1
|
Tanaka S, Soga N, Hirono K, Okawa K, Chin JYH, Kitahata S, Inoue T, Maruyama-Inoue M, Kadonosono K. TIME-COURSE RETINAL CIRCULATION CHANGES IN EYES WITH CENTRAL RETINAL ARTERY OCCLUSION USING LASER SPECKLE FLOWGRAPHY. Retina 2025; 45:133-140. [PMID: 39236301 DOI: 10.1097/iae.0000000000004264] [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: 09/07/2024]
Abstract
PURPOSE To evaluate retinal circulation in eyes with central retinal artery occlusion. METHODS The best-corrected visual acuity (BCVA), fluorescein angiography, and laser speckle flowgraphy values were measured at the first visit in 42 patients with central retinal artery occlusion (42 eyes; mean age, 66.0 ± 14.0 years). Laser speckle flowgraphy was performed at baseline, 1 week, and 1, 3, and 6 months; the difference between the mean blur rate vessel area and mean blur rate tissue area (MV-MT), reflecting the retinal vessel blood flow, was measured. Fluorescein angiography measured the arteriovenous passage time. Time-course changes in the MV-MT and BCVA, and the relationship between MV-MT and final BCVA were examined. RESULTS Mean blur rate vessel area and mean blur rate tissue area were significantly correlated with arteriovenous passage time ( P < 0.001). The MV-MT increased significantly after 1, 3, and 6 months ( P < 0.001). The visual acuity improved significantly until 1 month ( P < 0.01). Multivariate analysis showed a significant correlation between the baseline BCVA ( P < 0.001) and MV-MT at 1 month ( P = 0.01) and the final BCVA. Age and MV-MT significantly negatively correlated at 1 month ( P < 0.001). CONCLUSION The retinal circulation improved significantly within 1 month; younger patients had better retinal circulation improvement. The baseline BCVA and retinal circulation within 1 month correlated with the final BCVA.
Collapse
Affiliation(s)
- Shin Tanaka
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Naoki Soga
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Kazushi Hirono
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Kazuyoshi Okawa
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Jacob Y H Chin
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - Shohei Kitahata
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Tatsuya Inoue
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Maiko Maruyama-Inoue
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| | - Kazuaki Kadonosono
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan ; and
| |
Collapse
|
2
|
Baranykova S, Gupta RK, Kajdasz A, Wasilewska I, Macias M, Szybinska A, Węgierski T, Nahia KA, Mondal SS, Winata CL, Kuźnicki J, Majewski L. Loss of Stim2 in zebrafish induces glaucoma-like phenotype. Sci Rep 2024; 14:24442. [PMID: 39424970 PMCID: PMC11489432 DOI: 10.1038/s41598-024-74909-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Calcium is involved in vision processes in the retina and implicated in various pathologies, including glaucoma. Rod cells rely on store-operated calcium entry (SOCE) to safeguard against the prolonged lowering of intracellular calcium ion concentrations. Zebrafish that lacked the endoplasmic reticulum Ca2+ sensor Stim2 (stim2 knockout [KO]) exhibited impaired vision and lower light perception-related gene expression. We sought to understand mechanisms that are responsible for vision impairment in stim2 KO zebrafish. The single-cell RNA (scRNA) sequencing of neuronal cells from brains of 5 days postfertilization larvae distinguished 27 cell clusters, 10 of which exhibited distinct gene expression patterns, including amacrine and γ-aminobutyric acid (GABA)ergic retinal interneurons and GABAergic optic tectum cells. Five clusters exhibited significant changes in cell proportions between stim2 KO and controls, including GABAergic diencephalon and optic tectum cells. Transmission electron microscopy of stim2 KO zebrafish revealed decreases in width of the inner plexiform layer, ganglion cells, and their dendrites numbers (a hallmark of glaucoma). GABAergic neuron densities in the inner nuclear layer, including amacrine cells, as well as photoreceptors significantly decreased in stim2 KO zebrafish. Our study suggests a novel role for Stim2 in the regulation of neuronal insulin expression and GABAergic-dependent vision causing glaucoma-like retinal pathology.
Collapse
Affiliation(s)
- Sofiia Baranykova
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Rishikesh Kumar Gupta
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, 201313, India
| | - Arkadiusz Kajdasz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704, Poznan, Poland
- Xenstats sp. z o.o., Otwarta 1, 60-008, Poznan, Poland
| | - Iga Wasilewska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Adolfa Pawińskiego 5, 02-106, Warsaw, Poland
| | - Matylda Macias
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, 02-109, WarsawWarsaw, Poland
| | - Aleksandra Szybinska
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, 02-109, WarsawWarsaw, Poland
| | - Tomasz Węgierski
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, 02-109, WarsawWarsaw, Poland
| | - Karim Abu Nahia
- Laboratory of Zebrafish Developmental Genomics, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Shamba S Mondal
- Laboratory of Zebrafish Developmental Genomics, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Cecilia L Winata
- Laboratory of Zebrafish Developmental Genomics, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Jacek Kuźnicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland
| | - Lukasz Majewski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, 02-109, Warsaw, Poland.
| |
Collapse
|
3
|
Haque A, Trager NNM, Butler JT, Das A, Zaman V, Banik NL. A novel combination approach to effectively reduce inflammation and neurodegeneration in multiple sclerosis models. Neurochem Int 2024; 175:105697. [PMID: 38364938 PMCID: PMC10994736 DOI: 10.1016/j.neuint.2024.105697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by immune-mediated attacks on the central nervous system (CNS), resulting in demyelination and recurring T-cell responses. Unfortunately, there is no cure for it. Current therapies that target immunomodulation and/or immunosuppression show only modest beneficial effects, have many side effects, and do not block neurodegeneration or progression of the disease. Since neurodegeneration and in particular axonal degeneration is implicated in disability in progressive MS, development of novel therapeutic strategies to attenuate the neurodegenerative processes is imperative. This study aims to develop new safe and efficacious treatments that address both the inflammatory and neurodegenerative aspects of MS using its animal model, experimental allergic encephalomyelitis (EAE). In EAE, the cysteine protease calpain is upregulated in CNS tissue, and its activity correlates with neurodegeneration. Our immunologic studies on MS have indicated that increased calpain activity promotes pro-inflammatory T helper (Th)1 cells and the severity of the disease in EAE, suggesting that calpain inhibition could be a novel target to combat neurodegeneration in MS/EAE. While calpain inhibition by SNJ1945 reduced disease severity, treatment of EAE animals with a novel protease-resistant altered small peptide ligand (3aza-APL) that mimic myelin basic protein (MBP), also decreased the incidence of EAE, disease severity, infiltration of inflammatory cells, and protected myelin. A reduction in inflammatory T-cells with an increase in Tregs and myeloid suppressor cells is also found in EAE mice treated with SNJ1945 and 3aza-APL. Thus, a novel combination strategy was tested in chronic EAE mouse model in B10 mice which showed multiple pathological mechanisms could be addressed by simultaneous treatment with calpain inhibitor SNJ1945 and protease-resistant 3aza-APL to achieve a stronger therapeutic effect.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, 29401, USA.
| | - Nicole N M Trager
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jonathan T Butler
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Arabinda Das
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, 29401, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, 29401, USA.
| |
Collapse
|
4
|
Feng L, Dai S, Zhang C, Zhang W, Zhu W, Wang C, He Y, Song W. Ripa-56 protects retinal ganglion cells in glutamate-induced retinal excitotoxic model of glaucoma. Sci Rep 2024; 14:3834. [PMID: 38360971 PMCID: PMC10869350 DOI: 10.1038/s41598-024-54075-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/08/2024] [Indexed: 02/17/2024] Open
Abstract
Glaucoma is a prevalent cause of blindness globally, characterized by the progressive degeneration of retinal ganglion cells (RGCs). Among various factors, glutamate excitotoxicity stands out as a significant contributor of RGCs loss in glaucoma. Our study focused on Ripa-56 and its protective effect against NMDA-induced retinal damage in mice, aiming to delve into the potential underlying mechanism. The R28 cells were categorized into four groups: glutamate (Glu), Glu + Ripa-56, Ripa-56 and Control group. After 24 h of treatment, cell death was assessed by PI / Hoechst staining. Mitochondrial membrane potential changes, apoptosis and reactive oxygen species (ROS) production were analyzed using flow cytometry. The alterations in the expression of RIP-1, p-MLKL, Bcl-2, BAX, Caspase-3, Gpx4 and SLC7A11 were examined using western blot analysis. C57BL/6j mice were randomly divided into NMDA, NMDA + Ripa-56, Ripa-56 and control groups. Histological changes in the retina were evaluated using hematoxylin and eosin (H&E) staining. RGCs survival and the protein expression changes of RIP-1, Caspase-3, Bcl-2, Gpx4 and SLC7A11 were observed using immunofluorescence. Ripa-56 exhibited a significant reduction in the levels of RIP-1, p-MLKL, Caspase-3, and BAX induced by glutamate, while promoting the expression of Bcl-2, Gpx-4, and SLC7A1 in the Ripa-56-treated group. In our study, using an NMDA-induced normal tension glaucoma mice model, we employed immunofluorescence and H&E staining to observe that Ripa-56 treatment effectively ameliorated retinal ganglion cell loss, mitigating the decrease in retinal ganglion cell layer and bipolar cell layer thickness caused by NMDA. In this study, we have observed that Ripa-56 possesses remarkable anti- necroptotic, anti-apoptotic and anti-ferroptosis properties. It demonstrates the ability to combat not only glutamate-induced excitotoxicity in R28 cells, but also NMDA-induced retinal excitotoxicity in mice. Therefore, Ripa-56 could be used as a potential retinal protective agent.
Collapse
Affiliation(s)
- Lemeng Feng
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Shirui Dai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, People's Republic of China
| | - Cheng Zhang
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Wulong Zhang
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Weiming Zhu
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Chao Wang
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Ye He
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, People's Republic of China
| | - Weitao Song
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
5
|
Wada S, Tsuda S, Abe M, Nakazawa T, Urushihara H. A quality management system aiming to ensure regulatory-grade data quality in a glaucoma registry. PLoS One 2023; 18:e0286669. [PMID: 37267325 DOI: 10.1371/journal.pone.0286669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/20/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Disease/patient registries are underutilized despite their multiple advantages over clinical trials in the clinical evaluation of drugs, such as the capacity for long-term curation, provision of patient outcome data in routine clinical practice, and provision of benchmark data for comparison. Ensuring the fit-for-purpose quality of data generated from such registries is important to informing regulatory decision making. Here, we report the construction of a quality management system aiming to ensure regulatory-grade data quality for a registry of Japanese patients with glaucoma to evaluate long-term patient outcomes. METHODS The quality management system was established by reference to the risk-based approach in the ICH-E6 (R2) recommendations. The following three-component approach was taken: establishment of governance, computerized system validation (CSV), and implementation of risk assessment and control. Compliance of the system with the recommendations of regulatory guidelines relevant to use of the registry was assessed. RESULTS Governance by academic collaboration was established. This was followed by the development of a total of 15 standard operating procedures, including CSV, data management, monitoring, audit, and management of imaging data. The data management system was constructed based on a data management plan, which specified data/paper flow and data management procedures. The electronic data capture (EDC) system was audited by an external vendor, and configured and validated using the V-model framework as recommended in the GAMP5 guideline. Informed consent, eligibility assessment and major ophthalmology measurements were determined as Critical to Quality (CTQ) factors. A total of 22 risk items were identified and classified into three categories, and operationalized in the form of a risk control plan, which included training sessions and risk-based monitoring. The glaucoma registry addressed most quality recommendations in official guidelines issued by multiple health authorities, although two recommendations were not met. CONCLUSIONS We established and configured a quality management system for a glaucoma registry to ensure fit-for-purpose data quality for regulatory use, and to curate long-term follow-up data of glaucoma patients in a prospective manner.
