-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The identification of mechanisms precipitating neuronal death and injury is an intense area of investigation requiring reliable models to assess the effects of neuroprotective agents. Most are suboptimal since the effects of initial damage are diffuse and may not be reproducible or easily quantifiable. The ideal laboratory model should have the ability to (a) clearly detect evidence of neuronal injury and recovery, (b) accurately measure morphologically the extent of these changes, and (c) provide functional evidence for damage and recovery. Glaucoma is a disease of visual neurons in the eye and brain. In the visual system, neuroanatomical pathways and retinotopic organization are exquisitely defined, functional modalities are highly characterized and can be dissected physiologically, visual input parameters can be modified, visual functional output can be readily tested and measured, changes in the eye and the visual brain can be directly visualized and imaged, and pathological and compensatory changes in brain centers of vision can be examined and measured specifically. For these reasons, the glaucoma disease model is ideal for the study of response and recovery to injury in the central nervous system due to anterograde and retrograde degeneration from the eye to the brain and the brain to the eye, respectively. The study of this glaucoma model of transsynaptic brain injury may be relevant to understanding more complex pathways and point to new strategies to prevent disease progression in other neurodegenerative diseases.
Collapse
|
36
|
|
37
|
Abstract
Animal models are useful to elucidate the etiology and pathology of glaucoma and to develop novel and more effective therapies for the disease. Because of the substantial similarities between the rodent and primate eyes, and the advances of relevant study techniques, rat and mouse models of glaucoma have recently become popular as research tools. This review surveys research techniques used in the measurement of rodent intraocular pressure, and also the evaluation of pertinent morphologic, biochemical, and functional changes in the retina, optic nerve head, and optic nerve. This review further describes in detail the individual rodent models, some of which serve as surrogate models and do not entail ocular hypertension, whereas others involve transient or chronic increases of intraocular pressure. The technical considerations and theoretical concerns of these models, their advantages, and limitations, are also discussed.
Collapse
Affiliation(s)
- Iok-Hou Pang
- Glaucoma Research, Alcon Research, Ltd, Fort Worth, TX, USA.
| | | |
Collapse
|
38
|
Senatorov V, Malyukova I, Fariss R, Wawrousek EF, Swaminathan S, Sharan SK, Tomarev S. Expression of mutated mouse myocilin induces open-angle glaucoma in transgenic mice. J Neurosci 2006; 26:11903-14. [PMID: 17108164 PMCID: PMC6674879 DOI: 10.1523/jneurosci.3020-06.2006] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We developed a genetic mouse model of open-angle glaucoma by expression of mutated mouse myocilin (Myoc) in transgenic (Tg) mice. The Tyr423His point mutation, corresponding to the severe glaucoma-causing Tyr437His mutation in the human MYOC gene, was introduced into bacterial artificial chromosome DNA encoding the full-length mouse Myoc gene and long flanking regions. Both wild-type (Wt) and Tg animals expressed Myoc in tissues of the irido-corneal angle and the sclera. Expression of mutated Myoc induced the accumulation of Myoc in cell cytoplasm and prevented its secretion into the extracellular space. The levels of ATPase-1 were reduced in the irido-corneal angle of Tg mice compared with Wt animals. Tg mice demonstrated a moderate elevation of intraocular pressure, the loss of approximately 20% of the retinal ganglion cells (RGCs) in the peripheral retina, and axonal degeneration in the optic nerve. RGC depletion was associated with the shrinkage of their nuclei and DNA fragmentation in the peripheral retina. Pathological changes observed in the eyes of Tg mice are similar to those observed in glaucoma patients.
