1
|
Fradot V, Augustin S, Fontaine V, Marazova K, Guillonneau X, Sahel JA, Picaud S. Rodent Models of Retinal Degeneration: From Purified Cells in Culture to Living Animals. Cold Spring Harb Perspect Med 2024; 14:a041311. [PMID: 37848250 PMCID: PMC11444255 DOI: 10.1101/cshperspect.a041311] [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: 10/19/2023]
Abstract
Rodent models of retinal degeneration are essential for the development of therapeutic strategies. In addition to living animal models, we here also discuss models based on rodent cell cultures, such as purified retinal ganglion cells and retinal explants. These ex vivo models extend the possibilities for investigating pathological mechanisms and assessing the neuroprotective effect of pharmacological agents by eliminating questions on drug pharmacokinetics and bioavailability. The number of living rodent models has greatly increased with the possibilities to achieve transgenic modifications in animals for knocking in and out genes and mutations. The Cre-lox system has further enabled investigators to target specific genes or mutations in specific cells at specific stages. However, chemically or physically induced models can provide alternatives to such targeted gene modifications. The increased diversity of rodent models has widened our possibility to address most ocular pathologies for providing initial proof of concept of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Valérie Fradot
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Sébastien Augustin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Valérie Fontaine
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Katia Marazova
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - José A Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| |
Collapse
|
2
|
Jiang Z, Chernoff D, Galenchik-Chan A, Tomorri D, Honkanen RA, Duong TQ, Muir ER. Improved MRI methods to quantify retinal and choroidal blood flow applied to a model of glaucoma. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1385495. [PMID: 38984144 PMCID: PMC11182105 DOI: 10.3389/fopht.2024.1385495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/23/2024] [Indexed: 07/11/2024]
Abstract
Purpose Blood flow (BF) of the retinal and choroidal vasculatures can be quantitatively imaged using MRI. This study sought to improve methods of data acquisition and analysis for MRI of layer-specific retinal and choroidal BF and then applied this approach to detect reduced ocular BF in a well-established mouse model of glaucoma from both eyes. Methods Quantitative BF magnetic resonance imaging (MRI) was performed on glaucomatous DBA/2J and normal C57BL/6J mice. Arterial spin labeling MRI was applied to image retinal and choroidal BF using custom-made dual eye coils that could image both eyes during the same scan. Statistics using data from a single eye or two eyes were compared. BF values were calculated using two approaches. The BF rate per quantity of tissue was calculated as commonly done, and the peak BF values of the retinal and choroidal vasculatures were taken. Additionally, the BF rate per retinal surface area was calculated using a new analysis approach to attempt to reduce partial volume and variability by integrating BF over the retinal and choroidal depths. Results Ocular BF of both eyes could be imaged using the dual coil setup without effecting scan time. Intraocular pressure was significantly elevated in DBA/2J mice compared to C57BL/6J mice (P<0.01). Both retinal and choroidal BF were significantly decreased in DBA/2J mice in comparison to the age-matched normal C57BL/6J mice across all measurements (P < 0.01). From simulations, the values from the integrated BF analysis method had less partial volume effect, and from in vivo scans, this analysis approach also improved power. Conclusion The dual eye coil setup allows bilateral eye data acquisition, increasing the amount of data acquired without increasing acquisition times in vivo. The reduced ocular BF found using the improved acquisition and analysis approaches replicated the results of previous studies on DBA/2J mice. The ocular hypertensive stress-induced BF reduction found within these mice may represent changes associated with glaucomatous progression.
Collapse
Affiliation(s)
- Zhao Jiang
- Department of Radiology, Stony Brook University, Stony Brook, NY, United States
| | - Diane Chernoff
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Andre Galenchik-Chan
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - David Tomorri
- School of Health Professions, Stony Brook University, Stony Brook, NY, United States
| | - Robert A. Honkanen
- Department of Ophthalmology, Stony Brook University, Stony Brook, NY, United States
| | - Timothy Q. Duong
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Eric R. Muir
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Fraenkl SA, Simon Q, Yucel Y, Gupta N, Wittwer VV, Frueh BE, Tschanz SA. Impact of cerebral hypoperfusion-reperfusion on optic nerve integrity and visual function in the DBA/2J mouse model of glaucoma. BMJ Open Ophthalmol 2022; 7:bmjophth-2022-001078. [PMID: 36161839 PMCID: PMC9476133 DOI: 10.1136/bmjophth-2022-001078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
Objective One of the most important risk factors for developing a glaucomatous optic neuropathy is elevated intraocular pressure. Moreover, mechanisms such as altered perfusion have been postulated to injure the optical path. In a mouse model, we compare first negative effects of cerebral perfusion/reperfusion on the optic nerve structure versus alterations by elevated intraocular pressure. Second, we compare the alterations by isolated hypoperfusion-reperfusion and isolated intraocular pressure to the combination of both. Methods and analysis Mice were divided in four groups: (1) controls; (2) perfusion altered mice that underwent transient bi-common carotid artery occlusion (BCCAO) for 40 min; (3) glaucoma group (DBA/2J mice); (4) combined glaucoma and altered perfusion (DBA/2J mice with transient BCCAO). Optic nerve sections were stereologically examined 10–12 weeks after intervention. Results All experimental groups showed a decreased total axon number per optic nerve compared with controls. In DBA/2J and combined DBA/2J & BCCAO mice the significant decrease was roughly 50%, while BCCAO leaded to a 23% reduction of axon number, however reaching significance only in the direct t-test. The difference in axon number between BCCAO and both DBA/2J mice was almost 30%, lacking statistical significance due to a remarkably high variation in both DBA/2J groups. Conclusion Elevated intraocular pressure in the DBA/2J mouse model of glaucoma leads to a much more pronounced optic nerve atrophy compared with transient forebrain hypoperfusion and reperfusion by BCCAO. A supposed worsening effect of an altered perfusion added to the pressure-related damage could not be detected.
Collapse
Affiliation(s)
| | - Quentin Simon
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Yeni Yucel
- Keenan Research Centre, St Michael's Hospital Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada.,Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Neeru Gupta
- Keenan Research Centre, St Michael's Hospital Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada.,Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Valéry V Wittwer
- Department of Ophthalmology, Inselspital, University of Bern, Bern, Switzerland.,Ophthalmologic Network Organization (ONO), Geneva, Switzerland
| | - Beatrice E Frueh
- Department of Ophthalmology, Inselspital, University of Bern, Bern, Switzerland
| | | |
Collapse
|
4
|
Strickland RG, Garner MA, Gross AK, Girkin CA. Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma. Int J Mol Sci 2022; 23:8068. [PMID: 35897642 PMCID: PMC9329908 DOI: 10.3390/ijms23158068] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022] Open
Abstract
Glaucomatous optic neuropathy is the leading cause of irreversible blindness in the world. The chronic disease is characterized by optic nerve degeneration and vision field loss. The reduction of intraocular pressure remains the only proven glaucoma treatment, but it does not prevent further neurodegeneration. There are three major classes of cells in the human optic nerve head (ONH): lamina cribrosa (LC) cells, glial cells, and scleral fibroblasts. These cells provide support for the LC which is essential to maintain healthy retinal ganglion cell (RGC) axons. All these cells demonstrate responses to glaucomatous conditions through extracellular matrix remodeling. Therefore, investigations into alternative therapies that alter the characteristic remodeling response of the ONH to enhance the survival of RGC axons are prevalent. Understanding major remodeling pathways in the ONH may be key to developing targeted therapies that reduce deleterious remodeling.
Collapse
Affiliation(s)
- Ryan G. Strickland
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Mary Anne Garner
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Alecia K. Gross
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Christopher A. Girkin
- Department of Ophthalmology and Vision Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
5
|
Shalaby WS, Ahmed OM, Waisbourd M, Katz LJ. A Review of Potential Novel Glaucoma Therapeutic Options Independent of Intraocular Pressure. Surv Ophthalmol 2021; 67:1062-1080. [PMID: 34890600 DOI: 10.1016/j.survophthal.2021.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
Glaucoma, a progressive optic neuropathy characterized by retinal ganglion cell degeneration and visual field loss, is the leading cause of irreversible blindness worldwide. Intraocular pressure (IOP) is presently the only modifiable risk factor demonstrated to slow or halt disease progression; however, glaucomatous damage persists in almost 50% of patients despite significant IOP reduction. Many studies have investigated the non-IOP-related risk factors that contribute to glaucoma progression as well as interventions that can prevent or delay glaucomatous neurodegeneration and preserve vision throughout life, independently of IOP. A vast number of experimental studies have reported effective neuroprotection in glaucoma, and clinical studies are ongoing attempting to provide strong evidence of effectiveness of these interventions. In this review, we look into the current understanding of the pathophysiology of glaucoma and explore the recent advances in non-IOP related strategies for neuroprotection and neuroregeneration in glaucoma.
Collapse
Key Words
- AMD, Age-related macular degeneration
- BDNF, Brain derived neurotrophic factor
- CNTF, Ciliary neurotrophic factor
- GDNF, Glial‐derived neurotrophic factor
- Glaucoma
- IOP, Intraocular pressure
- LoGTS, Low-Pressure Glaucoma Treatment Study
- MRI, Magnetic resonance imaging
- MSCs, Mesenchymal stem cells
- NGF, Nerve growth factor
- NTG, Normal tension glaucoma
- OCTA, Optical coherence tomography angiography
- PBM, hotobiomodulation
- PDGF, Platelet derived growth factor
- POAG, Primary open angle glaucoma
- RGCs, Retinal ganglion cells
- TNF-α, Tumor necrosis factor- α
- bFGF, Basic fibroblast growth factor
- gene therapy
- intracranial pressure
- intraocular pressure
- neuroprotection
- ocular blood flow
- oxidative stress
- retinal ganglion cells
- stem cell therapy
Collapse
Affiliation(s)
- Wesam Shamseldin Shalaby
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Department of Ophthalmology, Tanta Medical School, Tanta University, Tanta, Gharbia, Egypt
| | - Osama M Ahmed
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Yale University School of Medicine, New Haven, CT, USA
| | - Michael Waisbourd
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Department of Ophthalmology, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - L Jay Katz
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Sato K, Sato T, Ohno-Oishi M, Ozawa M, Maekawa S, Shiga Y, Yabana T, Yasuda M, Himori N, Omodaka K, Fujita K, Nishiguchi KM, Ge S, Nakazawa T. CHOP deletion and anti-neuroinflammation treatment with hesperidin synergistically attenuate NMDA retinal injury in mice. Exp Eye Res 2021; 213:108826. [PMID: 34752818 DOI: 10.1016/j.exer.2021.108826] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/26/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022]
Abstract
Glaucoma is a leading cause of blindness worldwide and is characterized by degeneration associated with the death of retinal ganglion cells (RGCs). It is believed that glaucoma is a group of heterogeneous diseases with multifactorial pathomechanisms. Here, we investigate whether anti-inflammation treatment with an ER stress blockade can selectively promote neuroprotection against NMDA injury in the RGCs. Retinal excitotoxicity was induced with an intravitreal NMDA injection. Microglial activation and neuroinflammation were evaluated with Iba1 immunostaining and cytokine gene expression. A stable HT22 cell line transfected with an NF-kB reporter was used to assess NF-kB activity after hesperidin treatment. CHOP-deficient mice were used as a model of ER stress blockade. Retinal cell death was evaluated with a TUNEL assay. As results, in the NMDA injury group, Iba1-positive microglia increased 6 h after NMDA injection. Also at 6 h, pro-inflammatory cytokines and chemokine increased, including TNFα, IL-1b, IL-6 and MCP-1. In addition, the MCP-1 promoter-driven EGFP signal, which we previously identified as a stress signal in injured RGCs, also increased; hesperidin treatment suppressed this inflammatory response and reduced stressed RGCs. In CHOP-deficient mice that received an NMDA injection, the gene expression of pro-inflammatory cytokines, chemokines, markers of active microglia, and inflammatory regulators was greater than in WT mice. In WT mice, hesperidin treatment partially prevented retinal cell death after NMDA injury; this neuroprotective effect was enhanced in CHOP-deficient mice. These findings demonstrate that ER stress blockade is not enough by itself to prevent RGC loss due to neuroinflammation in the retina, but it has a synergistic neuroprotective effect after NMDA injury when combined with an anti-inflammatory treatment based on hesperidin.