Collapse
Affiliation(s)
- Shinsuke Wada
- Division of Drug Development and Regulatory Science, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Satoru Tsuda
- Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Maiko Abe
- Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, Japan
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisashi Urushihara
- Division of Drug Development and Regulatory Science, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| |
Collapse
|
6
|
Kotova IM, Pestereva NS, Traktirov DS, Absalyamova MT, Karpenko MN. Functions and distribution of calpain-calpastatin system components in brain during mammal ontogeny. Biochim Biophys Acta Gen Subj 2023; 1867:130345. [PMID: 36889447 DOI: 10.1016/j.bbagen.2023.130345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
Calpain and calpastatin are the key components of the calcium-dependent proteolytic system. Calpains are regulatory, calcium-dependent, cytoplasmic proteinases, and calpastatin is the endogenous inhibitor of calpains. Due to the correlation between changes in the activity of the calpain-calpastatin system in the brain and central nervous system (CNS) pathology states, this proteolytic system is a prime focus of research on CNS pathological processes, generally characterized by calpain activity upregulation. The present review aims to generalize existing data on cerebral calpain distribution and function through mammalian ontogenesis. Special attention is given to the most recent studies on the topic as more information on calpain-calpastatin system involvement in normal CNS development and functioning has become available. We also discuss data on calpain and calpastatin activity and production in different brain regions during ontogenesis as comparative analysis of these results in association with ontogeny processes can reveal brain regions and developmental stages with pronounced function of the calpain system.
Collapse
Affiliation(s)
- Irina M Kotova
- Institute of Experimental Medicine, St. Petersburg, Russia
| | | | | | | | | |
Collapse
|
7
|
Kiyota N, Namekata K, Nishijima E, Guo X, Kimura A, Harada C, Nakazawa T, Harada T. Effects of constitutively active K-Ras on axon regeneration after optic nerve injury. Neurosci Lett 2023; 799:137124. [PMID: 36780941 DOI: 10.1016/j.neulet.2023.137124] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/24/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
Visual disturbance after optic nerve injury is a serious problem. Attempts have been made to enhance the intrinsic ability of retinal ganglion cells (RGCs) to regenerate their axons, and the importance of PI3K/Akt and RAF/MEK/ERK signal activation has been suggested. Since these signals are shared with oncogenic signaling cascades, in this study, we focused on a constitutively active form of K-Ras, K-RasV12, to determine if overexpression of this molecule could stimulate axon regeneration. We confirmed that K-RasV12 phosphorylated Akt and ERK in vitro. Intravitreal delivery of AAV2-K-RasV12 increased the number of surviving RGCs and promoted 1.0 mm of axon regeneration one week after optic nerve injury without inducing abnormal proliferative effects in the RGCs. In addition, AAV2-K-RasV12 induced robust RGC axon regeneration, reaching as far as approximately 2.5 mm from the injury site, in eight weeks. Our findings suggest that AAV2-K-RasV12 could provide a good model for speedy and efficient analysis of the mechanism underlying axon regeneration in vivo.
Collapse
Affiliation(s)
- Naoki Kiyota
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Euido Nishijima
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
8
|
Calpains as mechanistic drivers and therapeutic targets for ocular disease. Trends Mol Med 2022; 28:644-661. [PMID: 35641420 PMCID: PMC9345745 DOI: 10.1016/j.molmed.2022.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022]
Abstract
Ophthalmic neurodegenerative diseases encompass a wide array of molecular pathologies unified by calpain dysregulation. Calpains are calcium-dependent proteases that perpetuate cellular death and inflammation when hyperactivated. Calpain inhibition trials in other organs have faced pharmacological challenges, but the eye offers many advantages for the development and testing of targeted molecular therapeutics, including small molecules, peptides, engineered proteins, drug implants, and gene-based therapies. This review highlights structural mechanisms underlying calpain activation, distinct cellular expression patterns, and in vivo models that link calpain hyperactivity to human retinal and developmental disease. Optimizing therapeutic approaches for calpain-mediated eye diseases can help accelerate clinically feasible strategies for treating calpain dysregulation in other diseased tissues.
Collapse
|
9
|
Boccuni I, Fairless R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life (Basel) 2022; 12:638. [PMID: 35629305 PMCID: PMC9147752 DOI: 10.3390/life12050638] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate neurotransmission and metabolism are finely modulated by the retinal network, where the efficient processing of visual information is shaped by the differential distribution and composition of glutamate receptors and transporters. However, disturbances in glutamate homeostasis can result in glutamate excitotoxicity, a major initiating factor of common neurodegenerative diseases. Within the retina, glutamate excitotoxicity can impair visual transmission by initiating degeneration of neuronal populations, including retinal ganglion cells (RGCs). The vulnerability of RGCs is observed not just as a result of retinal diseases but has also been ascribed to other common neurodegenerative and peripheral diseases. In this review, we describe the vulnerability of RGCs to glutamate excitotoxicity and the contribution of different glutamate receptors and transporters to this. In particular, we focus on the N-methyl-d-aspartate (NMDA) receptor as the major effector of glutamate-induced mechanisms of neurodegeneration, including impairment of calcium homeostasis, changes in gene expression and signalling, and mitochondrial dysfunction, as well as the role of endoplasmic reticular stress. Due to recent developments in the search for modulators of NMDA receptor signalling, novel neuroprotective strategies may be on the horizon.
Collapse
Affiliation(s)
- Isabella Boccuni
- Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Calpain Inhibitors as Potential Therapeutic Modulators in Neurodegenerative Diseases. Neurochem Res 2022; 47:1125-1149. [PMID: 34982393 DOI: 10.1007/s11064-021-03521-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023]
Abstract
It is considered a significant challenge to understand the neuronal cell death mechanisms with a suitable cure for neurodegenerative disorders in the coming years. Calpains are one of the best-considered "cysteine proteases activated" in brain disorders. Calpain is an important marker and mediator in the pathophysiology of neurodegeneration. Calpain activation being the essential neurodegenerative factor causing apoptotic machinery activation, it is crucial to develop reliable and effective approaches to prevent calpain-mediated apoptosis in degenerating neurons. It has been recently seen that the "inhibition of calpain activation" has appeared as a possible therapeutic target for managing neurodegenerative diseases. A systematic literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was conducted. The present article reviews the basic pathobiology and role of selective calpain inhibitors used in various neurodegenerative diseases as a therapeutic target.
Collapse
|
11
|
Kurysheva NI. [Neuroprotective properties of latanoprost]. Vestn Oftalmol 2022; 138:126-134. [PMID: 36004601 DOI: 10.17116/oftalma2022138041126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Glaucoma is the main cause of irreversible blindness in the world. Latanoprost - an ester prodrug of prostaglandin F2α (PGF2α) - was the first prostaglandin analogue used to treat glaucoma. The review shows that latanoprost possesses direct neuroprotective properties such as blocking the entry of calcium ions into neurons and inhibiting the action of caspase-3, inhibiting the activity of cyclooxygenase and activation of polypeptide 2B1 (OATP2B1) and Klotho protein. It is emphasized that when the drug is instilled into the eye, the concentration of the drug inside the vitreous body is twice as high as what is required to ensure the survival of retinal ganglion cells.
Collapse
Affiliation(s)
- N I Kurysheva
- Medical and Biological University of Innovations and Continuing Education of the State Research Center - Burnasyan Federal Biophysical Center, Moscow, Russia
- Ophthalmological Center of the State Research Center - Burnasyan Federal Biophysical Center, Moscow, Russia
| |
Collapse
|
12
|
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
|
13
|
Roscovitine, a Cyclin-Dependent Kinase-5 Inhibitor, Decreases Phosphorylated Tau Formation and Death of Retinal Ganglion Cells of Rats after Optic Nerve Crush. Int J Mol Sci 2021; 22:ijms22158096. [PMID: 34360858 PMCID: PMC8347789 DOI: 10.3390/ijms22158096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by abnormal metabolism of misfolded tau proteins and are progressive. Pathological phosphorylation of tau occurs in the retinal ganglion cells (RGCs) after optic nerve injuries. Cyclin-dependent kinase-5 (Cdk5) causes hyperphosphorylation of tau. To determine the roles played by Cdk5 in retinal degeneration, roscovitine, a Cdk5 inhibitor, was injected intravitreally after optic nerve crush (ONC). The neuroprotective effect of roscovitine was determined by the number of Tuj-1-stained RGCs on day 7. The change in the levels of phosphorylated tau, calpain-1, and cleaved α-fodrin was determined by immunoblots on day 3. The expression of P35/P25, a Cdk5 activator, in the RGCs was determined by immunohistochemistry. The results showed that roscovitine reduced the level of phosphorylated tau by 3.5- to 1.6-fold. Calpain-1 (2.1-fold) and cleaved α-fodrin (1.5-fold) were increased on day 3, suggesting that the calpain signaling pathway was activated. P35/P25 was accumulated in the RGCs that were poorly stained by Tuj-1. Calpain inhibition also reduced the increase in phosphorylated tau. The number of RGCs decreased from 2191 ± 178 (sham) to 1216 ± 122 cells/mm2 on day 7, and roscovitine preserved the level at 1622 ± 130 cells/mm2. We conclude that the calpain-mediated activation of Cdk5 is associated with the pathologic phosphorylation of tau.
Collapse
|
14
|
Masin L, Claes M, Bergmans S, Cools L, Andries L, Davis BM, Moons L, De Groef L. A novel retinal ganglion cell quantification tool based on deep learning. Sci Rep 2021; 11:702. [PMID: 33436866 PMCID: PMC7804414 DOI: 10.1038/s41598-020-80308-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023] Open
Abstract
Glaucoma is a disease associated with the loss of retinal ganglion cells (RGCs), and remains one of the primary causes of blindness worldwide. Major research efforts are presently directed towards the understanding of disease pathogenesis and the development of new therapies, with the help of rodent models as an important preclinical research tool. The ultimate goal is reaching neuroprotection of the RGCs, which requires a tool to reliably quantify RGC survival. Hence, we demonstrate a novel deep learning pipeline that enables fully automated RGC quantification in the entire murine retina. This software, called RGCode (Retinal Ganglion Cell quantification based On DEep learning), provides a user-friendly interface that requires the input of RBPMS-immunostained flatmounts and returns the total RGC count, retinal area and density, together with output images showing the computed counts and isodensity maps. The counting model was trained on RBPMS-stained healthy and glaucomatous retinas, obtained from mice subjected to microbead-induced ocular hypertension and optic nerve crush injury paradigms. RGCode demonstrates excellent performance in RGC quantification as compared to manual counts. Furthermore, we convincingly show that RGCode has potential for wider application, by retraining the model with a minimal set of training data to count FluoroGold-traced RGCs.