Collapse
Affiliation(s)
- Vladimir Senatorov
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Molecular and Developmental Biology, and
| | - Irina Malyukova
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Molecular and Developmental Biology, and
| | - Robert Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Eric F. Wawrousek
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Molecular and Developmental Biology, and
| | - Srividya Swaminathan
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702
| | - Shyam K. Sharan
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702
| | - Stanislav Tomarev
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Molecular and Developmental Biology, and
| |
Collapse
|
39
|
Kim CY, Kuehn MH, Anderson MG, Kwon YH. Intraocular pressure measurement in mice: a comparison between Goldmann and rebound tonometry. Eye (Lond) 2006; 21:1202-9. [PMID: 16946746 DOI: 10.1038/sj.eye.6702576] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The development of mouse models of glaucoma requires methods to accurately measure the intraocular pressure (IOP) in this species. The aim of this study was to compare the accuracy of IOP measurements in mice between modified Goldmann and rebound tonometers. METHODS IOP was measured either with a modified Goldmann or a rebound tonometer while simultaneously measuring the IOP using invasive manometry in enucleated eyes and in vivo. The level of IOP was controlled hydrostatically. The agreement and correlation between the IOP determined by invasive manometry and by either noninvasive method was evaluated. In addition, the IOP was determined by both noninvasive methods in a cohort of mice with laser-induced ocular hypertension (OHT), and the agreement and correlation between the two tonometry methods were evaluated. RESULTS Measured IOP by either noninvasive tonometer correlated well with those recorded simultaneously by invasive manometry (r=0.98 for rebound and r=0.94 for Goldmann). In mice with OHT, the IOP correlation between rebound and modified Goldmann was moderate (r=0.71); the IOP measured by modified Goldmann tonometry was consistently higher than that by rebound by approximately 5 mmHg. However, the relative per cent increases in IOP were similar between the two methods. CONCLUSION Both noninvasive methods of IOP measurements in mice are suitable to detect changes in IOP although rebound tonometry correlated better with the invasive manometry readings. The results suggest that the relative, rather than absolute, IOP offers a more reliable means of correlating findings from studies using different tonometers.
Collapse
Affiliation(s)
- C Y Kim
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA, USA
| | | | | | | |
Collapse
|
40
|
Harvey AR, Hu Y, Leaver SG, Mellough CB, Park K, Verhaagen J, Plant GW, Cui Q. Gene therapy and transplantation in CNS repair: The visual system. Prog Retin Eye Res 2006; 25:449-89. [PMID: 16963308 DOI: 10.1016/j.preteyeres.2006.07.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Normal visual function in humans is compromised by a range of inherited and acquired degenerative conditions, many of which affect photoreceptors and/or retinal pigment epithelium. As a consequence the majority of experimental gene- and cell-based therapies are aimed at rescuing or replacing these cells. We provide a brief overview of these studies, but the major focus of this review is on the inner retina, in particular how gene therapy and transplantation can improve the viability and regenerative capacity of retinal ganglion cells (RGCs). Such studies are relevant to the development of new treatments for ocular conditions that cause RGC loss or dysfunction, for example glaucoma, diabetes, ischaemia, and various inflammatory and neurodegenerative diseases. However, RGCs and associated central visual pathways also serve as an excellent experimental model of the adult central nervous system (CNS) in which it is possible to study the molecular and cellular mechanisms associated with neuroprotection and axonal regeneration after neurotrauma. In this review we present the current state of knowledge pertaining to RGC responses to injury, neurotrophic and gene therapy strategies aimed at promoting RGC survival, and how best to promote the regeneration of RGC axons after optic nerve or optic tract injury. We also describe transplantation methods being used in attempts to replace lost RGCs or encourage the regrowth of RGC axons back into visual centres in the brain via peripheral nerve bridges. Cooperative approaches including novel combinations of transplantation, gene therapy and pharmacotherapy are discussed. Finally, we consider a number of caveats and future directions, such as problems associated with compensatory sprouting and the reformation of visuotopic maps, the need to develop efficient, regulatable viral vectors, and the need to develop different but sequential strategies that target the cell body and/or the growth cone at appropriate times during the repair process.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, WA 6009, Australia
| | | | | | | | | | | | | | | |
Collapse
|
41
|