Collapse
Affiliation(s)
- Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Taimu Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Michiko Ohno-Oishi
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Mikako Ozawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shigeto Maekawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Takeshi Yabana
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Noriko Himori
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kosuke Fujita
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji M Nishiguchi
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shi Ge
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Miyagi, Japan.
| |
Collapse
|
7
|
Modeling Retinal Ganglion Cell Dysfunction in Optic Neuropathies. Cells 2021; 10:cells10061398. [PMID: 34198840 PMCID: PMC8227951 DOI: 10.3390/cells10061398] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
As in glaucoma and other optic neuropathies cellular dysfunction often precedes cell death, the assessment of retinal ganglion cell (RGC) function represents a key outcome measure for neuroprotective strategies aimed at targeting distressed but still viable cells. RGC dysfunction can be assessed with the pattern electroretinogram (PERG), a sensitive measure of electrical activity of RGCs that is recorded non-invasively in human subjects and mouse models. Here, we offer a conceptual framework based on an intuitive state-transition model used for disease management in patients to identify progressive, potentially reversible stages of RGC dysfunction leading to cell death in mouse models of glaucoma and other optic neuropathies. We provide mathematical equations to describe state-transitions with a set of modifiable parameters that alter the time course and severity of state-transitions, which can be used for hypothesis testing and fitting experimental PERG data. PERG dynamics as a function of physiological stimuli are also used to differentiate phenotypic and altered RGC response dynamics, to assess susceptibility to stressors and to assess reversible dysfunction upon pharmacological treatment.
Collapse
|
8
|
Marola OJ, Syc-Mazurek SB, Libby RT. DDIT3 (CHOP) contributes to retinal ganglion cell somal loss but not axonal degeneration in DBA/2J mice. Cell Death Discov 2019; 5:140. [PMID: 31632741 PMCID: PMC6787076 DOI: 10.1038/s41420-019-0220-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 09/04/2019] [Indexed: 01/08/2023] Open
Abstract
Glaucoma is an age-related neurodegenerative disease characterized by the progressive loss of retinal ganglion cells (RGCs). Chronic ocular hypertension, an important risk factor for glaucoma, leads to RGC axonal injury at the optic nerve head. This insult triggers molecularly distinct cascades governing RGC somal apoptosis and axonal degeneration. The molecular mechanisms activated by ocular hypertensive insult that drive both RGC somal apoptosis and axonal degeneration are incompletely understood. The cellular response to endoplasmic reticulum stress and induction of pro-apoptotic DNA damage inducible transcript 3 (DDIT3, also known as CHOP) have been implicated as drivers of neurodegeneration in many disease models, including glaucoma. RGCs express DDIT3 after glaucoma-relevant insults, and importantly, DDIT3 has been shown to contribute to both RGC somal apoptosis and axonal degeneration after acute induction of ocular hypertension. However, the role of DDIT3 in RGC somal and axonal degeneration has not been critically tested in a model of age-related chronic ocular hypertension. Here, we investigated the role of DDIT3 in glaucomatous RGC death using an age-related, naturally occurring ocular hypertensive mouse model of glaucoma, DBA/2J mice (D2). To accomplish this, a null allele of Ddit3 was backcrossed onto the D2 background. Homozygous Ddit3 deletion did not alter gross retinal or optic nerve head morphology, nor did it change the ocular hypertensive profile of D2 mice. In D2 mice, Ddit3 deletion conferred mild protection to RGC somas, but did not significantly prevent RGC axonal degeneration. Together, these data suggest that DDIT3 plays a minor role in perpetuating RGC somal apoptosis caused by chronic ocular hypertension-induced axonal injury, but does not significantly contribute to distal axonal degeneration.
Collapse
Affiliation(s)
- Olivia J Marola
- 1Department of Ophthalmology, Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY USA.,2Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY USA.,3The Center for Visual Sciences, University of Rochester, Rochester, NY USA
| | - Stephanie B Syc-Mazurek
- 1Department of Ophthalmology, Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY USA.,4Medical Scientist Training Program, University of Rochester Medical Center, Rochester, NY USA
| | - Richard T Libby
- 1Department of Ophthalmology, Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY USA.,3The Center for Visual Sciences, University of Rochester, Rochester, NY USA.,5Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY USA
| |
Collapse
|
9
|
Tawarayama H, Feng Q, Murayama N, Suzuki N, Nakazawa T. Cyclin-Dependent Kinase Inhibitor 2b Mediates Excitotoxicity-Induced Death of Retinal Ganglion Cells. ACTA ACUST UNITED AC 2019; 60:4479-4488. [DOI: 10.1167/iovs.19-27396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Hiroshi Tawarayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Qiwei Feng
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Namie Murayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriyuki Suzuki
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
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: 1.2] [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
|
Crosbie DE, Keaney J, Tam LCS, Daniel Stamer W, Campbell M, Humphries P. Age-related changes in eye morphology and aqueous humor dynamics in DBA/2J mice using contrast-enhanced ocular MRI. Magn Reson Imaging 2019; 59:10-16. [PMID: 30660703 DOI: 10.1016/j.mri.2019.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE Here, we are testing the hypothesis that dynamic contrast enhanced MRI (DCE-MRI) is a useful approach for non-invasively evaluating age-related changes in aqueous humor outflow and its contribution to elevated intraocular pressure in the DBA/2J model of pigmentary glaucoma. METHODS A rodent-specific 7 T MRI was used to assess eye anatomy (anterior chamber (AC) and vitreous body (VB) morphology, eye size, lens size) and aqueous humor dynamics (via intravenous administration of Gd-DTPA and Gd-BOPTA contrast agents) in C57BL/6 and DBA/2J mice at 3 and 9 months of age. RESULTS Gd-MRI was used to demonstrate an anterior solute pathway into the mouse AC. Topical latanoprost treatment in C57BL/6J mice reduced Gd-BOPTA accumulation in the AC. Age-related increases in AC area, AC depth and eye size were observed in DBA/2J mice compared to C57BL/6J mice. The rate of Gd-DTPA accumulation and peak Gd-DTPA intensity was lowest in 9-month old DBA/2J mice compared to 3-month old DBA/2J mice and C57BL/6J mice at both ages. Leakage of Gd-DTPA posteriorly into the VB was also observed in 9-month old DBA/2J mice. CONCLUSIONS These studies support the idea that age-related changes in aqueous humor outflow contribute to elevated intraocular pressure (IOP) in the DBA/2J model of pigmentary glaucoma. Gd-MRI is a valuable tool for better understanding of mechanisms and dynamics of aqueous humor circulation in normal and glaucomatous mouse eyes or following topical administration of medicines to reduce IOP.
Collapse
Affiliation(s)
- Darragh E Crosbie
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - James Keaney
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland; Ross University School of Veterinary Medicine, P. O. Box 334, Basseterre, St. Kitts, Saint Kitts and Nevis
| | - Lawrence C S Tam
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - W Daniel Stamer
- Departments of Ophthalmology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Matthew Campbell
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland.
| | - Peter Humphries
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland.
| |
Collapse
|
12
|
Adeghate J, Rahmatnejad K, Waisbourd M, Katz LJ. Intraocular pressure-independent management of normal tension glaucoma. Surv Ophthalmol 2018; 64:101-110. [PMID: 30300625 DOI: 10.1016/j.survophthal.2018.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 08/15/2018] [Accepted: 08/20/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Jennifer Adeghate
- Wills Eye Hospital, Glaucoma Research Department, Philadelphia, Pennsylvania, USA; Weill Cornell Medical College, Department of Ophthalmology, New York, New York, USA
| | - Kamran Rahmatnejad
- Wills Eye Hospital, Glaucoma Research Department, Philadelphia, Pennsylvania, USA
| | - Michael Waisbourd
- Wills Eye Hospital, Glaucoma Research Department, Philadelphia, Pennsylvania, USA; Thomas Jefferson University, Department of Ophthalmology, Philadelphia, Pennsylvania, USA; Tel-Aviv University Medical Center, Glaucoma Research Center, Tel-Aviv, Israel
| | - L Jay Katz
- Wills Eye Hospital, Glaucoma Research Department, Philadelphia, Pennsylvania, USA; Thomas Jefferson University, Department of Ophthalmology, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
13
|
Age-related Changes in Eye, Brain and Visuomotor Behavior in the DBA/2J Mouse Model of Chronic Glaucoma. Sci Rep 2018; 8:4643. [PMID: 29545576 PMCID: PMC5854610 DOI: 10.1038/s41598-018-22850-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/02/2018] [Indexed: 12/19/2022] Open
Abstract
Although elevated intraocular pressure (IOP) and age are major risk factors for glaucoma, their effects on glaucoma pathogenesis remain unclear. This study examined the onset and progression of glaucomatous changes to ocular anatomy and physiology, structural and physiological brain integrity, and visuomotor behavior in the DBA/2J mice via non-invasive tonometry, multi-parametric magnetic resonance imaging (MRI) and optokinetic assessments from 5 to 12 months of age. Using T2-weighted MRI, diffusion tensor MRI, and manganese-enhanced MRI, increasing IOP elevation at 9 and 12 months old coincided with anterior chamber deepening, altered fractional anisotropy and radial diffusivity of the optic nerve and optic tract, as well as reduced anterograde manganese transport along the visual pathway respectively in the DBA/2J mice. Vitreous body elongation and visuomotor function deterioration were observed until 9 months old, whereas axial diffusivity only decreased at 12 months old in diffusion tensor MRI. Under the same experimental settings, C57BL/6J mice only showed modest age-related changes. Taken together, these results indicate that the anterior and posterior visual pathways of the DBA/2J mice exhibit differential susceptibility to glaucomatous neurodegeneration observable by in vivo multi-modal examinations.