Collapse
Affiliation(s)
- Luca Masin
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| | - Marie Claes
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| | - Steven Bergmans
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| | - Lien Cools
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| | - Lien Andries
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| | - Benjamin M. Davis
- grid.83440.3b0000000121901201Glaucoma and Retinal Neurodegenerative Disease Research Group, Institute of Ophthalmology, University College London, London, UK ,grid.496779.2Central Laser Facility, Science and Technologies Facilities Council, UK Research and Innovation, Didcot, Oxfordshire UK
| | - Lieve Moons
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| | - Lies De Groef
- grid.5596.f0000 0001 0668 7884Department of Biology, Neural Circuit Development and Regeneration Research Group, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Li T, Wang Y, Chen J, Gao X, Pan S, Su Y, Zhou X. Co-delivery of brinzolamide and miRNA-124 by biodegradable nanoparticles as a strategy for glaucoma therapy. Drug Deliv 2020; 27:410-421. [PMID: 32133894 PMCID: PMC7067192 DOI: 10.1080/10717544.2020.1731861] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Co-delivery nanoparticles with characteristics of intracellular precision release drug have been generally accepted as an effective therapeutic strategy for eye diseases. In this study, we designed a new co-delivery system (miRNA/NP-BRZ) as a lasting therapeutic approach to prevent the neuro-destructive after the long-term treatment of glaucoma. Neuroprotective and intraocular pressure (IOP) response were assessed in in vivo and in vitro models of glaucoma. At the meaning time, we describe the preparation of miRNA/NP-BRZ, drug release characteristics, intraocular tracing, pharmacokinetic and pharmacodynamics study and toxicity test. We found that miRNA/NP-BRZ could remarkably decrease IOP and significantly prevent retinal ganglion cell (RGC) damages. The new formula of miRNA-124 encapsulated in PEG-PSA-BRZ nanoparticles exhibits high encapsulation efficiency (EE), drug-loading capacity (DC), and stable controlled-release efficacy (EC). Moreover, we also verified that the miRNA/NP-BRZ system is significantly neuroprotective and nontoxic as well as lowering IOP. This study shows our co-delivery drug system would have a wide potential on social and economic benefits for glaucoma.
Collapse
Affiliation(s)
- Tingting Li
- Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, China.,Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ye Wang
- Department of Biological pharmacy, School of Pharmacy, Jilin University, Changchun, China
| | - Jiahao Chen
- Department of Biological pharmacy, School of Pharmacy, Jilin University, Changchun, China
| | - Xiaoshu Gao
- Department of Biological pharmacy, School of Pharmacy, Jilin University, Changchun, China
| | - Siqi Pan
- Department of Biological pharmacy, School of Pharmacy, Jilin University, Changchun, China
| | - Yu Su
- Department of Biological pharmacy, School of Pharmacy, Jilin University, Changchun, China
| | - Xinrong Zhou
- Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, China
| |
Collapse
|
16
|
Asano T, Nagayo Y, Tsuda S, Ito A, Kobayashi W, Fujita K, Sato K, Nishiguchi KM, Kunikata H, Fujioka H, Kamiya M, Urano Y, Nakazawa T. Companion Diagnosis for Retinal Neuroprotective Treatment by Real-Time Imaging of Calpain Activation Using a Novel Fluorescent Probe. Bioconjug Chem 2020; 31:2241-2251. [PMID: 32840357 DOI: 10.1021/acs.bioconjchem.0c00435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Calpain activation induces retinal ganglion cell (RGC) death, while calpain inhibition suppresses RGC death, in animal studies. However, the role of calpain in human retinal disease is unclear. This study investigated a new strategy to study the role of calpain based on real-time imaging. We synthesized a novel fluorescent probe for calpain, acetyl-l-leucyl-l-methionine-hydroxymethyl rhodamine green (Ac-LM-HMRG) and used it for real-time imaging of calpain activation. The toxicity of Ac-LM-HMRG was evaluated with a lactate dehydrogenase cytotoxicity assay, retinal sections, and electroretinograms. Here, we performed real-time imaging of calpain activation in a rat model. First, we administered N-methyl-d-aspartate (NMDA) to induce retinal injury. Twenty minutes later, we administered an intravitreal injection of Ac-LM-HMRG. Real-time imaging was then completed with a noninvasive confocal scanning laser ophthalmoscope. The inhibitory effect of SNJ-1945 against calpain activation was also examined with the same real-time imaging method. Ac-LM-HMRG had no toxic effects. The number of Ac-LM-HMRG-positive cells in real-time imaging significantly increased after NMDA injury, and SNJ-1945 significantly lowered the number of Ac-LM-HMRG-positive cells. Real-time imaging with Ac-LM-HMRG was able to quickly quantify the NMDA-induced activation of calpain and the inhibitory effect of SNJ-1945. This technique, used as a companion diagnostic system, may aid research into the development of new neuroprotective therapies.
Collapse
Affiliation(s)
- Toshifumi Asano
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Yuri Nagayo
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Satoru Tsuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Azusa Ito
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Wataru Kobayashi
- Department of Retinal Disease Control, Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Kosuke Fujita
- Department of Retinal Disease Control, Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.,Collaborative Program of Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi 980-8574, Japan
| | - Hiroshi Kunikata
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.,Department of Retinal Disease Control, Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Hiroyoshi Fujioka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Mako Kamiya
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yasuteru Urano
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.,Department of Retinal Disease Control, Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Collaborative Program of Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi 980-8574, Japan.,Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Miyagi 980-8574, Japan
| |
Collapse
|
17
|
Fujita K, Nishiguchi KM, Sato K, Nakagawa Y, Nakazawa T. In vivo imaging of the light response in mouse retinal ganglion cells based on a neuronal activity-dependent promoter. Biochem Biophys Res Commun 2020; 521:471-477. [PMID: 31672273 DOI: 10.1016/j.bbrc.2019.10.155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 11/19/2022]
Abstract
Diseases of the retinal ganglion cells (RGCs) are an important cause of blindness, yet the light response of individual RGCs is difficult to assess in vivo, particularly in mammals, due to a lack of effective methods. We report a simple in vivo platform for imaging the light response of mouse RGCs based on a fluorescent reporter-tagged enhanced synaptic activity-responsive element (E-SARE) that mediates neuronal activity-dependent gene transcription. When E-SARE-driven d2Venus, packaged into an AAV vector, was injected intravitreally, light-responsive retinal neurons expressing d2Venus were visible at single-cell resolution using confocal ophthalmoscopy. Immunohistological assessment identified the majority of these cells as RGCs. In a murine model of RGC injury, the number of d2Venus-positive cells was correlated with the amplitude of light-induced responses and with visual acuity, measured electrophysiologically at the visual cortex, indicating that the vector can be used as a tool to assess visual function in RGCs. The platform described herein allows a simple in vivo assessment of RGC function, which should help basic research into the mechanisms of RGC death and the development of treatments for diseases involving the RGCs.
Collapse
Affiliation(s)
- Kosuke Fujita
- Department of Ophthalmic Imaging and Information Analytics, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Kota Sato
- Collaborative Program of Ophthalmic Drug Discovery, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Yurika Nakagawa
- Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Toru Nakazawa
- Department of Ophthalmic Imaging and Information Analytics, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Advanced Ophthalmic Medicine, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Collaborative Program of Ophthalmic Drug Discovery, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan.
| |
Collapse
|
18
|
Katayama S, Sato K, Nakazawa T. In vivo and in vitro knockout system labelled using fluorescent protein via microhomology-mediated end joining. Life Sci Alliance 2019; 3:3/1/e201900528. [PMID: 31874862 PMCID: PMC6932181 DOI: 10.26508/lsa.201900528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
Gene knockout is important for understanding gene function and genetic disorders. The CRISPR/Cas9 system has great potential to achieve this purpose. However, we cannot distinguish visually whether a gene is knocked out and in how many cells it is knocked out among a population of cells. Here, we developed a new system that enables the labelling of knockout cells with fluorescent protein through microhomology-mediated end joining-based knock-in. Using a combination with recombinant adeno-associated virus, we delivered our system into the retina, where the expression of Staphylococcus aureus Cas9 was driven by a retina ganglion cell (RGC)-specific promoter, and knocked out carnitine acetyltransferase (CAT). We evaluated RGCs and revealed that CAT is required for RGC survival. Furthermore, we applied our system to Keap1 and confirmed that Keap1 is not expressed in fluorescently labelled cells. Our system provides a promising framework for cell type-specific genome editing and fluorescent labelling of gene knockout based on knock-in.
Collapse
Affiliation(s)
- Shota Katayama
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toru Nakazawa
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan .,Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
19
|
Assessing retinal ganglion cell death and neuroprotective agents using real time imaging. Brain Res 2019; 1714:65-72. [DOI: 10.1016/j.brainres.2019.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 01/01/2023]
|
20
|
Takeuchi H, Inagaki S, Morozumi W, Nakano Y, Inoue Y, Kuse Y, Mizoguchi T, Nakamura S, Funato M, Kaneko H, Hara H, Shimazawa M. VGF nerve growth factor inducible is involved in retinal ganglion cells death induced by optic nerve crush. Sci Rep 2018; 8:16443. [PMID: 30401804 PMCID: PMC6219571 DOI: 10.1038/s41598-018-34585-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022] Open
Abstract
VGF nerve growth factor inducible (VGF) is a polypeptide that is induced by neurotrophic factors and is involved in neurite growth and neuroprotection. The mRNA of the Vgf gene has been detected in the adult rat retina, however the roles played by VGF in the retina are still undetermined. Thus, the purpose of this study was to determine the effects of VGF on the retinal ganglion cells (RGCs) of mice in the optic nerve crush (ONC) model, rat-derived primary cultured RGCs and human induced pluripotent stem cells (iPSCs)-derived RGCs. The mRNA and protein of Vgf were upregulated after the ONC. Immunostaining showed that the VGF was located in glial cells including Müller glia and astrocytes but not in the retinal neurons and their axons. AQEE-30, a VGF peptide, suppressed the loss of RGCs induced by the ONC, and it increased survival rat-derived RGCs and promoted the outgrowth of neurites of rat and human iPSCs derived RGCs in vitro. These findings indicate that VGF plays important roles in neuronal degeneration and has protective effects against the ONC on RGCs. Thus, VGF should be considered as a treatment of RGCs degeneration.