MacLaren RE, Buch PK, Smith AJ, Balaggan KS, MacNeil A, Taylor JS, Osborne NN, Ali RR. CNTF gene transfer protects ganglion cells in rat retinae undergoing focal injury and branch vessel occlusion. Exp Eye Res 2006; 83:1118-27. [PMID: 16831422 DOI: 10.1016/j.exer.2006.05.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2005] [Revised: 04/20/2006] [Accepted: 05/26/2006] [Indexed: 11/29/2022]
Abstract
Ciliary neurotrophic factor (CNTF) has been shown to protect ganglion cells in a variety of acute ischaemia models. Here we assess the efficacy of local CNTF gene transfer in protecting retinal ganglion cells when there is focal ischaemia combined with interruption of axoplasmic flow. This dual injury may be more representative of the pathological mechanisms operating in acute retinal diseases, such as vascular events acting at the optic nerve head. Fourteen rats received an intravitreal injection of an adeno-associated viral (AAV) vector expressing a secretable form of CNTF into the right eye and a control vector into the left eye. Three weeks later, each rat underwent a symmetrical small vertical 2mm standardised retinal crush injury approximately 2mm temporal to the optic disc. The injury also occluded the temporal retinal arteriole so that the axon crush was combined with an acute retinal infarction visible on fundoscopy. Changes in the damaged sector were compared histologically four weeks after injury and ganglion cell survival was estimated by comparing cell counts on retinal flat-mounts immunostained with RT-97 antibody. This mode of injury led to a profound loss of both the inner nuclear and ganglion cell layers, but was limited to the lesioned sector. In AAV.CNTF-treated eyes approximately 12% of ganglion cells survived compared with approximately 2% in control eyes (p=0.01). The scotopic electroretinogram (ERG), however, was reduced to about 50% in AAV.CNTF-treated eyes, both before and after injury. We therefore show that CNTF gene transfer confers neuroprotection to ganglion cells undergoing an acute ischaemic injury combined with interruption of axoplasmic flow. This approach may be relevant to optic nerve trauma and a variety of retinal vascular diseases that lead to swelling of the optic nerve head, provided CNTF can be delivered in a way that does not significantly suppress retinal function.
Collapse
Affiliation(s)
- Robert E MacLaren
- Division of Molecular Therapy, Institute of Ophthalmology, University College London, London EC1V 9EL, UK.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Anderson MG, Libby RT, Mao M, Cosma IM, Wilson LA, Smith RS, John SWM. Genetic context determines susceptibility to intraocular pressure elevation in a mouse pigmentary glaucoma. BMC Biol 2006; 4:20. [PMID: 16827931 PMCID: PMC1543659 DOI: 10.1186/1741-7007-4-20] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 07/07/2006] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND DBA/2J (D2) mice develop an age-related form of glaucoma. Their eyes progressively develop iris pigment dispersion and iris atrophy followed by increased intraocular pressure (IOP) and glaucomatous optic nerve damage. Mutant alleles of the Gpnmb and Tyrp1 genes are necessary for the iris disease, but it is unknown whether alleles of other D2 gene(s) are necessary for the distinct later stages of disease. We initiated a study of congenic strains to further define the genetic requirements and disease mechanisms of the D2 glaucoma. RESULTS To further understand D2 glaucoma, we created congenic strains of mice on the C57BL/6J (B6) genetic background. B6 double-congenic mice carrying D2-derived Gpnmb and Tyrp1 mutations develop a D2-like iris disease. B6 single-congenics with only the Gpnmb and Tyrp1 mutations develop milder forms of iris disease. Genetic epistasis experiments introducing a B6 tyrosinase mutation into the congenic strains demonstrated that both the single and double-congenic iris diseases are rescued by interruption of melanin synthesis. Importantly, our experiments analyzing mice at ages up to 27 months indicate that the B6 double-congenic mice are much less prone to IOP elevation and glaucoma than are D2 mice. CONCLUSION As demonstrated here, the Gpnmb and Tyrp1 iris phenotypes are both individually dependent on tyrosinase function. These results support involvement of abnormal melanosomal events in the diseases caused by each gene. In the context of the inbred D2 mouse strain, the glaucoma phenotype is clearly influenced by more genes than just Gpnmb and Tyrp1. Despite the outward similarity of pigment-dispersing iris disease between D2 and the B6 double-congenic mice, the congenic mice are much less susceptible to developing high IOP and glaucoma. These new congenic strains provide a valuable new resource for further studying the genetic and mechanistic complexity of this form of glaucoma.
Collapse
Affiliation(s)
- Michael G Anderson
- The Jackson Laboratory, Bar Harbor, ME, USA
- Department of Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | | | - Mao Mao
- Department of Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | | | | | | | - Simon WM John
- The Jackson Laboratory, Bar Harbor, ME, USA
- Howard Hughes Medical Institute, Bar Harbor, ME, USA
- Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|