Collapse
|
14
|
Rizzo MI, Greco A, De Virgilio A, Gallo A, Taverniti L, Fusconi M, Conte M, Pagliuca G, Turchetta R, de Vincentiis M. Glaucoma: recent advances in the involvement of autoimmunity. Immunol Res 2018; 65:207-217. [PMID: 27475096 DOI: 10.1007/s12026-016-8837-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Glaucomatous optic neuropathy is the most commonly acquired optic neuropathy encountered in clinical practice. It is the second leading cause of blindness globally, after cataracts, but it presents a greater public health challenge than cataracts, because the blindness it causes is irreversible. It has pathogenesis still largely unknown and no established cure. Alterations in serum antibody profiles, upregulation, and downregulation have been described, but it still remains elusive if the autoantibodies seen in glaucoma are an epiphenomenon or causative. Hypertension, diabetes, and hearing disorders also are associated. This review is a glaucoma update with focus about the recent advances in the last 15 years.
Collapse
Affiliation(s)
- Maria Ida Rizzo
- ENT Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy.,Department of Surgical Science, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| | - Antonio Greco
- Department of Surgical Science, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| | - Armando De Virgilio
- ENT Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy. .,Department of Surgical Science, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy.
| | - Andrea Gallo
- Otorhinolaryngology Section, Department of Medico-Surgical Sciences and Biotechnologies, ''Sapienza'' University of Rome, Corso della Repubblica, 79, 04100, Latina, LT, Italy
| | - Luciano Taverniti
- Ophthalmology Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| | - Massimo Fusconi
- ENT Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| | - Michela Conte
- ENT Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| | - Giulio Pagliuca
- Otorhinolaryngology Section, Department of Medico-Surgical Sciences and Biotechnologies, ''Sapienza'' University of Rome, Corso della Repubblica, 79, 04100, Latina, LT, Italy
| | - Rosaria Turchetta
- ENT Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| | - Marco de Vincentiis
- ENT Section, Department Organs of Sense, University of Rome "La Sapienza", Viale del Policlinico 155, 00100, Rome, Italy
| |
Collapse
|
15
|
Chou TH, Musada GR, Romano GL, Bolton E, Porciatti V. Anesthetic Preconditioning as Endogenous Neuroprotection in Glaucoma. Int J Mol Sci 2018; 19:E237. [PMID: 29342845 PMCID: PMC5796185 DOI: 10.3390/ijms19010237] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/15/2022] Open
Abstract
Blindness in glaucoma is the result of death of Retinal Ganglion Cells (RGCs) and their axons. RGC death is generally preceded by a stage of reversible dysfunction and structural remodeling. Current treatments aimed at reducing intraocular pressure (IOP) are ineffective or incompletely effective in management of the disease. IOP-independent neuroprotection or neuroprotection as adjuvant to IOP lowering in glaucoma remains a challenge as effective agents without side effects have not been identified yet. We show in DBA/2J mice with spontaneous IOP elevation and glaucoma that the lifespan of functional RGCs can be extended by preconditioning RGCs with retrobulbar lidocaine in one eye at four months of age that temporary blocks RGC axonal transport. The contralateral, PBS-injected eye served as control. Lidocaine-induced impairment of axonal transport to superior colliculi was assessed by intravitreal injection of cholera toxin B. Long-term (nine months) effect of lidocaine were assessed on RGC electrical responsiveness (PERG), IOP, expression of relevant protein (BDNF, TrkB, PSD95, GFAP, Synaptophysin, and GAPDH) and RGC density. While lidocaine treatment did not alter the age-related increase of IOP, TrkB expression was elevated, GFAP expression was decreased, RGC survival was improved by 35%, and PERG function was preserved. Results suggest that the lifespan of functional RGCs in mouse glaucoma can be extended by preconditioning RGCs in early stages of the disease using a minimally invasive treatment with retrobulbar lidocaine, a common ophthalmologic procedure. Lidocaine is inexpensive, safe and is approved by Food and Drug Administration (FDA) to be administered intravenously.
Collapse
Affiliation(s)
- Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL 33136, USA.
| | - Ganeswara Rao Musada
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL 33136, USA.
| | - Giovanni Luca Romano
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL 33136, USA.
| | - Elizabeth Bolton
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL 33136, USA.
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
16
|
Sánchez-López E, Egea MA, Davis BM, Guo L, Espina M, Silva AM, Calpena AC, Souto EMB, Ravindran N, Ettcheto M, Camins A, García ML, Cordeiro MF. Memantine-Loaded PEGylated Biodegradable Nanoparticles for the Treatment of Glaucoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:1701808. [PMID: 29154484 DOI: 10.1002/smll.201701808] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/10/2017] [Indexed: 05/20/2023]
Abstract
Glaucoma is a multifactorial neurodegenerative disease associated with retinal ganglion cells (RGC) loss. Increasing reports of similarities in glaucoma and other neurodegenerative conditions have led to speculation that therapies for brain neurodegenerative disorders may also have potential as glaucoma therapies. Memantine is an N-methyl-d-aspartate (NMDA) antagonist approved for Alzheimer's disease treatment. Glutamate-induced excitotoxicity is implicated in glaucoma and NMDA receptor antagonism is advocated as a potential strategy for RGC preservation. This study describes the development of a topical formulation of memantine-loaded PLGA-PEG nanoparticles (MEM-NP) and investigates the efficacy of this formulation using a well-established glaucoma model. MEM-NPs <200 nm in diameter and incorporating 4 mg mL-1 of memantine were prepared with 0.35 mg mL-1 localized to the aqueous interior. In vitro assessment indicated sustained release from MEM-NPs and ex vivo ocular permeation studies demonstrated enhanced delivery. MEM-NPs were additionally found to be well tolerated in vitro (human retinoblastoma cells) and in vivo (Draize test). Finally, when applied topically in a rodent model of ocular hypertension for three weeks, MEM-NP eye drops were found to significantly (p < 0.0001) reduce RGC loss. These results suggest that topical MEM-NP is safe, well tolerated, and, most promisingly, neuroprotective in an experimental glaucoma model.
Collapse
Affiliation(s)
- Elena Sánchez-López
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, 08028, Spain
- Biomedical Research and Networking Center in Neurodegenerative diseases (CIBERNED), Madrid, 28031, Spain
| | - Maria Antonia Egea
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, 08028, Spain
| | - Benjamin Michael Davis
- Glaucoma and Retinal Neurodegeneration Research, Visual Neuroscience, UCL Institute of Ophthalmology, Bath Street, London, EC1V 9EL, UK
| | - Li Guo
- Glaucoma and Retinal Neurodegeneration Research, Visual Neuroscience, UCL Institute of Ophthalmology, Bath Street, London, EC1V 9EL, UK
| | - Marta Espina
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, 08028, Spain
| | - Amelia Maria Silva
- Department of Biology and Environment, School of Life and Environmental sciences (ECVA, UTAD), and Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB-UTAD), University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5001-801, Vila Real, Portugal
| | - Ana Cristina Calpena
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, 08028, Spain
| | - Eliana Maria Barbosa Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC) and REQUIMTE/Group of Pharmaceutical Technology, Polo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Nivedita Ravindran
- Glaucoma and Retinal Neurodegeneration Research, Visual Neuroscience, UCL Institute of Ophthalmology, Bath Street, London, EC1V 9EL, UK
| | - Miren Ettcheto
- Biomedical Research and Networking Center in Neurodegenerative diseases (CIBERNED), Madrid, 28031, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, 08028, Spain
| | - Antonio Camins
- Biomedical Research and Networking Center in Neurodegenerative diseases (CIBERNED), Madrid, 28031, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, 08028, Spain
| | - Maria Luisa García
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, 08028, Spain
| | - Maria Francesca Cordeiro
- Glaucoma and Retinal Neurodegeneration Research, Visual Neuroscience, UCL Institute of Ophthalmology, Bath Street, London, EC1V 9EL, UK
- Western Eye Hospital, Imperial College Healthcare Trust, London, UK
| |
Collapse
|
17
|
Kalogerou M, Kolovos P, Prokopiou E, Papagregoriou G, Deltas C, Malas S, Georgiou T. Omega-3 fatty acids protect retinal neurons in the DBA/2J hereditary glaucoma mouse model. Exp Eye Res 2017; 167:128-139. [PMID: 29258748 DOI: 10.1016/j.exer.2017.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 11/15/2017] [Accepted: 12/11/2017] [Indexed: 12/31/2022]
Abstract
The purpose of this study was to evaluate the neuroprotective effects of omega-3 polyunsaturated fatty acid (ω3-PUFA) supplementation, alone or in combination with timolol eye drops, in a mouse model of hereditary glaucoma. DBA/2J mice (8.5-month-old) were assigned to an ω3-PUFAs + timolol, ω3-PUFAs only, timolol only, or an untreated group. Treated mice received a daily gavage administration of eicosapentaenoic acid (EPA) and docosahexaenoic acid and/or topical instillation of timolol (0.5%) once a day for 3 months. Blood was analysed regularly to determine ω3-PUFA levels and retinas were histologically analysed. Real-time PCR and Western blot were performed for retinal pro-inflammatory cytokines and macrophages. Blood arachidonic acid/EPA ratio gradually decreased and reached the desired therapeutic range (1-1.5) after 4 weeks of daily gavage with ω3-PUFAs in the ω3-PUFAs + timolol and ω3-PUFAs only groups. Retinal ganglion cell densities were significantly higher in the ω3-PUFAs + timolol (1303.77 ± 139.62/mm2), ω3-PUFAs only (768.40 ± 52.44/mm2) and timolol only (910.57 ± 57.28/mm2) groups than in the untreated group (323.39 ± 95.18/mm2). ω3-PUFA supplementation alone or timolol alone, significantly increased protein expression levels of M1 macrophage-secreted inducible nitric oxide synthase and M2 macrophage-secreted arginase-1 in the retina, which led to significant decreases in the expression levels of tumour necrosis factor-α (TNF-α). ω3-PUFA supplementation alone also resulted in significantly reduced expression of interleukin-18 (IL-18). ω3-PUFA + timolol treatment had no effect on the expression level of any of the aforementioned mediators in the retina. Supplementation with ω3-PUFAs has neuroprotective effect in the retinas of DBA/2J mice that is enhanced when combined with timolol eye drops. The continued inflammation following ω3-PUFAs + timolol treatment suggests that downregulation of IL-18 and TNF-α may not be the only factors involved in ω3-PUFA-mediated neuroprotection in the retina.