Collapse
Affiliation(s)
- Hiroto Takeuchi
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoshi Inagaki
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan.,Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Wataru Morozumi
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Yukimichi Nakano
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Inoue
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Yoshiki Kuse
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Takahiro Mizoguchi
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Michinori Funato
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Hideo Kaneko
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Hideaki Hara
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
21
|
Mahaman YAR, Huang F, Kessete Afewerky H, Maibouge TMS, Ghose B, Wang X. Involvement of calpain in the neuropathogenesis of Alzheimer's disease. Med Res Rev 2018; 39:608-630. [PMID: 30260518 PMCID: PMC6585958 DOI: 10.1002/med.21534] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/11/2018] [Accepted: 07/29/2018] [Indexed: 01/02/2023]
Abstract
Alzheimer’s disease (AD) is the most common (60% to 80%) age‐related disease associated with dementia and is characterized by a deterioration of behavioral and cognitive capacities leading to death in few years after diagnosis, mainly due to complications from chronic illness. The characteristic hallmarks of the disease are extracellular senile plaques (SPs) and intracellular neurofibrillary tangles (NFTs) with neuropil threads, which are a direct result of amyloid precursor protein (APP) processing to Aβ, and τ hyperphosphorylation. However, many indirect underlying processes play a role in this event. One of these underlying mechanisms leading to these histological hallmarks is the uncontrolled hyperactivation of a family of cysteine proteases called calpains. Under normal physiological condition calpains participate in many processes of cells’ life and their activation is tightly controlled. However, with an increase in age, increased oxidative stress and other excitotoxicity assaults, this regulatory system becomes impaired and result in increased activation of these proteases involving them in the pathogenesis of various diseases including neurodegeneration like AD. Reviewed here is a pool of data on the implication of calpains in the pathogenesis of AD, the underlying molecular mechanism, and the potential of targeting these enzymes for AD therapeutics.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Henok Kessete Afewerky
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tanko Mahamane Salissou Maibouge
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bishwajit Ghose
- Department of Social Medicine and Health Management, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Division of Neurodegenerative Disorders, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
22
|
Sato K, Saigusa D, Saito R, Fujioka A, Nakagawa Y, Nishiguchi KM, Kokubun T, Motoike IN, Maruyama K, Omodaka K, Shiga Y, Uruno A, Koshiba S, Yamamoto M, Nakazawa T. Metabolomic changes in the mouse retina after optic nerve injury. Sci Rep 2018; 8:11930. [PMID: 30093719 PMCID: PMC6085332 DOI: 10.1038/s41598-018-30464-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
In glaucoma, although axonal injury drives retinal ganglion cell (RGC) death, little is known about the underlying pathomechanisms. To provide new mechanistic insights and identify new biomarkers, we combined latest non-targeting metabolomics analyses to profile altered metabolites in the mouse whole retina 2, 4, and 7 days after optic nerve crush (NC). Ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry and liquid chromatography Fourier transform mass spectrometry covering wide spectrum of metabolites in combination highlighted 30 metabolites that changed its concentration after NC. The analysis displayed similar changes for purine nucleotide and glutathione as reported previously in another animal model of axonal injury and detected multiple metabolites that increased after the injury. After studying the specificity of the identified metabolites to RGCs in histological sections using imaging mass spectrometry, two metabolites, i.e., L-acetylcarnitine and phosphatidylcholine were increased not only preceding the peak of RGC death in the whole retina but also at the RGC layer (2.3-fold and 1.2-fold, respectively). These phospholipids propose novel mechanisms of RGC death and may serve as early biomarkers of axonal injury. The combinatory metabolomics analyses promise to illuminate pathomechanisms, reveal biomarkers, and allow the discovery of new therapeutic targets of glaucoma.
Collapse
Affiliation(s)
- Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.,Department of Ophthalmic imaging and information analytics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan.,LEAP, Japan Agency for Medical Research and Development (AMED), Chiyoda, Tokyo, Japan
| | - Ritsumi Saito
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Amane Fujioka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yurika Nakagawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Taiki Kokubun
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ikuko N Motoike
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Department of Systems Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Kazuichi Maruyama
- Department of Innovative Visual Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.,Department of Ophthalmic imaging and information analytics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Seizo Koshiba
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan. .,Department of Ophthalmic imaging and information analytics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan. .,Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan. .,Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| |
Collapse
|
23
|
Sato K, Shiga Y, Nakagawa Y, Fujita K, Nishiguchi KM, Tawarayama H, Murayama N, Maekawa S, Yabana T, Omodaka K, Katayama S, Feng Q, Tsuda S, Nakazawa T. Ecel1 Knockdown With an AAV2-Mediated CRISPR/Cas9 System Promotes Optic Nerve Damage-Induced RGC Death in the Mouse Retina. Invest Ophthalmol Vis Sci 2018; 59:3943-3951. [PMID: 30073365 DOI: 10.1167/iovs.18-23784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To assess the therapeutic potential of endothelin-converting enzyme-like 1 (Ecel1) in a mouse model of optic nerve crush. Methods Ecel1 expression was evaluated with real time quantitative (qRT)-PCR, Western blotting, and immunohistochemistry in mouse retinas after optic nerve crush. Vinblastine administration to the optic nerve and the intravitreal injection of N-methyl-d-aspartate (NMDA) were used to assess Ecel1 gene expression. Ecel1 was deleted with an adeno-associated viral (AAV) clustered regulatory interspaced short palindromic repeat (CRISPR)/Cas9 system, and retinal ganglion cell (RGC) survival was investigated with retrograde labeling, qRT-PCR, and visual evoked potential. Results Optic nerve crush induced Ecel1 expression specifically in the RGCs, peaking on day 4 after optic nerve crush. Ecel1 gene expression was induced by the vinblastine-induced inhibition of axonal flow, but not by NMDA-induced excitotoxicity, even though both are triggers of RGC death. Knockdown of Ecel1 promoted the loss of RGCs after optic nerve crush. Conclusions Our data suggest that Ecel1 induction is part of the retinal neuroprotective response to axonal injury in mice. These findings might provide insight into novel therapeutic targets for the attenuation of RGC damage, such as occurs in traumatic optic neuropathy.
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
| | - Yukihiro Shiga
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yurika Nakagawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kosuke Fujita
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hiroshi Tawarayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Namie Murayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shigeto Maekawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Takeshi Yabana
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shota Katayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Qiwei Feng
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Satoru Tsuda
- 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
| |
Collapse
|
24
|
Liu XF, Zhou DD, Xie T, Hao JL, Malik TH, Lu CB, Qi J, Pant OP, Lu CW. The Nrf2 Signaling in Retinal Ganglion Cells under Oxidative Stress in Ocular Neurodegenerative Diseases. Int J Biol Sci 2018; 14:1090-1098. [PMID: 29989056 PMCID: PMC6036726 DOI: 10.7150/ijbs.25996] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/22/2018] [Indexed: 12/28/2022] Open
Abstract
Retinal ganglion cells (RGCs) are one of the important cell types affected in many ocular neurodegenerative diseases. Oxidative stress is considered to be involved in retinal RGCs death in ocular neurodegenerative diseases. More and more attention has been focused on studying the agents that may have neuroprotective effects. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key nuclear transcription factor for the systemic antioxidant defense system. This review elucidates the underlying mechanism of the Nrf2-mediated neuroprotective effects on RGCs in ocular neurodegenerative diseases, such as diabetic retinopathy and retinal ischemia-reperfusion injury. Several Nrf2 inducers that shield RGCs from oxidative stress-induced neurodegeneration via regulating Nrf2 signaling are discussed.
Collapse
Affiliation(s)
- Xiu-Fen Liu
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Dan-Dan Zhou
- Department of Radiology, The First Hospital of Jilin University, Jilin, China
| | - Tian Xie
- Department of . Neurosurgery, The People's Hospital of Jilin Province, Jilin, China
| | - Ji-Long Hao
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Tayyab Hamid Malik
- Department of Gastroenterology, The First Hospital of Jilin University, Jilin, China
| | - Cheng-Bo Lu
- Department of Cardiology, The First Hospital of Jiamusi University, Heilongjiang, China
| | - Jing Qi
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Om Prakash Pant
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Cheng-Wei Lu
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
25
|
Evangelho K, Mogilevskaya M, Losada-Barragan M, Vargas-Sanchez JK. Pathophysiology of primary open-angle glaucoma from a neuroinflammatory and neurotoxicity perspective: a review of the literature. Int Ophthalmol 2017; 39:259-271. [PMID: 29290065 DOI: 10.1007/s10792-017-0795-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/11/2017] [Indexed: 11/29/2022]
Abstract
PURPOSE Glaucoma is the leading cause of blindness in humans, affecting 2% of the population. This disorder can be classified into various types including primary, secondary, glaucoma with angle closure and with open angle. The prevalence of distinct types of glaucoma differs for each particular region of the world. One of the most common types of this disease is primary open-angle glaucoma (POAG), which is a complex inherited disorder characterized by progressive retinal ganglion cell death, optic nerve head excavation and visual field loss. Nowadays, POAG is considered an optic neuropathy, while intraocular pressure is proposed to play a fundamental role in its pathophysiology and especially in optic disk damage. However, the exact mechanism of optic nerve head damage remains a topic of debate. This literature review aims to bring together the information on the pathophysiology of primary open-angle glaucoma, particularly focusing on neuroinflammatory mechanisms leading to the death of the retinal ganglion cell. METHODS A literature search was done on PubMed using key words including primary open-angle glaucoma, retinal ganglion cells, Müller cells, glutamate, glial cells, ischemia, hypoxia, exitotoxicity, neuroinflammation, axotomy and neurotrophic factors. The literature was reviewed to collect the information published about the pathophysiologic mechanisms of RGC death in the POAG, from a neuroinflammatory and neurotoxicity perspective. RESULTS Proposed mechanisms for glaucomatous damage are a result of pressure in RGC followed by ischemia, hypoxia of the ONH, and consequently death due to glutamate-induced excitotoxicity, deprivation of energy and oxygen, increase in levels of inflammatory mediators and alteration of trophic factors flow. These events lead to blockage of anterograde and retrograde axonal transport with ensuing axotomy and eventually blindness. CONCLUSIONS The damage to ganglion cells and eventually glaucomatous injury can occur via various mechanisms including baric trauma, ischemia and impact of metabolic toxins, which triggers an inflammatory process and secondary degeneration in the ONH.
Collapse
Affiliation(s)
- Karine Evangelho
- Grupo de Investigación en Ciencias Biomédicas GRINCIBIO, Facultad de medicina, Sede Bogotá, Universidad Antonio Nariño, Bogotá, Colombia
| | - Maria Mogilevskaya
- Grupo de Investigación en Ingeniería Clínica - Hospital Universitario la Samaritana GINIC-HUS, Sede Bogotá, ECCI, Bogotá, Colombia
| | - Monica Losada-Barragan
- Grupo de Biología Celular y Funcional e Ingeniería de Biomoléculas, Facultad de Ciencias, Sede Bogotá, Universidad Antonio Nariño, Bogotá, Colombia
| | - Jeinny Karina Vargas-Sanchez
- Grupo de Investigación en Ciencias Biomédicas GRINCIBIO, Facultad de medicina, Sede Bogotá, Universidad Antonio Nariño, Bogotá, Colombia.
| |
Collapse
|
26
|
Schaefer K, Mahajan M, Gore A, Tsang SH, Bassuk AG, Mahajan VB. Calpain-5 gene expression in the mouse eye and brain. BMC Res Notes 2017; 10:602. [PMID: 29157313 PMCID: PMC5697233 DOI: 10.1186/s13104-017-2927-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/10/2017] [Indexed: 01/10/2023] Open
Abstract
Objective Our objective was to characterize CAPN5 gene expression in the mouse central nervous system. Mouse brain and eye sections were probed with two high-affinity RNA oligonucleotide analogs designed to bind CAPN5 RNA and one scramble, control oligonucleotide. Images were captured in brightfield. Results CAPN5 RNA probes were validated on mouse breast cancer tumor tissue. In the eye, CAPN5 was expressed in the ganglion cell, inner nuclear and outer nuclear layers of the retina. Signal could not be detected in the ciliary body or the iris because of the high density of melanin. In the brain, CAPN5 was expressed in the granule cell layers of the hippocampus and cerebellum. There was scattered expression in pons. The visual cortex showed faint signal. Most signal in the brain was in a punctate pattern.
Collapse
Affiliation(s)
- Kellie Schaefer
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - MaryAnn Mahajan
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Anuradha Gore
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Stephen H Tsang
- Bernard and Shirlee Brown Glaucoma Laboratory, Department of Pathology and Cell Biology, Department of Ophthalmology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | - Vinit B Mahajan
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA. .,Palo Alto Veterans Administration, Palo Alto, CA, USA.
| |
Collapse
|
27
|
Stankowska DL, Mueller BH, Oku H, Ikeda T, Dibas A. Neuroprotective effects of inhibitors of Acid-Sensing ion channels (ASICs) in optic nerve crush model in rodents. Curr Eye Res 2017; 43:84-95. [PMID: 29111855 DOI: 10.1080/02713683.2017.1383442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE The purpose of the current study was to assess the potential involvement of acid-sensing ion channel 1 (ASIC1) in retinal ganglion cell (RGC) death and investigate the neuroprotective effects of inhibitors of ASICs in promoting RGC survival following optic nerve crush (ONC). RESULTS ASIC1 protein was significantly increased in optic nerve extracts at day 7 following ONC in rats. Activated calpain-1 increased at 2 and 7 days following ONC as evidenced by increased degradation of α-fodrin, known substrate of calpain. Glial fibrillary acidic protein levels increased significantly at 2 and 7 days post-injury. By contrast, glutamine synthetase increased at 2 days while decreased at 7 days. The inhibition of ASICs with amiloride and psalmotoxin-1 significantly increased RGC survival in rats following ONC (p < 0.05, one-way ANOVA). The mean number of surviving RGCs in rats (n = 6) treated with amiloride (100 µM) following ONC was 1477 ± 98 cells/mm2 compared with ONC (1126 ± 101 cells/mm2), where psalmotoxin-1 (1 μM) treated rats (n = 6) and subjected to ONC had 1441 ± 63 RGCs/mm2 compared with ONC (1065 ± 76 RGCs/mm2). Average number of RGCs in control rats (n = 12) was 2092 ± 46 cells/mm2. Blocking of ASICs also significantly increased RGC survival from ischemic-like insult from 473 ± 80 to 842 ± 49 RGCs/mm2 (for psalmotoxin-1) and from 628 ± 53 RGCs/mm2 to 890 ± 55 RGCs/mm2 (for amiloride) with p ≤ 0.05, using one-way ANOVA. Acidification (a known activator of ASIC1) increased intracellular Ca2+ ([Ca2+]i) in rat primary RGCs, which was statistically blocked by pretreatment with 100 nM psalmotoxin-1. CONCLUSIONS ASIC1 up-regulation-induced influx of extracellular calcium may be responsible for activation of calcium-sensitive calpain-1 in the retina. Calpain-1 induced degradation of α-fodrin and leads to morphological changes and eventually neuronal death. Therefore, blockers of ASIC1 can be used as potential therapeutics in the treatment of optic nerve degeneration. ABBREVIATIONS 4-(2-Aminoethyl) benzenesulfonyl fluoride hydrochloride (AEBSF); acid-sensing ion channels (ASICs); analysis of variance (ANOVA); bicinchoninic acid (BCA); brain-derived neurotrophic factor (BDNF); central nervous system (CNS); ciliary neurotrophic factor (CNTF); dimethyl sulfoxide (DMSO); endoplasmic reticulum (ER); ethylene glycol-bis(β-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA); ethylenediaminetetraacetic acid (EDTA); Food and Drug Administration (FDA); glial fibrillary acidic protein (GFAP); glutamine synthetase (GS); intraocular pressure (IOP); kilodalton (kDa); Krebs-Ringer Buffer (KRB); optic nerve crush (ONC); phosphate-buffered saline (PBS); plasma membrane (PM); polymerase chain reaction (PCR); retinal ganglion cell (RGC); RNA Binding Protein With Multiple Splicing (RBPMS); room temperature (RT); standard error of the mean (SEM).
Collapse
Affiliation(s)
- Dorota L Stankowska
- a North Texas Eye Research Institute , University of North Texas Health Science Center , Fort Worth
| | - Brett H Mueller
- a North Texas Eye Research Institute , University of North Texas Health Science Center , Fort Worth
| | - Hidehiro Oku
- b Department of Ophthalmology , Osaka Medical College , Takatsuki Osaka , Japan
| | - Tsunehiko Ikeda
- b Department of Ophthalmology , Osaka Medical College , Takatsuki Osaka , Japan
| | - Adnan Dibas
- a North Texas Eye Research Institute , University of North Texas Health Science Center , Fort Worth
| |
Collapse
|
28
|
Smith AW, Ray SK, Das A, Nozaki K, Rohrer B, Banik NL. Calpain inhibition as a possible new therapeutic target in multiple sclerosis. AIMS MOLECULAR SCIENCE 2017; 4:446-462. [PMID: 40181912 PMCID: PMC11967729 DOI: 10.3934/molsci.2017.4.446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Multiple sclerosis (MS), the most common chronic autoimmune inflammatory disease of the central nervous system (CNS), is characterized by demyelination and neurodegeneration. In particular, neurodegeneration is a major factor in disease progression with neuronal death and irreversible axonal damage leading to disability. MS is manageable with current therapies that are directed towards immunomodulation but there are no available therapies for neuroprotection. The complex pathophysiology and heterogeneity of MS indicate that therapeutic agents should be directed to both the inflammatory and neurodegenerative arms of the disease. Activity of the Ca2+ activated protease calpain has been previously implicated in progression of MS and its primary animal model, experimental autoimmune encephalomyelitis (EAE). The effects of calpain inhibitors in EAE involve downregulation of Th1/Th17 inflammatory responses and promotion of regulatory T cells, overall leading to decreased inflammatory cell infiltration in CNS tissues. Furthermore, analysis of brains, spinal cords and optic nerves from EAE animals revealed decreases in axon degeneration, motor neuron and retinal ganglion cell death. This resulted in improved severity of paralysis and preservation of visual function. Taken together, the studies presented in this brief review suggest that use of calpain inhibitors in combination with an immunomodulatory agent may be a potential therapeutic strategy for MS and optic neuritis.
Collapse
Affiliation(s)
- Amena W. Smith
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, SC, USA
| | - Arabinda Das
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Kenkichi Nozaki
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Baerbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Naren L. Banik
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
29
|
Nakamura O, Moritoh S, Sato K, Maekawa S, Murayama N, Himori N, Omodaka K, Sogon T, Nakazawa T. Bilberry extract administration prevents retinal ganglion cell death in mice via the regulation of chaperone molecules under conditions of endoplasmic reticulum stress. Clin Ophthalmol 2017; 11:1825-1834. [PMID: 29066860 PMCID: PMC5644593 DOI: 10.2147/opth.s145159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose To investigate the effect of bilberry extract anthocyanins on retinal ganglion cell (RGC) survival after optic nerve crush. Additionally, to determine details of the mechanism of the neuroprotective effect of bilberry extract anthocyanins and the involvement of endoplasmic reticulum stress suppression in the mouse retina. Materials and methods Anthocyanins in bilberry extract (100 mg/kg/day or 500 mg/kg/day) were administrated orally to C57BL/6J mice. The expression levels of various molecular chaperones were assessed with quantitative reverse-transcription polymerase chain reaction, Western blotting, and immunohistochemistry. RGC survival was evaluated by measuring the gene expression of RGC markers and counting retrogradely labeled RGCs after optic nerve crush. Results The protein levels of Grp78 and Grp94 increased significantly in mice after bilberry extract administration. Increased Grp78 and Grp94 levels were detected in the inner nuclear layer and ganglion cell layer of the retina, surrounding the RGCs. Gene expression of Chop, Bax, and Atf4 increased in mice after optic nerve crush and decreased significantly after oral bilberry extract administration. RGC survival after nerve crush also increased with bilberry extract administration. Conclusion These results indicate that oral bilberry extract administration suppresses RGC death. Bilberry extract administration increased Grp78 and Grp94 protein levels, an effect which may underlie the neuroprotective effect of bilberry extract after optic nerve crush. Thus, bilberry extract has a potential role in neuroprotective treatments for retinal injuries, such as those which occur in glaucoma.
Collapse
Affiliation(s)
- Orie Nakamura
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Satoru Moritoh
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - 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
| | - Shigeto Maekawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Namie Murayama
- 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.,Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tetsuya Sogon
- R&D Department, Wakasa Seikatsu Co., Ltd., Kyoto, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Department of Retinal Disease Control, 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 Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
30
|
Maekawa S, Sato K, Fujita K, Daigaku R, Tawarayama H, Murayama N, Moritoh S, Yabana T, Shiga Y, Omodaka K, Maruyama K, Nishiguchi KM, Nakazawa T. The neuroprotective effect of hesperidin in NMDA-induced retinal injury acts by suppressing oxidative stress and excessive calpain activation. Sci Rep 2017; 7:6885. [PMID: 28761134 PMCID: PMC5537259 DOI: 10.1038/s41598-017-06969-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/22/2017] [Indexed: 12/21/2022] Open
Abstract
We found that hesperidin, a plant-derived bioflavonoid, may be a candidate agent for neuroprotective treatment in the retina, after screening 41 materials for anti-oxidative properties in a primary retinal cell culture under oxidative stress. We found that the intravitreal injection of hesperidin in mice prevented reductions in markers of the retinal ganglion cells (RGCs) and RGC death after N-methyl-D-aspartate (NMDA)-induced excitotoxicity. Hesperidin treatment also reduced calpain activation, reactive oxygen species generation and TNF-α gene expression. Finally, hesperidin treatment improved electrophysiological function, measured with visual evoked potential, and visual function, measured with optomotry. Thus, we found that hesperidin suppressed a number of cytotoxic factors associated with NMDA-induced cell death signaling, such as oxidative stress, over-activation of calpain, and inflammation, thereby protecting the RGCs in mice. Therefore, hesperidin may have potential as a therapeutic supplement for protecting the retina against the damage associated with excitotoxic injury, such as occurs in glaucoma and diabetic retinopathy.