Collapse
MESH Headings
- Administration, Ophthalmic
- Adrenergic beta-Antagonists/therapeutic use
- Animals
- Arachidonic Acid/blood
- Arginase/metabolism
- Blotting, Western
- Cell Survival
- Disease Models, Animal
- Drug Combinations
- Eicosapentaenoic Acid/blood
- Fatty Acids, Omega-3/administration & dosage
- Female
- Glaucoma, Open-Angle/genetics
- Glaucoma, Open-Angle/metabolism
- Glaucoma, Open-Angle/prevention & control
- Interleukin-18/metabolism
- Intraocular Pressure/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Nitric Oxide Synthase Type II/metabolism
- Ophthalmic Solutions
- Optic Nerve Diseases/genetics
- Optic Nerve Diseases/metabolism
- Optic Nerve Diseases/prevention & control
- Real-Time Polymerase Chain Reaction
- Retinal Ganglion Cells/drug effects
- Timolol/therapeutic use
- Tonometry, Ocular
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Maria Kalogerou
- Ophthalmos Research and Educational Institute, Morfou 48, Egkomi, 2417 Nicosia, Cyprus.
| | - Panagiotis Kolovos
- Ophthalmos Research and Educational Institute, Morfou 48, Egkomi, 2417 Nicosia, Cyprus.
| | - Ekatherine Prokopiou
- Ophthalmos Research and Educational Institute, Morfou 48, Egkomi, 2417 Nicosia, Cyprus.
| | - Gregory Papagregoriou
- Molecular Medicine Research Centre and Laboratory of Molecular and Medical Genetics, Department of Biological Sciences, University of Cyprus, Kallipoleos 75, 1678 Nicosia, Cyprus.
| | - Constantinos Deltas
- Molecular Medicine Research Centre and Laboratory of Molecular and Medical Genetics, Department of Biological Sciences, University of Cyprus, Kallipoleos 75, 1678 Nicosia, Cyprus.
| | - Stavros Malas
- Developmental and Functional Genetics Group, The Cyprus Institute of Neurology and Genetics, 1683 Nicosia, Cyprus.
| | - Tassos Georgiou
- Ophthalmos Research and Educational Institute, Morfou 48, Egkomi, 2417 Nicosia, Cyprus.
| |
Collapse
|
18
|
Williams PA, Harder JM, John SWM. Glaucoma as a Metabolic Optic Neuropathy: Making the Case for Nicotinamide Treatment in Glaucoma. J Glaucoma 2017; 26:1161-1168. [PMID: 28858158 PMCID: PMC5854489 DOI: 10.1097/ijg.0000000000000767] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondrial dysfunction may be an important, if not essential, component of human glaucoma. Using transcriptomics followed by molecular and neurobiological techniques, we have recently demonstrated that mitochondrial dysfunction within retinal ganglion cells is an early feature in the DBA/2J mouse model of inherited glaucoma. Guided by these findings, we discovered that the retinal level of nicotinamide adenine dinucleotide (NAD, a key molecule for mitochondrial health) declines in an age-dependent manner. We hypothesized that this decline in NAD renders retinal ganglion cells susceptible to damage during periods of elevated intraocular pressure. To replete NAD levels in this glaucoma, we administered nicotinamide (the amide of vitamin B3). At the lowest dose tested, nicotinamide robustly protected from glaucoma (~70% of eyes had no detectable glaucomatous neurodegeneration). At this dose, nicotinamide had no influence on intraocular pressure and so its effect was neuroprotective. At the highest dose tested, 93% of eyes had no detectable glaucoma. This represents a ~10-fold decrease in the risk of developing glaucoma. At this dose, intraocular pressure still became elevated but there was a reduction in the degree of elevation showing an additional benefit. Thus, nicotinamide is unexpectedly potent at preventing this glaucoma and is an attractive option for glaucoma therapeutics. Our findings demonstrate the promise for both preventing and treating glaucoma by interventions that bolster metabolism during increasing age and during periods of elevated intraocular pressure. Nicotinamide prevents age-related declines in NAD (a decline that occurs in different genetic contexts and species). NAD precursors are reported to protect from a variety of neurodegenerative conditions. Thus, nicotinamide may provide a much needed neuroprotective treatment against human glaucoma. This manuscript summarizes human data implicating mitochondria in glaucoma, and argues for studies to further assess the safety and efficacy of nicotinamide in human glaucoma care.
Collapse
Affiliation(s)
- Pete A Williams
- The Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Jeffrey M Harder
- The Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Simon W M John
- The Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, USA
- Department of Ophthalmology, Tufts University of Medicine, Boston, MA, USA
| |
Collapse
|
19
|
Norrin protects optic nerve axons from degeneration in a mouse model of glaucoma. Sci Rep 2017; 7:14274. [PMID: 29079753 PMCID: PMC5660254 DOI: 10.1038/s41598-017-14423-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/10/2017] [Indexed: 11/25/2022] Open
Abstract
Norrin is a secreted signaling molecule activating the Wnt/β-catenin pathway. Since Norrin protects retinal neurons from experimental acute injury, we were interested to learn if Norrin attenuates chronic damage of retinal ganglion cells (RGC) and their axons in a mouse model of glaucoma. Transgenic mice overexpressing Norrin in the retina (Pax6-Norrin) were generated and crossed with DBA/2J mice with hereditary glaucoma and optic nerve axonal degeneration. One-year old DBA/2J/Pax6-Norrin animals had significantly more surviving optic nerve axons than their DBA/2J littermates. The protective effect correlated with an increase in insulin-like growth factor (IGF)-1 mRNA and an enhanced Akt phosphorylation in DBA/2J/Pax6-Norrin mice. Both mouse strains developed an increase in intraocular pressure during the second half of the first year and marked degenerative changes in chamber angle, ciliary body and iris structure. The degenerations were slightly attenuated in the chamber angle of DBA/2J/Pax6-Norrin mice, which showed a β-catenin increase in the trabecular meshwork. We conclude that high levels of Norrin and the subsequent constitutive activation of Wnt/β-catenin signaling in RGC protect from glaucomatous axonal damage via IGF-1 causing increased activity of PI3K-Akt signaling. Our results identify components of a protective signaling network preventing degeneration of optic nerve axons in glaucoma.
Collapse
|
20
|
JUN is important for ocular hypertension-induced retinal ganglion cell degeneration. Cell Death Dis 2017; 8:e2945. [PMID: 28726785 PMCID: PMC5550879 DOI: 10.1038/cddis.2017.338] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/06/2017] [Accepted: 06/12/2017] [Indexed: 12/14/2022]
Abstract
Ocular hypertension, a major risk factor for glaucoma, is thought to trigger glaucomatous neurodegeneration through injury to retinal ganglion cell (RGC) axons. The molecular signaling pathway leading from ocular hypertension to RGC degeneration, however, is not well defined. JNK signaling, a component of the mitogen-activated protein kinase (MAPK) family, and its canonical target, the transcription factor JUN, have been shown to regulate neurodegeneration in many different systems. JUN is expressed after glaucoma-relevant injuries and Jun deficiency protects RGCs after mechanical injury to the optic nerve. Here, we tested the importance of JNK–JUN signaling for RGC death after ocular hypertensive axonal injury in an age-related, mouse model of ocular hypertension. Immunohistochemistry was performed to evaluate JUN expression in ocular hypertensive DBA/2J mice. JUN was expressed in a temporal and spatial pattern consistent with a role in glaucomatous injury. To determine the importance of JUN in ocular hypertension-induced RGC death, a floxed allele of Jun and a retinal expressed cre recombinase (Six3-cre) were backcrossed onto the DBA/2J background. Intraocular pressure (IOP) and gross morphology of the retina and optic nerve head were assessed to determine whether removing Jun from the developing retina altered IOP elevation or retinal development. Jun deficiency in the retina did not alter DBA/2J IOP elevation or retinal development. Optic nerves and retinas were assessed at ages known to have glaucomatous damage in DBA/2J mice. Jun deficiency protected RGC somas from ocular hypertensive injury, but did not protect RGC axons from glaucomatous neurodegeneration. Jun is a major regulator of RGC somal degeneration after glaucomatous ocular hypertensive injury. These results suggest in glaucomatous neurodegeneration, JNK–JUN signaling has a major role as a pro-death signaling pathway between axonal injury and somal degeneration.
Collapse
|
21
|
Harder JM, Braine CE, Williams PA, Zhu X, MacNicoll KH, Sousa GL, Buchanan RA, Smith RS, Libby RT, Howell GR, John SWM. Early immune responses are independent of RGC dysfunction in glaucoma with complement component C3 being protective. Proc Natl Acad Sci U S A 2017; 114:E3839-E3848. [PMID: 28446616 PMCID: PMC5441748 DOI: 10.1073/pnas.1608769114] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Various immune response pathways are altered during early, predegenerative stages of glaucoma; however, whether the early immune responses occur secondarily to or independently of neuronal dysfunction is unclear. To investigate this relationship, we used the Wlds allele, which protects from axon dysfunction. We demonstrate that DBA/2J.Wlds mice develop high intraocular pressure (IOP) but are protected from retinal ganglion cell (RGC) dysfunction and neuroglial changes that otherwise occur early in DBA/2J glaucoma. Despite this, immune pathways are still altered in DBA/2J.Wlds mice. This suggests that immune changes are not secondary to RGC dysfunction or altered neuroglial interactions, but may be directly induced by the increased strain imposed by high IOP. One early immune response following IOP elevation is up-regulation of complement C3 in astrocytes of DBA/2J and DBA/2J.Wlds mice. Unexpectedly, because the disruption of other complement components, such as C1Q, is protective in glaucoma, C3 deficiency significantly increased the number of DBA/2J eyes with nerve damage and RGC loss at an early time point after IOP elevation. Transcriptional profiling of C3-deficient cultured astrocytes implicated EGFR signaling as a hub in C3-dependent responses. Treatment with AG1478, an EGFR inhibitor, also significantly increased the number of DBA/2J eyes with glaucoma at the same early time point. These findings suggest that C3 protects from early glaucomatous damage, a process that may involve EGFR signaling and other immune responses in the optic nerve head. Therefore, therapies that target specific components of the complement cascade, rather than global inhibition, may be more applicable for treating human glaucoma.
Collapse
Affiliation(s)
| | | | | | - Xianjun Zhu
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME 04609
| | | | | | | | | | - Richard T Libby
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY 14642
| | | | - Simon W M John
- The Jackson Laboratory, Bar Harbor, ME 04609
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME 04609
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
22
|
Maes ME, Schlamp CL, Nickells RW. BAX to basics: How the BCL2 gene family controls the death of retinal ganglion cells. Prog Retin Eye Res 2017; 57:1-25. [PMID: 28064040 DOI: 10.1016/j.preteyeres.2017.01.002] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022]
Abstract
Retinal ganglion cell (RGC) death is the principal consequence of injury to the optic nerve. For several decades, we have understood that the RGC death process was executed by apoptosis, suggesting that there may be ways to therapeutically intervene in this cell death program and provide a more direct treatment to the cells and tissues affected in diseases like glaucoma. A major part of this endeavor has been to elucidate the molecular biological pathways active in RGCs from the point of axonal injury to the point of irreversible cell death. A major component of this process is the complex interaction of members of the BCL2 gene family. Three distinct family members of proteins orchestrate the most critical junction in the apoptotic program of RGCs, culminating in the activation of pro-apoptotic BAX. Once active, BAX causes irreparable damage to mitochondria, while precipitating downstream events that finish off a dying ganglion cell. This review is divided into two major parts. First, we summarize the extent of knowledge of how BCL2 gene family proteins interact to facilitate the activation and function of BAX. This area of investigation has rapidly changed over the last few years and has yielded a dramatically different mechanistic understanding of how the intrinsic apoptotic program is run in mammalian cells. Second, we provided a comprehensive analysis of nearly two decades of investigation of the role of BAX in the process of RGC death, much of which has provided many important insights into the overall pathophysiology of diseases like glaucoma.