Collapse
Affiliation(s)
- Shigeto Maekawa
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kota Sato
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kosuke Fujita
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Reiko Daigaku
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Hiroshi Tawarayama
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Namie Murayama
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Satoru Moritoh
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Takeshi Yabana
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Toru Nakazawa
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
| |
Collapse
|
31
|
Schaefer KA, Toral MA, Velez G, Cox AJ, Baker SA, Borcherding NC, Colgan DF, Bondada V, Mashburn CB, Yu CG, Geddes JW, Tsang SH, Bassuk AG, Mahajan VB. Calpain-5 Expression in the Retina Localizes to Photoreceptor Synapses. Invest Ophthalmol Vis Sci 2017; 57:2509-21. [PMID: 27152965 PMCID: PMC4868102 DOI: 10.1167/iovs.15-18680] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose We characterize calpain-5 (CAPN5) expression in retinal and neuronal subcellular compartments. Methods CAPN5 gene variants were classified using the exome variant server, and RNA-sequencing was used to compare expression of CAPN5 mRNA in the mouse and human retina and in retinoblastoma cells. Expression of CAPN5 protein was ascertained in humans and mice in silico, in mouse retina by immunohistochemistry, and in neuronal cancer cell lines and fractionated central nervous system tissue extracts by Western analysis with eight antibodies targeting different CAPN5 regions. Results Most CAPN5 genetic variation occurs outside its protease core; and searches of cancer and epilepsy/autism genetic databases found no variants similar to hyperactivating retinal disease alleles. The mouse retina expressed one transcript for CAPN5 plus those of nine other calpains, similar to the human retina. In Y79 retinoblastoma cells, the level of CAPN5 transcript was very low. Immunohistochemistry detected CAPN5 expression in the inner and outer nuclear layers and at synapses in the outer plexiform layer. Western analysis of fractionated retinal extracts confirmed CAPN5 synapse localization. Western blots of fractionated brain neuronal extracts revealed distinct subcellular patterns and the potential presence of autoproteolytic CAPN5 domains. Conclusions CAPN5 is moderately expressed in the retina and, despite higher expression in other tissues, hyperactive disease mutants of CAPN5 only manifest as eye disease. At the cellular level, CAPN5 is expressed in several different functional compartments. CAPN5 localization at the photoreceptor synapse and with mitochondria explains the neural circuitry phenotype in human CAPN5 disease alleles.
Collapse
Affiliation(s)
- Kellie A Schaefer
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States 2Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Marcus A Toral
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States 2Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States 3Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, United States
| | - Gabriel Velez
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States 2Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States 3Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, United States
| | - Allison J Cox
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States
| | - Sheila A Baker
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States 5Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States
| | - Nicholas C Borcherding
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States 3Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, United States
| | - Diana F Colgan
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States 2Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Vimala Bondada
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Charles B Mashburn
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Chen-Guang Yu
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - James W Geddes
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Stephen H Tsang
- Barbara & Donald Jonas Stem Cell Laboratory, and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Pathology & Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States 9Neurology, University of Iowa, Iowa City, Iowa, United States
| | - Vinit B Mahajan
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States 2Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
32
|
Fujita K, Nishiguchi KM, Shiga Y, Nakazawa T. Spatially and Temporally Regulated NRF2 Gene Therapy Using Mcp-1 Promoter in Retinal Ganglion Cell Injury. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:130-141. [PMID: 28480312 PMCID: PMC5415330 DOI: 10.1016/j.omtm.2017.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/12/2017] [Indexed: 02/03/2023]
Abstract
Retinal ganglion cell degeneration triggered by axonal injury is believed to underlie many ocular diseases, including glaucoma and optic neuritis. In these diseases, retinal ganglion cells are affected unevenly, both spatially and temporally, such that healthy and unhealthy cells coexist in different patterns at different time points. Herein, we describe a temporally and spatially regulated adeno-associated virus gene therapy aiming to reduce undesired off-target effects on healthy retinal neurons. The Mcp-1 promoter previously shown to be activated in stressed retinal ganglion cells following murine optic nerve injury was combined with the neuroprotective intracellular transcription factor Nrf2. In this model, Mcp-1 promoter-driven NRF2 expression targeting only stressed retinal ganglion cells showed efficacy equivalent to non-selective cytomegalovirus promoter-driven therapy for preventing cell death. However, cytomegalovirus promoter-mediated NRF2 transcription induced cellular stress responses and death of Brn3A-positive uninjured retinal ganglion cells. Such undesired effects were reduced substantially by adopting the Mcp-1 promoter. Combining a stress-responsive promoter and intracellular therapeutic gene is a versatile approach for specifically targeting cells at risk of degeneration. This strategy may be applicable to numerous chronic ocular and non-ocular conditions.
Collapse
Affiliation(s)
- Kosuke Fujita
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Toru Nakazawa
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| |
Collapse
|
33
|
Sun D, Moore S, Jakobs TC. Optic nerve astrocyte reactivity protects function in experimental glaucoma and other nerve injuries. J Exp Med 2017; 214:1411-1430. [PMID: 28416649 PMCID: PMC5413323 DOI: 10.1084/jem.20160412] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 12/05/2016] [Accepted: 03/08/2017] [Indexed: 01/06/2023] Open
Abstract
Reactive remodeling of optic nerve head astrocytes is consistently observed in glaucoma and other optic nerve injuries. However, it is unknown whether this reactivity is beneficial or harmful for visual function. In this study, we used the Cre recombinase (Cre)-loxP system under regulation of the mouse glial fibrillary acidic protein promoter to knock out the transcription factor signal transducer and activator of transcription 3 (STAT3) from astrocytes and test the effect this has on reactive remodeling, ganglion cell survival, and visual function after experimental glaucoma and nerve crush. After injury, STAT3 knockout mice displayed attenuated astrocyte hypertrophy and reactive remodeling; astrocytes largely maintained their honeycomb organization and glial tubes. These changes were associated with increased loss of ganglion cells and visual function over a 30-day period. Thus, reactive astrocytes play a protective role, preserving visual function. STAT3 signaling is an important mediator of various aspects of the reactive phenotype within optic nerve astrocytes.
Collapse
Affiliation(s)
- Daniel Sun
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - Sara Moore
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
34
|
Ikuta Y, Aoyagi S, Tanaka Y, Sato K, Inada S, Koseki Y, Onodera T, Oikawa H, Kasai H. Creation of nano eye-drops and effective drug delivery to the interior of the eye. Sci Rep 2017; 7:44229. [PMID: 28290486 PMCID: PMC5349510 DOI: 10.1038/srep44229] [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: 08/16/2016] [Accepted: 02/06/2017] [Indexed: 01/22/2023] Open
Abstract
Nano eye-drops are a new type of ophthalmic treatment with increased potency and reduced side effects. Compounds in conventional eye-drops barely penetrate into the eye because the cornea, located at the surface of eye, has a strong barrier function for preventing invasion of hydrophilic or large-sized materials from the outside. In this work, we describe the utility of nano eye-drops utilising brinzolamide, a commercially available glaucoma treatment drug, as a target compound. Fabrication of the nanoparticles of brinzolamide prodrug increases the eye penetration rate and results in high drug efficacy, compared with that of commercially available brinzolamide eye-drops formulated as micro-sized structures. In addition, the resulting nano eye-drops were not toxic to the corneal epithelium after repeated administration for 1 week. The nano eye-drops may have applications as a next-generation ophthalmic treatment.
Collapse
Affiliation(s)
- Yoshikazu Ikuta
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Shigenobu Aoyagi
- Ouchi Shinko Chemical Industrial Co., Ltd., Research and Development Center, 111 Shimojyukumae, Sukagawa, Fukushima 962-0806, Japan
| | - Yuji Tanaka
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kota Sato
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Satoshi Inada
- Ouchi Shinko Chemical Industrial Co., Ltd., Research and Development Center, 111 Shimojyukumae, Sukagawa, Fukushima 962-0806, Japan
| | - Yoshitaka Koseki
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Tsunenobu Onodera
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Hidetoshi Oikawa
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Hitoshi Kasai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| |
Collapse
|
35
|
Yamamoto K, Sato K, Yukita M, Yasuda M, Omodaka K, Ryu M, Fujita K, Nishiguchi KM, Machida S, Nakazawa T. The neuroprotective effect of latanoprost acts via klotho-mediated suppression of calpain activation after optic nerve transection. J Neurochem 2017; 140:495-508. [PMID: 27859240 PMCID: PMC5299490 DOI: 10.1111/jnc.13902] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022]
Abstract
Latanoprost was first developed for use in glaucoma therapy as an ocular hypotensive agent targeting the prostaglandin F2α (FP) receptor. Subsequently, latanoprost showed a neuroprotective effect, an additional pharmacological action. However, although it is well-known that latanoprost exerts an ocular hypotensive effect via the FP receptor, it is not known whether this is also true of its neuroprotective effect. Klotho was firstly identified as the gene linked to the suppression of aging phenotype: the defect of klotho gene in mice results aging phenotype such as hypokinesis, arteriosclerosis, and short lifespan. After that, the function of klotho was also reported to maintain calcium homeostasis and to exert a neuroprotective effect in various models of neurodegenerative disease. However, the function of klotho in eyes including retina is still poorly understood. Here, we show that klotho is a key factor underlying the neuroprotective effect of latanoprost during post-axotomy retinal ganglion cell (RGC) degeneration. Importantly, a quantitative RT-PCR gene expression analysis of klotho in sorted rat retinal cells revealed that the highest expression level of klotho in the retina was in the RGCs. Latanoprost acid, the biologically active form of latanoprost, inhibits post-traumatic calpain activation and concomitantly facilitates the expression and shedding of klotho in axotomized RGCs. This expression profile is a good match with the localization, not of the FP receptor, but of organic anion transporting polypeptide 2B1, known as a prostaglandin transporter, in the ocular tissue. Furthermore, an organic anion transporting polypeptide 2B1 inhibitor suppressed latanoprost acid-mediated klotho shedding ex vivo, whereas an FP receptor antagonist did not. The klotho fragments shed from the RGCs reduced the intracellular level of reactive oxygen species, and a specific klotho inhibitor accelerated and increased RGC death after axotomy. We conclude that the shed klotho fragments might contribute to the attenuation of axonal injury-induced calpain activation and oxidative stress, thereby protecting RGCs from post-traumatic neuronal degeneration.
Collapse
Affiliation(s)
- Kotaro Yamamoto
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
| | - Kota Sato
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
- Department of Ophthalmic Imaging and Information AnalyticsTohoku University Graduate School of MedicineMiyagiJapan
| | - Masayoshi Yukita
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
| | - Masayuki Yasuda
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
| | - Kazuko Omodaka
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
- Department of Ophthalmic Imaging and Information AnalyticsTohoku University Graduate School of MedicineMiyagiJapan
| | - Morin Ryu
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
| | - Kosuke Fujita
- Department of Retinal Disease ControlTohoku University Graduate School of MedicineMiyagiJapan
| | - Koji M. Nishiguchi
- Department of Advanced Ophthalmic MedicineTohoku University Graduate School of MedicineMiyagiJapan
| | - Shigeki Machida
- Department of OphthalmologyDokkyo Medical University Koshigaya HospitalSaitamaJapan
- Department of OphthalmologyIwate Medical University School of MedicineIwateJapan
| | - Toru Nakazawa
- Department of OphthalmologyTohoku University Graduate School of MedicineMiyagiJapan
- Department of Ophthalmic Imaging and Information AnalyticsTohoku University Graduate School of MedicineMiyagiJapan
- Department of Retinal Disease ControlTohoku University Graduate School of MedicineMiyagiJapan
- Department of Advanced Ophthalmic MedicineTohoku University Graduate School of MedicineMiyagiJapan
| |
Collapse
|
36
|
Involvement of P2X7 receptor in neuronal degeneration triggered by traumatic injury. Sci Rep 2016; 6:38499. [PMID: 27929040 PMCID: PMC5144087 DOI: 10.1038/srep38499] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022] Open
Abstract
Axonal injury is a common feature of central nervous system insults that culminates with the death of the affected neurons, and an irreversible loss of function. Inflammation is an important component of the neurodegenerative process, where the microglia plays an important role by releasing proinflammatory factors as well as clearing the death neurons by phagocytosis. Here we have identified the purinergic signaling through the P2X7 receptor as an important component for the neuronal death in a model of optic nerve axotomy. We have found that in P2X7 receptor deficient mice there is a delayed loss of retinal ganglion cells and a decrease of phagocytic microglia at early times points after axotomy. In contralateral to the axotomy retinas, P2X7 receptor controlled the numbers of phagocytic microglia, suggesting that extracellular ATP could act as a danger signal activating the P2X7 receptor in mediating the loss of neurons in contralateral retinas. Finally, we show that intravitreal administration of the selective P2X7 receptor antagonist A438079 also delays axotomy-induced retinal ganglion cell death in retinas from wild type mice. Thus, our work demonstrates that P2X7 receptor signaling is involved in neuronal cell death after axonal injury, being P2X7 receptor antagonism a potential therapeutic strategy.
Collapse
|
37
|
Ko JA, Minamoto A, Sugimoto Y, Kiuchi Y. Down-regulation of semaphorin 3F in rat retinal ganglion cells in response to optic nerve crush. Cell Biochem Funct 2016; 34:378-84. [DOI: 10.1002/cbf.3200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/26/2016] [Accepted: 05/26/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Ji-Ae Ko
- Department of Ophthalmology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima City Hiroshima Japan
| | - Akira Minamoto
- Department of Ophthalmology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima City Hiroshima Japan
| | - Yosuke Sugimoto
- Department of Ophthalmology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima City Hiroshima Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima City Hiroshima Japan
| |
Collapse
|
38
|
Geeraerts E, Dekeyster E, Gaublomme D, Salinas-Navarro M, De Groef L, Moons L. A freely available semi-automated method for quantifying retinal ganglion cells in entire retinal flatmounts. Exp Eye Res 2016; 147:105-113. [PMID: 27107795 DOI: 10.1016/j.exer.2016.04.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/08/2016] [Accepted: 04/18/2016] [Indexed: 01/13/2023]
Abstract
Glaucomatous optic neuropathies are characterized by progressive loss of retinal ganglion cells (RGCs), the neurons that connect the eye to the brain. Quantification of these RGCs is a cornerstone in experimental optic neuropathy research and commonly performed via manually quantifying parts of the retina. However, this is a time-consuming process subject to inter- and intra-observer variability. Here we present a freely available ImageJ script to semi-automatically quantify RGCs in entire retinal flatmounts after immunostaining for the RGC-specific transcription factor Brn3a. The blob-like signal of Brn3a-immunopositive RGCs is enhanced via eigenvalues of the Hessian matrix and the resulting local maxima are counted as RGCs. After the user has outlined the retinal flatmount area, the total RGC number and retinal area are reported and an isodensity map, showing the RGC density distribution across the retina, is created. The semi-automated quantification shows a very strong correlation (Pearson's r ≥ 0.99) with manual counts for both widefield and confocal images, thereby validating the data generated via the developed script. Moreover, application of this method in established glaucomatous optic neuropathy models such as N-methyl-D-aspartate-induced excitotoxicity, optic nerve crush and laser-induced ocular hypertension revealed RGC loss conform with literature. Compared to manual counting, the described automated quantification method is faster and shows user-independent consistency. Furthermore, as the script detects the RGC number in entire retinal flatmounts, the method allows detection of regional differences in RGC density. As such, it can help advance research investigating the degenerative mechanisms of glaucomatous optic neuropathies and the effectiveness of new neuroprotective treatments. Because the script is flexible and easy to optimize due to a low number of critical parameters, it can potentially be applied in combination with other tissues or alternative labeling protocols.
Collapse
Affiliation(s)
- E Geeraerts
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - E Dekeyster
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - D Gaublomme
- Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Centre, Ghent University, Ghent, Belgium; Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - M Salinas-Navarro
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - L De Groef
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - L Moons
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
39
|
Tsuda S, Tanaka Y, Kunikata H, Yokoyama Y, Yasuda M, Ito A, Nakazawa T. Real-time imaging of RGC death with a cell-impermeable nucleic acid dyeing compound after optic nerve crush in a murine model. Exp Eye Res 2016; 146:179-188. [PMID: 27013099 DOI: 10.1016/j.exer.2016.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/16/2015] [Accepted: 03/17/2016] [Indexed: 11/16/2022]
Abstract
The retinal ganglion cells (RGCs) are the main source of therapeutic targets for neuroprotective glaucoma treatment, and evaluating RGCs is key for effective glaucoma care. Thus, we developed a minimally invasive, quick, real-time method to evaluate RGC death in mice. In this article we describe the details of our method, report new results obtained from C57BL/6J mice, and report that our method was usable in wild type (WT) and knockout (KO) mice lacking an RGC-death-suppressing gene. It used a non-invasive confocal scanning laser ophthalmoscope (cSLO) and a low molecular weight, photo-switching, cell-impermeant, fluorescent nucleic acid dyeing compound, SYTOX orange (SO). The RGCs were retrogradely labeled with Fluorogold (FG), the optic nerve was crushed (ONC), and SO was injected into the vitreous. After ten minutes, RGC death was visualized with cSLO in vivo. The retinas were then extracted and flat mounted for histological observation. SO-labeled RGCs were counted in vivo and FG-labeled RGCs were counted in retinal flat mounts. The time course of RGC death was examined in Calpastatin KO mice and wild type (WT) mice. Our in vivo imaging method revealed that SO-positive dead RGCs were mainly present from 4 to 6 days after ONC, and the peak of RGC death was after 5 days. Moreover, the number of SO-positive dead RGCs after 5 days differed significantly in the Calpastatin KO mice and the WT mice. Counting FG-labeled RGCs in isolated retinas confirmed these results. Thus, real-time imaging with SO was able to quickly quantify ONC-induced RGC death. This technique may aid research into RGC death and the development of new neuroprotective therapies for glaucoma.
Collapse
Affiliation(s)
- Satoru Tsuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Japan
| | - Yuji Tanaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Japan
| | - Hiroshi Kunikata
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Japan; Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yu Yokoyama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Japan
| | - Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Japan
| | - Azusa Ito
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 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.
| |
Collapse
|
40
|
In vivo cellular imaging of various stress/response pathways using AAV following axonal injury in mice. Sci Rep 2015; 5:18141. [PMID: 26670005 PMCID: PMC4680972 DOI: 10.1038/srep18141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/10/2015] [Indexed: 11/30/2022] Open
Abstract
Glaucoma, a leading cause of blindness worldwide, is instigated by various factors, including axonal injury, which eventually leads to a progressive loss of retinal ganglion cells (RGCs). To study various pathways reportedly involved in the pathogenesis of RGC death caused by axonal injury, seven pathways were investigated. Pathway-specific fluorescent protein-coded reporters were each packaged into an adeno-associated virus (AAV). After producing axonal injury in the eye, injected with AAV to induce RGC death, the temporal activity of each stress-related pathway was monitored in vivo through the detection of fluorescent RGCs using confocal ophthalmoscopy. We identified the activation of ATF6 and MCP-1 pathways involved in endoplasmic reticulum stress and macrophage recruitment, respectively, as early markers of RGC stress that precede neuronal death. Conversely, inflammatory responses probed by NF-κB and cell-death-related pathway p53 were most prominent in the later phases, when RGC death was already ongoing. AAV-mediated delivery of stress/response reporters followed by in vivo cellular imaging is a powerful strategy to characterize the temporal aspects of complex molecular pathways involved in retinal diseases. The identification of promoter elements that are activated before the death of RGCs enables the development of pre-emptive gene therapy, exclusively targeting the early phases of diseased cells.
Collapse
|
41
|
Yukita M, Machida S, Nishiguchi KM, Tsuda S, Yokoyama Y, Yasuda M, Maruyama K, Nakazawa T. Molecular, anatomical and functional changes in the retinal ganglion cells after optic nerve crush in mice. Doc Ophthalmol 2015; 130:149-56. [PMID: 25560383 DOI: 10.1007/s10633-014-9478-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/30/2014] [Indexed: 01/11/2023]
Abstract
PURPOSE Optic nerve crush (ONC) and subsequent axonal damage can be used in rodents to study the mechanism of retinal ganglion cell (RGC) degeneration. Here, we examined electroretinograms (ERGs) in post-ONC mice to investigate changes in the positive scotopic threshold response (pSTR). We then compared these changes with molecular and morphological changes to identify early objective biomarkers of RGC dysfunction. METHODS Fifty 12-week-old C57BL/6 mice were included. ONC was used to induce axonal injury in the right eye of each animal, with the left eye used as a control. The expression of the RGC markers Brn3a and Brn3b was measured on days 1, 2, 3, 5 and 7 after ONC with quantitative real-time PCR. ERGs were recorded under dark adaptation with the stimulus intensity increasing from -6.2 to 0.43 log cd-s/m(2) on days 1, 2, 3, 5, 7 and 10 after ONC. The pSTR, a- and b-wave amplitudes were measured. Inner retinal thickness around the optic nerve head was measured with spectral-domain optical coherence tomography on days 0, 2, 5, 7 and 10 after ONC. RESULTS The expression of Brn3a and Brn3b began to significantly decrease on day 1 and day 2, respectively (P < 0.01). The amplitude of the pSTR underwent rapid, significant deterioration on day 3, after which it fell gradually (P < 0.01), while the a- and b-wave amplitudes remained unchanged throughout the experiment. Inner retinal thickness gradually decreased, with the most significant reduction on day 10 (P < 0.01). CONCLUSIONS Decrease in pSTR likely reflected the early loss of RGC function after ONC and that declining expression of RGC-specific genes preceded anatomical and functional changes in the RGCs.