Collapse
Affiliation(s)
- Margaret E Maes
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Cassandra L Schlamp
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Borrás T. The Pathway From Genes to Gene Therapy in Glaucoma: A Review of Possibilities for Using Genes as Glaucoma Drugs. Asia Pac J Ophthalmol (Phila) 2017; 6:80-93. [PMID: 28161916 PMCID: PMC6005701 DOI: 10.22608/apo.2016126] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/27/2016] [Indexed: 12/19/2022] Open
Abstract
Treatment of diseases with gene therapy is advancing rapidly. The use of gene therapy has expanded from the original concept of re-placing the mutated gene causing the disease to the use of genes to con-trol nonphysiological levels of expression or to modify pathways known to affect the disease. Genes offer numerous advantages over conventional drugs. They have longer duration of action and are more specific. Genes can be delivered to the target site by naked DNA, cells, nonviral, and viral vectors. The enormous progress of the past decade in molecular bi-ology and delivery systems has provided ways for targeting genes to the intended cell/tissue and safe, long-term vectors. The eye is an ideal organ for gene therapy. It is easily accessible and it is an immune-privileged site. Currently, there are clinical trials for diseases affecting practically every tissue of the eye, including those to restore vision in patients with Leber congenital amaurosis. However, the number of eye trials compared with those for systemic diseases is quite low (1.8%). Nevertheless, judg-ing by the vast amount of ongoing preclinical studies, it is expected that such number will increase considerably in the near future. One area of great need for eye gene therapy is glaucoma, where a long-term gene drug would eliminate daily applications and compliance issues. Here, we review the current state of gene therapy for glaucoma and the possibilities for treating the trabecular meshwork to lower intraocular pressure and the retinal ganglion cells to protect them from neurodegeneration.
Collapse
Affiliation(s)
- Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Emerging Concepts in Glaucoma and Review of the Literature. Am J Med 2016; 129:1000.e7-1000.e13. [PMID: 27125182 DOI: 10.1016/j.amjmed.2016.03.038] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 01/04/2023]
Abstract
Glaucoma is the most commonly acquired optic neuropathy. It represents a public health challenge because it causes an irreversible blindness. Emerging evidence indicates that the pathogenesis of glaucoma depends on several interacting pathogenetic mechanisms, which include mechanical effects by an increased intraocular pressure, decreased neutrophine-supply, hypoxia, excitotoxicity, oxidative stress, and the involvement of autoimmune processes. In particular, alterations in serum antibody profiles have been described. However, it is still unclear whether the autoantibodies seen in glaucoma are an epiphenomenon or causative. Oxidative stress appears to be a critical factor in the neurodestructive consequences of mitochondrial dysfunction, glial activation response, and uncontrolled activity of the immune system during glaucomatous neurodegeneration. In addition, hearing loss has been identified in association with glaucoma. A higher prevalence of antiphosphatidylserine antibodies of the immunoglobulin G class was seen in normal-tension glaucoma patients with hearing loss in comparison with normal-tension glaucoma patients with normacusis. This finding suggests a similar pathological pathway as a sign for generalized disease.
Collapse
|
25
|
Chandra S, Muir ER, Deo K, Kiel JW, Duong TQ. Effects of Dorzolamide on Retinal and Choroidal Blood Flow in the DBA/2J Mouse Model of Glaucoma. Invest Ophthalmol Vis Sci 2016; 57:826-31. [PMID: 26934140 PMCID: PMC4777278 DOI: 10.1167/iovs.15-18291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To test the hypothesis that acute topical dorzolamide (DZ) decreases intraocular pressure (IOP) and increases retinal and choroidal blood flow in the DBA/2J mouse model of glaucoma. METHODS Retinal and choroidal blood flow were measured in 4- and 9-month-old DBA/2J mice, and 4-month C57BL/6 (control) mice under isoflurane anesthesia using magnetic resonance imaging. Ocular blood flow was measured at baseline, and 1 and 2 hours after topical dorzolamide. Intraocular pressure was measured using a rebound tonometer in a subset of animals at the same time points. RESULTS Baseline IOP in the 4-month-old DBA/2J mice and C57BL/6 mice was not significantly different (P > 0.05), and IOP in both groups was less than in the 9-month-old DBA/2J mice (P < 0.05 for both). Compared to baseline, dorzolamide reduced IOP at 1 and 2 hours after dorzolamide in the 4- (P < 0.05) and 9-month-old (P < 0.01) DBA/2J mice, but not in the C57BL/6J mice (P > 0.05). Baseline retinal blood flow was lower in the 4-month and 9-month-old DBA/2J mice compared with the 4-month-old C57BL/6J mice (P < 0.05). Baseline choroidal blood flow in the 9-month-old DBA/2J mice was less than in the C57BL/6J mice (P < 0.05). Compared with baseline, both retinal and choroidal blood flow increased at 1-hour post-dorzolamide and remained elevated 2 hours later in the 9-month-old DBA/2J mice (P < 0.05). CONCLUSIONS Dorzolamide lowers IOP and raises retinal and choroidal blood flow in older DBA/2J mice, consistent with the study hypothesis.
Collapse
|
26
|
Celiker H, Yuksel N, Solakoglu S, Karabas L, Aktar F, Caglar Y. Neuroprotective Effects of Memantine in the Retina of Glaucomatous Rats: An Electron Microscopic Study. J Ophthalmic Vis Res 2016; 11:174-82. [PMID: 27413498 PMCID: PMC4926565 DOI: 10.4103/2008-322x.183934] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose: In this experimental study, the effects of systemic memantine administration on the retinal ultrastructure of experimentally induced glaucomatous rats were investigated. Methods: Twenty-four Wistar albino rats were included in this study. Glaucoma was induced by injecting sodium hyaluronate into the anterior chamber of the rats for a period of three weeks. As a control, 8 rats were sham treated (Group C). Glaucoma induced animals were divided into two groups; Group M (n = 8) received a single daily dose of 10 mg/kg memantine, and Group G received the same volume of saline (n = 8), via intraperitoneal route for a period of six weeks, starting with the induction of glaucoma. Then, all rats were sacrificed and the retinas were prepared for electron microscopic examination. Electron microscopic damage findings were graded between 0 and 4 and mean damage scores for each cell or layer was calculated for each group. Statistical comparison was made between group G and group M. Results: Including the photoreceptor cells, marked ultrastructural changes were observed in the retinas of the animals in group G. The ultrastructural changes in group M were modest and there was no significant cell death. Statistical findings indicated these results. Conclusion: Results of the present study suggest that memantine treatment, when started in the early phase of glaucomatous process, may help to preserve the retinal ultrastructure and thus prevent neuronal injury in experimentally induced glaucoma.
Collapse
Affiliation(s)
- Hande Celiker
- Department of Ophthalmology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Nursen Yuksel
- Department of Ophthalmology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Seyhun Solakoglu
- Department of Histology and Embryology, School of Medicine, Istanbul University, Istanbul, Turkey
| | - Levent Karabas
- Department of Ophthalmology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Fadime Aktar
- Department of Histology and Embryology, School of Medicine, Istanbul University, Istanbul, Turkey
| | - Yusuf Caglar
- Department of Ophthalmology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
27
|
Sawada K, Hiraoka M, Ohguro H. Effect of Antiglaucoma Medicine on Intraocular Pressure in DBA/2J Mice. Ophthalmic Res 2016; 55:205-11. [PMID: 26942413 DOI: 10.1159/000444057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/16/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The DBA/2J mouse strain is known to develop glaucomatous changes. Intraocular pressure (IOP) fluctuations affected by age or antiglaucoma drug administration were compared among three mouse strains, DBA/2J, C57BL/6, and BALB/c. METHODS IOP was measured using the TonoLab tonometer under systemic anesthesia. For each mouse strain, the effects of age and topical administration of antiglaucoma medications (timolol maleate, dorzolamide, brimonidine tartrate, and travoprost) were assessed, and results were compared among the three strains. RESULTS IOP started to rise in DBA/2J mice at 21 weeks of age. The highest values of IOP were distributed from 18 to 51 mm Hg. Eighty percent of DBA/2J mice showed maximum IOP at either 35 or 46 weeks. IOP of C57BL/6 ranged from 9 to 14 mm Hg as the mice aged. Treatment with any of the antiglaucoma medications resulted in IOP-lowering effects in all three strains. The difference in levels before and after administration ranged from 6 to 10 mm Hg on average in DBA/2J. CONCLUSION DBA/2J mice are a useful animal model to study the effects of antiglaucoma therapy.
Collapse
Affiliation(s)
- Kanako Sawada
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | | | | |
Collapse
|
28
|
Abstract
Glaucoma is a progressive optic neuropathy that causes characteristic changes of the optic nerve and visual field in relation to intraocular pressure (IOP). It is now known that glaucoma can occur at statistically normal IOPs and prevalence studies have shown that normal tension glaucoma (NTG) is more common than previously thought. While IOP is believed to be the predominant risk factor in primary open angle glaucoma (POAG), IOP-independent risk factors, such as vascular dysregulation, are believed to play an important part in the pathogenesis of NTG. Though certain distinguishing phenotypic features of NTG have been reported, such as an increased frequency of disc hemorrhages, acquired pits of the optic nerve and characteristic patterns of disc cupping and visual field loss, there is much overlap of the clinical findings in NTG with POAG, suggesting that NTG is likely part of a continuum of open angle glaucomas. However, IOP modification is still the mainstay of treatment in NTG. As in traditional POAG, reduction of IOP can be achieved with the use of medications, laser trabeculoplasty or surgery. Studies now show that the choice of medication may also be important in determining the outcomes of these patients. Though it is likely that future treatment of NTG will involve modification of both IOP and IOP-independent risk factors, current efforts to develop IOP-independent neuroprotective treatments have not yet proven to be effective in humans.
Collapse
Affiliation(s)
| | - Joseph Caprioli
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, USA
| |
Collapse
|
29
|
Using genetic mouse models to gain insight into glaucoma: Past results and future possibilities. Exp Eye Res 2015; 141:42-56. [PMID: 26116903 DOI: 10.1016/j.exer.2015.06.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/16/2015] [Accepted: 06/23/2015] [Indexed: 12/18/2022]
Abstract
While all forms of glaucoma are characterized by a specific pattern of retinal ganglion cell death, they are clinically divided into several distinct subclasses, including normal tension glaucoma, primary open angle glaucoma, congenital glaucoma, and secondary glaucoma. For each type of glaucoma there are likely numerous molecular pathways that control susceptibility to the disease. Given this complexity, a single animal model will never precisely model all aspects of all the different types of human glaucoma. Therefore, multiple animal models have been utilized to study glaucoma but more are needed. Because of the powerful genetic tools available to use in the laboratory mouse, it has proven to be a highly useful mammalian system for studying the pathophysiology of human disease. The similarity between human and mouse eyes coupled with the ability to use a combination of advanced cell biological and genetic tools in mice have led to a large increase in the number of studies using mice to model specific glaucoma phenotypes. Over the last decade, numerous new mouse models and genetic tools have emerged, providing important insight into the cell biology and genetics of glaucoma. In this review, we describe available mouse genetic models that can be used to study glaucoma-relevant disease/pathobiology. Furthermore, we discuss how these models have been used to gain insights into ocular hypertension (a major risk factor for glaucoma) and glaucomatous retinal ganglion cell death. Finally, the potential for developing new mouse models and using advanced genetic tools and resources for studying glaucoma are discussed.
Collapse
|
30
|
Porciatti V. Electrophysiological assessment of retinal ganglion cell function. Exp Eye Res 2015; 141:164-70. [PMID: 25998495 DOI: 10.1016/j.exer.2015.05.008] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/17/2015] [Accepted: 05/17/2015] [Indexed: 01/22/2023]
Abstract
The function of retinal ganglion cells (RGCs) can be non-invasively assessed in experimental and genetic models of glaucoma by means of variants of the ERG technique that emphasize the activity of inner retina neurons. The best understood technique is the Pattern Electroretinogram (PERG) in response to contrast-reversing gratings or checkerboards, which selectively depends on the presence of functional RGCs. In glaucoma models, the PERG can be altered before histological loss of RGCs; PERG alterations may be either reversed with moderate IOP lowering or exacerbated with moderate IOP elevation. Under particular luminance-stimulus conditions, the Flash-ERG displays components that may reflect electrical activity originating in the proximal retina and be altered in some experimental glaucoma models (positive Scotopic Threshold response, pSTR; negative Scotopic Threshold Response, nSTR; Photopic Negative Response, PhNR; Oscillatory Potentials, OPs; multifocal ERG, mfERG). It is not yet known which of these components is most sensitive to glaucomatous damage. Electrophysiological assessment of RGC function appears to be a necessary outcome measure in experimental glaucoma models, which complements structural assessment and may even predict it. Neuroprotective strategies could be tested based on enhancement of baseline electrophysiological function that results in improved RGC survival. The use of electrophysiology in glaucoma models may be facilitated by specifically designed instruments that allow high throughput, robust assessment of electrophysiological function.
Collapse
Affiliation(s)
- Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, McKnight Vision Research Center, 1638 NW 10th Ave., Miami, FL 33136, United States.
| |
Collapse
|
31
|
Kimura A, Namekata K, Guo X, Noro T, Harada C, Harada T. Valproic acid prevents NMDA-induced retinal ganglion cell death via stimulation of neuronal TrkB receptor signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:756-64. [PMID: 25542970 DOI: 10.1016/j.ajpath.2014.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/04/2014] [Accepted: 11/10/2014] [Indexed: 12/27/2022]
Abstract
Valproic acid (VPA) is widely prescribed for treatment of epilepsy, mood disorders, migraines, and neuropathic pain. It exerts its therapeutic benefits through multiple mechanisms, including enhancement of GABAergic activity, activation of prosurvival protein kinases, and inhibition of histone deacetylase. Increasing evidence suggests that VPA possesses neuroprotective properties. We examined neuroprotective effects of VPA in an N-methyl-d-aspartate (NMDA) excitotoxicity model, which mimics some of the pathological features of glaucoma. In vivo retinal imaging using optical coherence tomography revealed that NMDA-induced retinal degeneration was suppressed in the VPA-treated retina, and histological analyses confirmed that VPA reduced retinal ganglion cell death. In vivo electrophysiological analyses demonstrated that visual impairment was prevented in the VPA-treated retina, clearly establishing both histological and functional effects of VPA. Brain-derived neurotrophic factor (BDNF) expression was up-regulated in Müller glial cells, and neuroprotective effects of VPA on retinal ganglion cells were significantly reduced in a conditional knockout mouse strain with deletion of tropomyosin receptor kinase B (TrkB), a receptor for BDNF from retinal ganglion cells. The results show that VPA stimulates BDNF up-regulation in Müller glial cells and provides direct evidence that neuronal TrkB is important in VPA-mediated neuroprotection. Also, VPA suppresses oxidative stress induced by NMDA in the retina. Our findings raise intriguing possibilities that the widely prescribed drug VPA may be useful for treatment of glaucoma.
Collapse
Affiliation(s)
- Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takahiko Noro
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
32
|
Howell GR, MacNicoll KH, Braine CE, Soto I, Macalinao DG, Sousa GL, John SWM. Combinatorial targeting of early pathways profoundly inhibits neurodegeneration in a mouse model of glaucoma. Neurobiol Dis 2014; 71:44-52. [PMID: 25132557 DOI: 10.1016/j.nbd.2014.07.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/27/2014] [Accepted: 07/16/2014] [Indexed: 12/18/2022] Open
Abstract
The endothelin system is implicated in various human and animal glaucomas. Targeting the endothelin system has great promise as a treatment for human glaucoma, but the cell types involved and the exact mechanisms of action are not clearly elucidated. Here, we report a detailed characterization of the endothelin system in specific cell types of the optic nerve head (ONH) during glaucoma in DBA/2J mice. First, we show that key components of the endothelin system are expressed in multiple cell types. We discover that endothelin 2 (EDN2) is expressed in astrocytes as well as microglia/monocytes in the ONH. The endothelin receptor type A (Ednra) is expressed in vascular endothelial cells, while the endothelin receptor type B (Ednrb) receptor is expressed in ONH astrocytes. Second, we show that Macitentan treatment protects from glaucoma. Macitentan is a novel, orally administered, dual endothelin receptor antagonist with greater affinity, efficacy and safety than previous antagonists. Finally, we test the combinatorial effect of targeting both the endothelin and complement systems as a treatment for glaucoma. Similar to endothelin, the complement system is implicated in a variety of human and animal glaucomas, and has great promise as a treatment target. We discovered that combined targeting of the endothelin (Bosentan) and complement (C1qa mutation) systems is profoundly protective. Remarkably, 80% of DBA/2J eyes subjected to this combined inhibition developed no detectable glaucoma. This opens an exciting new avenue for neuroprotection in glaucoma.
Collapse
Affiliation(s)
- Gareth R Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA.
| | | | | | - Ileana Soto
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | - Gregory L Sousa
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Simon W M John
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA; The Howard Hughes Medical Institute, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA; Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
33
|
Pérez de Lara MJ, Santano C, Guzmán-Aránguez A, Valiente-Soriano FJ, Avilés-Trigueros M, Vidal-Sanz M, de la Villa P, Pintor J. Assessment of inner retina dysfunction and progressive ganglion cell loss in a mouse model of glaucoma. Exp Eye Res 2014; 122:40-9. [PMID: 24631335 DOI: 10.1016/j.exer.2014.02.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 02/13/2014] [Accepted: 02/25/2014] [Indexed: 12/11/2022]
Abstract
The DBA/2J mouse is a model of ocular hypertension and retinal ganglion cell (RGC) degeneration, the main features of which are iris pigment dispersion (IPD) and iris stromal atrophy (ISA). These animals also experience glaucomatous changes, including an increase in intraocular pressure (IOP) beginning at about 9-12 months of age and sectorial RGC death in the retina. The aim of this study was to determine the onset of functional changes exhibited by DBA/2J mice in the inner retina. This was performed by means of electroretinographic recordings (scotopic threshold response, STR) and their correlation with morphological changes (loss of RGCs). To this end, we recorded the scotopic threshold response in control C57BL/6J and in DBA/2J mice at different ages. The RGCs, in both DBA/2J and C57BL/6J animals, were identified at 15 months of age by retrograde tracing with an analogue of fluorogold, hydroxystilbamidine methanesulfonate (OHSt), applied on the superior colliculi. Whole mount retinas were processed to quantify the population of RGCs identified by fluorogold tracing and Brn3a immunodetection, and were counted using image analysis software; an isodensity contour plot was generated for each retina. DBA/2J mice showed a significant reduction in the positive STR (pSTR) amplitudes at 12 months of age, as compared to control C57BL/6J mice of the same age. The pSTR mean amplitude decreased to approximately 27.82% of the values recorded in control mice (p = 0.0058). STR responses decreased in both strains as a result of the natural process of aging, but the decrease was more pronounced in DBA/2J mice. Furthermore, quantification of the total number of RGCs identified by OHSt and Brn3a expression showed a reduced population of RGCs in DBA/2J mice as compared to control mice. Regression analysis revealed significant correlations between the decrease in pSTR and a non-homogeneous reduction in the number of RGCs throughout the retina. Our results indicate the existence of a correlation between retinal function impairment and RGC loss. This functional and morphological analysis allows a reliable assessment of the progression of the disease.
Collapse
Affiliation(s)
- María J Pérez de Lara
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037 Madrid, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain
| | - Concepción Santano
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037 Madrid, Spain
| | - Ana Guzmán-Aránguez
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037 Madrid, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain
| | - F Javier Valiente-Soriano
- Experimental Ophthalmology Laboratory, Dept. of Ophthalmology, College of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", Murcia Institute of Bio-Health Research (IMIB), E-30100 Murcia, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain
| | - Marcelino Avilés-Trigueros
- Experimental Ophthalmology Laboratory, Dept. of Ophthalmology, College of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", Murcia Institute of Bio-Health Research (IMIB), E-30100 Murcia, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain
| | - Manuel Vidal-Sanz
- Experimental Ophthalmology Laboratory, Dept. of Ophthalmology, College of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", Murcia Institute of Bio-Health Research (IMIB), E-30100 Murcia, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain
| | - Pedro de la Villa
- Department of Systems Biology, University of Alcalá, 28871 Alcalá de Henares, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037 Madrid, Spain; Red Española Temática de Investigación Cooperativa en Patología Ocular Prevalente y Crónica, Instituto de Salud Carlos III, Madrid, PC, Spain.
| |
Collapse
|
34
|
Chua B, Goldberg I. Neuroprotective agents in glaucoma therapy: recent developments and future directions. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.10.55] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Enhanced expression of NR2B subunits of NMDA receptors in the inherited glaucomatous DBA/2J mouse retina. Neural Plast 2013; 2013:670254. [PMID: 24175101 PMCID: PMC3794641 DOI: 10.1155/2013/670254] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/14/2013] [Indexed: 01/12/2023] Open
Abstract
DBA/2J mouse has been used as a model for spontaneous secondary glaucoma. Here, we investigated changes in expression of NMDA receptor (NMDAR) subunits and Cdk5/p35/NMDAR signaling in retinas of DBA/2J mice using Western blot technique. The protein levels of NR1 and NR2A subunits in retinas of DBA/2J mice at all ages (6–12 months) were not different from those in age-matched C57BL/6 mice. In contrast, the protein levels of NR2B subunits, in addition to age-dependent change, significantly increased with elevated intraocular pressure (IOP) in DBA/2J mice at 6 and 9 months as compared with age-matched controls. Moreover, expression of Cdk5, p35 and ratio of p-NR2AS1232/NR2A progressively increased with time in both strains, suggestive of activated Cdk5/p35 signaling pathway. However, the changes in these proteins were in the same levels in both strain mice, except a significant increase of p35 proteins at 6 months in DBA/2J mice. Meanwhile, the protein levels of Brn-3a, a retinal ganglion cell (RGC) maker, remarkably decreased at 9–12 months in DBA/2J mice, which was in parallel with the changes of NR2B expression. Our results suggest that elevated IOP-induced increase in expression of NR2B subunits of NMDARs may be involved in RGC degeneration of DBA/2J mice.
Collapse
|
36
|
|
37
|
Howell GR, Soto I, Ryan M, Graham LC, Smith RS, John SWM. Deficiency of complement component 5 ameliorates glaucoma in DBA/2J mice. J Neuroinflammation 2013; 10:76. [PMID: 23806181 PMCID: PMC3708765 DOI: 10.1186/1742-2094-10-76] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/12/2013] [Indexed: 01/08/2023] Open
Abstract
Background Glaucoma is an age-related neurodegenerative disorder involving the loss of retinal ganglion cells (RGCs), which results in blindness. Studies in animal models have shown that activation of inflammatory processes occurs early in the disease. In particular, the complement cascade is activated very early in DBA/2J mice, a widely used mouse model of glaucoma. A comprehensive analysis of the role of the complement cascade in DBA/2J glaucoma has not been possible because DBA/2J mice are naturally deficient in complement component 5 (C5, also known as hemolytic complement, Hc), a key mediator of the downstream processes of the complement cascade, including the formation of the membrane attack complex. Methods To assess the role of C5 in DBA/2J glaucoma, we backcrossed a functional C5 gene from strain C57BL/6J to strain DBA/2J for at least 10 generations. The prevalence and severity of glaucoma was evaluated using ocular examinations, IOP measurements, and assessments of optic nerve damage and RGC degeneration. To understand how C5 affects glaucoma, C5 expression was assessed in the retinas and optic nerves of C5-sufficient DBA/2J mice, using immunofluorescence. Results C5-sufficient DBA/2J mice developed a more severe glaucoma at an earlier age than standard DBA/2J mice, which are therefore protected by C5 deficiency. Components of the membrane attack complex were found to be deposited at sites of axonal injury in the optic nerve head and associated with RGC soma in the retina. Conclusion C5 plays an important role in glaucoma, with its deficiency lessening disease severity. These results highlight the importance of fully understanding the role of the complement cascade in neurodegenerative diseases. Inhibiting C5 may be beneficial as a therapy for human glaucoma.
Collapse
Affiliation(s)
- Gareth R Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Inman DM, Lambert WS, Calkins DJ, Horner PJ. α-Lipoic acid antioxidant treatment limits glaucoma-related retinal ganglion cell death and dysfunction. PLoS One 2013; 8:e65389. [PMID: 23755225 PMCID: PMC3673940 DOI: 10.1371/journal.pone.0065389] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 04/30/2013] [Indexed: 01/21/2023] Open
Abstract
Oxidative stress has been implicated in neurodegenerative diseases, including glaucoma. However, due to the lack of clinically relevant models and expense of long-term testing, few studies have modeled antioxidant therapy for prevention of neurodegeneration. We investigated the contribution of oxidative stress to the pathogenesis of glaucoma in the DBA/2J mouse model of glaucoma. Similar to other neurodegenerative diseases, we observed lipid peroxidation and upregulation of oxidative stress-related mRNA and protein in DBA/2J retina. To test the role of oxidative stress in disease progression, we chose to deliver the naturally occurring, antioxidant α-lipoic acid (ALA) to DBA/2J mice in their diet. We used two paradigms for ALA delivery: an intervention paradigm in which DBA/2J mice at 6 months of age received ALA in order to intervene in glaucoma development, and a prevention paradigm in which DBA/2J mice were raised on a diet supplemented with ALA, with the goal of preventing glaucoma development. At 10 and 12 months of age (after 4 and 11 months of dietary ALA respectively), we measured changes in genes and proteins related to oxidative stress, retinal ganglion cell (RGC) number, axon transport, and axon number and integrity. Both ALA treatment paradigms showed increased antioxidant gene and protein expression, increased protection of RGCs and improved retrograde transport compared to control. Measures of lipid peroxidation, protein nitrosylation, and DNA oxidation in retina verified decreased oxidative stress in the prevention and intervention paradigms. These data demonstrate the utility of dietary therapy for reducing oxidative stress and improving RGC survival in glaucoma.
Collapse
Affiliation(s)
- Denise M Inman
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States of America.
| | | | | | | |
Collapse
|
39
|
Functional protective effects of long-term memantine treatment in the DBA/2J mouse. Doc Ophthalmol 2013; 126:221-32. [DOI: 10.1007/s10633-013-9380-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 03/04/2013] [Indexed: 01/02/2023]
|
40
|
Rejdak R, Junemann A, Grieb P, Thaler S, Schuettauf F, Chorągiewicz T, Zarnowski T, Turski WA, Zrenner E. Kynurenic acid and kynurenine aminotransferases in retinal aging and neurodegeneration. Pharmacol Rep 2012; 63:1324-34. [PMID: 22358081 DOI: 10.1016/s1734-1140(11)70697-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 06/24/2011] [Indexed: 10/25/2022]
Abstract
The kynurenine aminotransferases (KATs) KAT I and KAT II are pivotal to the synthesis of kynurenic acid (KYNA), the only known endogenous glutamate receptor antagonist and neuroprotectant. KAT I and II have been found in avian, rodent, and human retina. Expression of KAT I in Müller cell endfeet and KAT II in retinal ganglion cells has been documented. Developmental changes in KAT expression and KYNA concentration in the avian and rodent retina have also been found. Studies of retinal neurodegeneration have shown alterations in KYNA synthesis in the retina in response to retinal ganglion cell loss. In DBA/2J mice, a model of ocular hypertension, an age-dependent decrease of retinal KYNA and KATs was found. In the corpora amylacea in the human retina intensive KAT I and II immunoreactivity was demonstrated. In summary, these findings point to the potential involvement of KYNA in the mechanisms of retinal aging and neurodegeneration.
Collapse
Affiliation(s)
- Robert Rejdak
- Centre for Ophthalmology, University of Tübingen, Roentgenweg 11, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Unterlauft JD, Eichler W, Kuhne K, Yang XM, Yafai Y, Wiedemann P, Reichenbach A, Claudepierre T. Pigment epithelium-derived factor released by Müller glial cells exerts neuroprotective effects on retinal ganglion cells. Neurochem Res 2012; 37:1524-33. [PMID: 22410737 PMCID: PMC3368109 DOI: 10.1007/s11064-012-0747-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Revised: 02/22/2012] [Accepted: 02/27/2012] [Indexed: 01/25/2023]
Abstract
Survival of retinal ganglion cells (RGC) is compromised in several vision-threatening disorders such as ischemic and hypertensive retinopathies and glaucoma. Pigment epithelium-derived factor (PEDF) is a naturally occurring pleiotropic secreted factor in the retina. PEDF produced by retinal glial (Müller) cells is suspected to be an essential component of neuron-glial interactions especially for RGC, as it can protect this neuronal type from ischemia-induced cell death. Here we show that PEDF treatment can directly affect RGC survival in vitro. Using Müller cell-RGC-co-cultures we observed that activity of Müller-cell derived soluble mediators can attenuate hypoxia-induced damage and RGC loss. Finally, neutralizing the activity of PEDF in glia-conditioned media partially abolished the neuroprotective effect of glia, leading to an increased neuronal death in hypoxic condition. Altogether our results suggest that PEDF is crucially involved in the neuroprotective process of reactive Müller cells towards RGC.
Collapse
Affiliation(s)
- Jan Darius Unterlauft
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Liebigstrasse 10-14, 04103, Leipzig, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Primary open angle glaucoma and Alzheimer’s disease have long been established as two separate pathological entities, primarily affecting the elderly. The progressive, irreversible course of both diseases has significant implications on an aging population. As the complex pathophysiology of the two diseases has progressively unraveled over the past two decades, common pathophysiological changes have also been elucidated. Some of these mechanisms have established a strong grounding, whilst others remain principally speculative. The mutual neuropathological changes in primary open angle glaucoma and Alzheimer’s disease have facilitated the development of neuroprotective strategies. While most of these strategies are still in the preclinical phase, they have shown great promise in experimental animal studies. Further understanding of the common pathophysiology of primary open angle glaucoma and Alzheimer’s disease and their timeline may have great implications on early diagnosis and effective therapeutic targeting.
Collapse
Affiliation(s)
- Mukhtar Bizrah
- Glaucoma & Retinal Neurodegeneration Research Group, Visual Neuroscience, UCL Institute of Ophthalmology, London, UK; Western Eye Hospital, Imperial College Healthcare Trust, London, UK
- Guy’s & St Thomas’ NHS Foundation Trust, London, UK
| | - Li Guo
- Glaucoma & Retinal Neurodegeneration Research Group, Visual Neuroscience, UCL Institute of Ophthalmology, London, UK; Western Eye Hospital, Imperial College Healthcare Trust, London, UK
| | - Maria Francesca Cordeiro
- Glaucoma & Retinal Neurodegeneration Research Group, Visual Neuroscience, UCL Institute of Ophthalmology, London, UK; Western Eye Hospital, Imperial College Healthcare Trust, London, UK
| |
Collapse
|
43
|
Crish SD, Calkins DJ. Neurodegeneration in glaucoma: progression and calcium-dependent intracellular mechanisms. Neuroscience 2010; 176:1-11. [PMID: 21187126 DOI: 10.1016/j.neuroscience.2010.12.036] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/16/2010] [Accepted: 12/18/2010] [Indexed: 01/18/2023]
Abstract
Glaucoma is an age-related optic neuropathy involving sensitivity to ocular pressure. The disease is now seen increasingly as one of the central nervous system, as powerful new approaches highlight an increasing number of similarities with other age-related neurodegenerations such as Alzheimer's and Parkinson's. While the etiologies of these diseases are diverse, they involve many important common elements including compartmentalized programs of degeneration targeting axons, dendrites and finally cell bodies. Most age-related degenerations display early functional deficits that precede actual loss of neuronal substrate. These are linked to several specific neurochemical cascades that can be linked back to dysregulation of Ca(2+)-dependent processes. We are now in the midst of identifying similar cascades in glaucoma. Here we review recent evidence on the pathological progression of neurodegeneration in glaucoma and some of the Ca(2+)-dependent mechanisms that could underlie these changes. These mechanisms present clear implications for efforts to develop interventions targeting neuronal loss directly and make glaucoma an attractive model for both interrogating and informing other neurodegenerative diseases.
Collapse
Affiliation(s)
- S D Crish
- Department of Pharmaceutical Sciences, Northeastern Ohio Universities Colleges of Medicine and Pharmacy, Rootstown, OH 44272, USA
| | | |
Collapse
|
44
|
Abstract
The N-methyl-D-aspartate receptor (NMDAR) co-agonist D-serine is important in a number of different processes in the CNS, ranging from synaptic plasticity to disease states, including schizophrenia. D-serine appears to be the major co-agonist acting on retinal ganglion cell NMDA receptors, but the cell type from which it originates and whether its release can be modulated by activity are unknown. In this study, we utilized a mutant mouse line with elevated d-serine to investigate this question. Direct measurements of extracellular D-serine using capillary electrophoresis demonstrate that D-serine can be released from the intact mouse retina through an α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR) dependent mechanism. α-Amino-3-hydroxyl-5-methyl-4-isoxazole-propionate-evoked D-serine release persisted in the presence of a cocktail of neural inhibitors but was abolished after administration of a glial toxin. These findings provide the first evidence that extracellular D-serine levels in the retina can be modulated, and that such modulation is contingent upon glial cell activity.
Collapse
Affiliation(s)
- Steve J Sullivan
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA.
| | | |
Collapse
|
45
|
Heiduschka P, Julien S, Schuettauf F, Schnichels S. Loss of retinal function in aged DBA/2J mice - New insights into retinal neurodegeneration. Exp Eye Res 2010; 91:779-83. [PMID: 20832401 DOI: 10.1016/j.exer.2010.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 08/27/2010] [Accepted: 09/01/2010] [Indexed: 02/07/2023]
Abstract
The DBA/2J mouse is a common animal model of glaucoma. The intraocular pressure increases with age, and retinal ganglion cells (RGC) degenerate, usually starting at an age of approximately six months. In this study, we used two-year-old DBA/2J mice presuming an end-point of RGC degeneration. We investigated visual function in these animals using electroretinography (ERG) and visual evoked potentials (VEP), and we checked the number of remaining RGC by retrograde staining. Almost no RGC were left in the retina, and VEP were hardly recordable. Surprisingly, also ERG amplitudes of scotopic a-waves and b-waves, photopic b-waves and oscillatory potentials were decreased significantly by approximately 40% compared to amplitudes measured in age-matched C57BL/6J mice. The latencies were not changed in DBA/2J mice compared to C57BL/6J mice, and so were the ratios between amplitudes of a-waves, b-waves and oscillatory potentials. Our results indicate that, in addition to degeneration of RGC, also photoreceptors are affected by pathological processes in the eye caused by the mutations present in DBA/2J mice.
Collapse
|
46
|
Abstract
The development of treatments that slow photoreceptor death could profoundly improve patient wellbeing in those with inherited retinal degenerations. Over recent years, it has emerged that extracellular adenosine-tri-phosphate (ATP) regulates the function of photoreceptors in rodents and primates. Moreover, when the retina is exposed to high levels of ATP, rapid death of photoreceptors occurs, which can be blocked by pretreatment with antagonists to P2X receptors. Compounds that inhibit the action of extracellular ATP slow photoreceptor loss in an animal model of inherited retinal degeneration. In this article, I provide an overview of our work in relation to other research in this area and suggest a model by which ATP contributes to photoreceptor death in inherited retinal degenerations.
Collapse
|
47
|
Rammes G, Danysz W, Parsons CG. Pharmacodynamics of memantine: an update. Curr Neuropharmacol 2010; 6:55-78. [PMID: 19305788 PMCID: PMC2645549 DOI: 10.2174/157015908783769671] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 05/10/2007] [Accepted: 11/05/2007] [Indexed: 01/12/2023] Open
Abstract
Memantine received marketing authorization from the European Agency for the Evaluation of Medicinal Products (EMEA) for the treatment of moderately severe to severe Alzheimer s disease (AD) in Europe on 17(th) May 2002 and shortly thereafter was also approved by the FDA for use in the same indication in the USA. Memantine is a moderate affinity, uncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist with strong voltage-dependency and fast kinetics. Due to this mechanism of action (MOA), there is a wealth of other possible therapeutic indications for memantine and numerous preclinical data in animal models support this assumption. This review is intended to provide an update on preclinical studies on the pharmacodynamics of memantine, with an additional focus on animal models of diseases aside from the approved indication. For most studies prior to 1999, the reader is referred to a previous review [196].In general, since 1999, considerable additional preclinical evidence has accumulated supporting the use of memantine in AD (both symptomatic and neuroprotective). In addition, there has been further confirmation of the MOA of memantine as an uncompetitive NMDA receptor antagonist and essentially no data contradicting our understanding of the benign side effect profile of memantine.
Collapse
Affiliation(s)
- G Rammes
- Clinical Neuropharmacology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | | |
Collapse
|
48
|
A Thy1-CFP DBA/2J mouse line with cyan fluorescent protein expression in retinal ganglion cells. Vis Neurosci 2009; 26:453-65. [PMID: 19930759 DOI: 10.1017/s095252380999023x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A DBA/2J (D2) transgenic mouse line with cyan fluorescent protein (CFP) reporter expression in ganglion cells was developed for the analysis of ganglion cells during progressive glaucoma. The Thy1-CFP D2 (CFP-D2) line was created by congenically breeding the D2 line, which develops pigmentary glaucoma, and the Thy1-CFP line, which expresses CFP in ganglion cells. Microsatellite marker analysis of CFP-D2 progeny verified the genetic inclusion of the D2 isa and ipd loci. Specific mutations within these loci lead to dysfunctional melanosomal proteins and glaucomatous phenotype in D2 mice. Polymerase chain reaction analysis confirmed the inclusion of the Thy1-CFP transgene. CFP-fluorescent ganglion cells, 6-20 microm in diameter, were distributed in all retinal regions, CFP processes were throughout the inner plexiform layer, and CFP-fluorescent axons were in the fiber layer and optic nerve head. Immunohistochemistry with antibodies to ganglion cell markers NF-L, NeuN, Brn3a, and SMI32 was used to confirm CFP expression in ganglion cells. Immunohistochemistry with antibodies to amacrine cell markers HPC-1 and ChAT was used to confirm weak CFP expression in cholinergic amacrine cells. CFP-D2 mice developed a glaucomatous phenotype, including iris disease, ganglion cell loss, attrition of the fiber layer, and elevated intraocular pressure. A CFP-D2 transgenic line with CFP-expressing ganglion cells was developed, which has (1) a predominantly D2 genetic background, (2) CFP-expressing ganglion cells, and (3) age-related progressive glaucoma. This line will be of value for experimental studies investigating ganglion cells and their axons in vivo and in vitro during the progressive development of glaucoma.
Collapse
|
49
|
Thaler S, Fiedorowicz M, Rejdak R, Choragiewicz TJ, Sulejczak D, Stopa P, Zarnowski T, Zrenner E, Grieb P, Schuettauf F. Neuroprotective effects of tempol on retinal ganglion cells in a partial optic nerve crush rat model with and without iron load. Exp Eye Res 2009; 90:254-60. [PMID: 19883642 DOI: 10.1016/j.exer.2009.10.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 10/23/2009] [Accepted: 10/23/2009] [Indexed: 12/31/2022]
Abstract
Iron overload can contribute to oxidative stress in many tissues. We studied the effects of pretreatment with iron dextran on RGC loss in a calibrated partial optic nerve crush (PONC) model in rats, along with the protection offered by tempol (4-hydroxy-2,2,6,6-tetramethylpiperidinyl-1-oxyl, a membrane-permeable superoxide dismutase mimetic and free-radical scavenger), in the same experimental paradigm. A total of 40 rats in 6 groups of 5-8 animals each underwent PONC in one eye and sham crush in the other. Animals were pretreated with a single iron dextran load 24 h prior to PONC, and treated with tempol 6 h before and then once daily after PONC. Control animals were treated with PBS. RGC were retrogradely labeled with a fluorescent marker; all data are expressed in percent of the RGC count in the respective sham-treated eye. Immunohistochemistry was performed to visualize 3-nitrotyrosine, a marker of nitroxidative stress. PONC without iron pretreatment resulted in the survival of only 31.4% of labeled RGC after 7 days. Even fewer RGC (12.7%) survived after PONC with iron pretreatment. However, tempol in doses of 20 mg/kg of body weight (BW) significantly attenuated this effect when given as described above; in the group without iron pretreatment the number of surviving RGC doubled from 31.4% to 62.1%. In the group with iron pretreatment the survival rate of RGC increased even more pronouncedly, from 12.7% without tempol to 46.2% with tempol. Tempol in doses of 1 mg/kg BW and 5 mg/kg BW showed no significant rescue of RGC. Immunostaining showed nitrotyrosine-positive RGCs in PONC but not in sham-treated eyes and an increase in positive cells after iron load. Tempol treatment reduced nitrotyrosine staining in both the iron and non-iron groups. Our results demonstrate that PONC results in significantly greater RGC damage when iron pretreatment is performed, and that the compound tempol may provide additional protection for RGC in cases of neuronal damage both with and without prior iron treatment.
Collapse
Affiliation(s)
- Sebastian Thaler
- Centre for Ophthalmology, University of Tübingen, Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wang X, Archibald ML, Stevens K, Baldridge WH, Chauhan BC. Cyan fluorescent protein (CFP) expressing cells in the retina of Thy1-CFP transgenic mice before and after optic nerve injury. Neurosci Lett 2009; 468:110-4. [PMID: 19879331 DOI: 10.1016/j.neulet.2009.10.077] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2009] [Revised: 10/14/2009] [Accepted: 10/23/2009] [Indexed: 11/18/2022]
Abstract
We investigated the specificity of cyan fluorescent protein (CFP) expression in retinal ganglion cells (RGCs) of the transgenic Thy1-CFP (B6.Cg-Tg(Thy1-CFP)23Jrs/J) mouse line, and the characteristics of these cells after optic nerve injury. RGCs of adult Thy1-CFP mice were retrogradely labeled with fluorochrome (2% fluorogold [FG]) from the superior colliculi (SC). Animals were sacrificed 7 days after RGC labeling. Retinas were fixed and whole-mounted. CFP and FG-positive cells were visualized and imaged separately. Cells positive for CFP, FG, or co-labeled were counted. In another group of animals, the left optic nerves were transected 7 days after FG labeling. They were sacrificed 7 or 21 days after transection. The retinas were whole-mounted and the characteristics of CFP-expressing cells examined. CFP-expressing cells were distributed evenly throughout the retinas of Thy1-CFP mice. The average densities of CFP and FG-positive cells in the retina were 2778+/-216 and 3230+/-157 cells/mm(2), respectively. 93.2+/-1.6% of CFP-expressing cells were also labeled with FG. However, only 79.9+/-2.5% of FG-labeled RGCs expressed CFP. The number of CFP-expressing cells decreased dramatically after transection. Cells with spindle shape, immunohistochemically identified as microglia, were seen in the retina with CFP expression at both 7 and 21 days after optic nerve transection. In retinas of Thy1-CFP mice, CFP is expressed by the large majority of RGCs, but not exclusively by RGCs. CFP is internalized by phagocytosing cells after injury to RGCs.
Collapse
Affiliation(s)
- Xu Wang
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | | |
Collapse
|