Collapse
Affiliation(s)
- Masayoshi Yukita
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aoba, Sendai, Miyagi, 980-8574, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Yasuda M, Tanaka Y, Nishiguchi KM, Ryu M, Tsuda S, Maruyama K, Nakazawa T. Retinal transcriptome profiling at transcription start sites: a cap analysis of gene expression early after axonal injury. BMC Genomics 2014; 15:982. [PMID: 25407019 PMCID: PMC4246558 DOI: 10.1186/1471-2164-15-982] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/23/2014] [Indexed: 12/01/2022] Open
Abstract
Background Glaucoma is characterized by progressive loss of the visual field and death of retinal ganglion cells (RGCs), a process that is mediated, in part, by axonal injury. However, the molecular pathomechanisms linking RGC death and axonal injury remain largely unknown. Here, we examined these mechanisms with a cap analysis of gene expression (CAGE), which allows the comprehensive quantification of transcription initiation across the entire genome. We aimed to identify changes in gene expression patterns and to predict the resulting alterations in the protein network in the early phases of axonal injury in mice. Results We performed optic nerve crush (ONC) in mice to model axonal injury. Two days after ONC, the retinas were isolated, RNA was extracted, and a CAGE library was constructed and sequenced. CAGE data for ONC eyes and sham-treated eyes was compared, revealing 180 differentially expressed genes. Among them, the Bcat1 gene, involved in the catabolism of branched-chain amino acid transaminase, showed the largest change in expression (log2 fold-change = 6.70). In some differentially expressed genes, alternative transcription start sites were observed in the ONC eyes, highlighting the dynamism of transcription initiation in a state of disease. In silico pathway analysis predicted that ATF4 was the most significant upstream regulator orchestrating pathological processes after ONC. Its downstream candidate targets included Ddit3, which is known to induce cell death under endoplasmic reticulum stress. In addition, a regulatory network comprising IFNG, P38 MAPK, and TP53 was predicted to be involved in the induction of cell death. Conclusion Through CAGE, we have identified differentially expressed genes that may account for the link between axonal injury and RGC death. Furthermore, an in silico pathway analysis provided a global view of alterations in the networks of key regulators of biological pathways that presumably take place in ONC. We thus believe that our study serves as a valuable resource to understand the molecular processes that define axonal injury-driven RGC death. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-982) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
| |
Collapse
|
43
|
Neuroprotective effect against axonal damage-induced retinal ganglion cell death in apolipoprotein E-deficient mice through the suppression of kainate receptor signaling. Brain Res 2014; 1586:203-12. [PMID: 25160129 DOI: 10.1016/j.brainres.2014.08.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 11/22/2022]
Abstract
Apolipoprotein E (ApoE) plays important roles in the body, including a carrier of cholesterols, an anti-oxidant, and a ligand for the low-density lipoprotein receptors. In the nervous system, the presence of ApoE4 isoforms is associated with Alzheimer's disease. ApoE gene polymorphisms are also associated with glaucoma, but the function of ApoE in the retina remains unclear. In this study, we investigated the role of ApoE in axonal damage-induced RGC death. ApoE was detected in the astrocytes and Müller cells in the wild-type (WT) retina. RGC damage was induced in adult ApoE-deficient mice (male, 10-12 weeks old) through ocular hypertension (OH), optic nerve crush (NC), or by administering kainic acid (KA) intravitreally. The WT mice were treated with a glutamate receptor antagonist (MK801 or CNQX) 30 min before performing NC or left untreated. Seven days later, the retinas were flat mounted and Fluorogold-labeled RGCs were counted. We found that the RGCs in the ApoE-deficient mice were resistant to OH-induced RGC death and optic nerve degeneration 4 weeks after induction. In WT mice, NC effectively induced RGC death (control: 4085±331 cells/mm(2), NC: 1728±170 cells/mm(2)). CNQX, an inhibitor of KA receptors, suppressed this RGC death (3031±246 cells/mm(2)), but MK801, an inhibitor of NMDA receptors, did not (1769±212 cells/mm(2)). This indicated the involvement of KA receptor signaling in NC-induced RGC death. We found that NC- or KA-induced RGC death was significantly less in the ApoE-deficient mice than in the WT mice. These data suggest that the ApoE deficiency had a neuroprotective effect against axonal damage-induced RGC death by suppressing the KA receptor signaling.
Collapse
|
44
|
Yokoyama Y, Maruyama K, Yamamoto K, Omodaka K, Yasuda M, Himori N, Ryu M, Nishiguchi KM, Nakazawa T. The role of calpain in an in vivo model of oxidative stress-induced retinal ganglion cell damage. Biochem Biophys Res Commun 2014; 451:510-5. [PMID: 25111816 DOI: 10.1016/j.bbrc.2014.08.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/01/2014] [Indexed: 12/17/2022]
Abstract
PURPOSE In this study, we set out to establish an in vivo animal model of oxidative stress in the retinal ganglion cells (RGCs) and determine whether there is a link between oxidative stress in the RGCs and the activation of calpain, a major part of the apoptotic pathway. MATERIALS AND METHODS Oxidative stress was induced in the RGCs of C57BL/6 mice by the intravitreal administration of 2,2'-azobis (2-amidinopropane) dihydrochloride (AAPH, 30mM, 2μl). Control eyes were injected with 2μl of vehicle. Surviving Fluorogold (FG)-labeled RGCs were then counted in retinal flat mounts. Double staining with CellROX and Annexin V was performed to investigate the co-localization of free radical generation and apoptosis. An immunoblot assay was used both to indirectly evaluate calpain activation in the AAPH-treated eyes by confirming α-fodrin cleavage, and also to evaluate the effect of SNJ-1945 (a specific calpain inhibitor: 4% w/v, 100mg/kg, intraperitoneal administration) in these eyes. RESULTS Intravitreal administration of AAPH led to a significant decrease in FG-labeled RGCs 7days after treatment (control: 3806.7±575.2RGCs/mm(2), AAPH: 3156.1±371.2RGCs/mm(2), P<0.01). CellROX and Annexin V signals were co-localized in the FG-labeled RGCs 24h after AAPH injection. An immunoblot assay revealed a cleaved α-fodrin band that increased significantly 24h after AAPH administration. Intraperitoneally administered SNJ-1945 prevented the cleavage of α-fodrin and had a neuroprotective effect against AAPH-induced RGC death (AAPH: 3354.0±226.9RGCs/mm(2), AAPH+SNJ-1945: 3717.1±614.6RGCs/mm(2), P<0.01). CONCLUSION AAPH administration was an effective model of oxidative stress in the RGCs, showing that oxidative stress directly activated the calpain pathway and induced RGC death. Furthermore, inhibition of the calpain pathway protected the RGCs after AAPH administration.
Collapse
Affiliation(s)
- Yu Yokoyama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kotaro Yamamoto
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kazuko Omodaka
- 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
| | - Morin Ryu
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Koji M Nishiguchi
- 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 Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan.
| |
Collapse
|
45
|
Himori N, Maruyama K, Yamamoto K, Yasuda M, Ryu M, Omodaka K, Shiga Y, Tanaka Y, Nakazawa T. Critical neuroprotective roles of heme oxygenase-1 induction against axonal injury-induced retinal ganglion cell death. J Neurosci Res 2014; 92:1134-42. [PMID: 24799032 DOI: 10.1002/jnr.23398] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/10/2014] [Accepted: 04/01/2014] [Indexed: 12/26/2022]
Abstract
Although axonal damage induces significant retinal ganglion cell (RGC) death, small numbers of RGCs are able to survive up to 7 days after optic nerve crush (NC) injury. To develop new treatments, we set out to identify patterns of change in the gene expression of axonal damage-resistant RGCs. To compensate for the low density of RGCs in the retina, we performed retrograde labeling of these cells with 4Di-10ASP in adult mice and 7 days after NC purified the RGCs with fluorescence-activated cell sorting. Gene expression in the cells was determined with a microarray, and the expression of Ho-1 was determined with quantitative PCR (qPCR). Changes in protein expression were assessed with immunohistochemistry and immunoblotting. Additionally, the density of Fluoro-gold-labeled RGCs was counted in retinas from mice pretreated with CoPP, a potent HO-1 inducer. The microarray and qPCR analyses showed increased expression of Ho-1 in the post-NC RGCs. Immunohistochemistry also showed that HO-1-positive cells were present in the ganglion cell layer (GCL), and cell counting showed that the proportion of HO-1-positive cells in the GCL rose significantly after NC. Seven days after NC, the number of RGCs in the CoPP-treated mice was significantly higher than in the control mice. Combined pretreatment with SnPP, an HO-1 inhibitor, suppressed the neuroprotective effect of CoPP. These results reflect changes in HO-1 activity to RGCs that are a key part of RGC survival. Upregulation of HO-1 signaling may therefore be a novel therapeutic strategy for glaucoma.
Collapse
Affiliation(s)
- Noriko Himori
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yasuda M, Tanaka Y, Ryu M, Tsuda S, Nakazawa T. RNA sequence reveals mouse retinal transcriptome changes early after axonal injury. PLoS One 2014; 9:e93258. [PMID: 24676137 PMCID: PMC3968129 DOI: 10.1371/journal.pone.0093258] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 03/04/2014] [Indexed: 12/23/2022] Open
Abstract
Glaucoma is an ocular disease characterized by progressive retinal ganglion cell (RGC) death caused by axonal injury. However, the underlying mechanisms involved in RGC death remain unclear. In this study, we investigated changes in the transcriptome profile following axonal injury in mice (C57BL/6) with RNA sequencing (RNA-seq) technology. The experiment group underwent an optic nerve crush (ONC) procedure to induce axonal injury in the right eye, and the control group underwent a sham procedure. Two days later, we extracted the retinas and performed RNA-seq and a pathway analysis. We identified 177 differentially expressed genes with RNA-seq, notably the endoplasmic reticulum (ER) stress-related genes Atf3, Atf4, Atf5, Chac1, Chop, Egr1 and Trb3, which were significantly upregulated. The pathway analysis revealed that ATF4 was the most significant upstream regulator. The antioxidative response-related genes Hmox1 and Srxn1, as well as the immune response-related genes C1qa, C1qb and C1qc, were also significantly upregulated. To our knowledge, this is the first reported RNA-seq investigation of the retinal transcriptome and molecular pathways in the early stages after axonal injury. Our results indicated that ER stress plays a key role under these conditions. Furthermore, the antioxidative defense and immune responses occurred concurrently in the early stages after axonal injury. We believe that our study will lead to a better understanding of and insight into the molecular mechanisms underlying RGC death after axonal injury.
Collapse
Affiliation(s)
- Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Tanaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Morin Ryu
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Tsuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| |
Collapse
|
47
|
Suzuki R, Oka T, Tamada Y, Shearer TR, Azuma M. Degeneration and dysfunction of retinal neurons in acute ocular hypertensive rats: involvement of calpains. J Ocul Pharmacol Ther 2014; 30:419-28. [PMID: 24660785 DOI: 10.1089/jop.2013.0100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Retinal ischemic diseases primarily lead to damage of the inner retinal neurons. Electrophysiological studies also suggest impairment of the inner retinal neurons. Our recent studies with acute ocular hypertensive rats confirmed damage predominantly in the inner retinal layer along with the ganglion cell layer, changes that are ameliorated by the calpain inhibitor SNJ-1945. However, we do not know which specific neuronal cells in the inner retinal layer are damaged by calpains. Thus, the purpose of the present study was to identify specific calpain-damaged neuronal cells in the inner retina from acute ocular hypertensive rats. METHODS Intraocular pressure was elevated to 110 mm Hg for 40 min. One hour after ocular hypertension (OH), SNJ-1945 was administrated as a single oral dose of 50 mg/kg. Retinal function was assessed by scotopic electroretinography (ERG). Histological degeneration was evaluated by hematoxylin and eosin, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end-labeling (TUNEL), and immunostaining in thin sections and flat mounts of the retina. Calpain activation was determined by proteolysis of the calpain substrate α-spectrin. RESULTS OH caused calpain activation, increased TUNEL-positive staining, decreased thickness of the inner nuclear layer (INL), and decreased amplitudes of the ERG a- and b-waves and oscillatory potentials (OPs). SNJ-1945 significantly inhibited calpain activation and the decrease in ERG values. Interestingly, the changes in the b-wave and OPs amplitudes were significantly correlated to changes in the thickness of the INL. In the inner retinal layer, the numbers of rod bipolar, cone-ON bipolar, and amacrine cells were decreased after OH. SNJ-1945 suppressed the loss of cone-ON bipolar and amacrine cells, but did not inhibit the loss of rod bipolar cells. We also observed increased glial fibrillary acid protein-positive staining in the Müller cells after OH and the treatment with SNJ-1945. CONCLUSIONS Calpains may contribute to ischemic retinal dysfunction by causing the loss of cone-ON bipolar and amacrine cells and causing the activation of Müller cells. Calpain inhibitor SNJ-1945 may be a candidate compound for treatment of retinal ischemic disease.
Collapse
Affiliation(s)
- Rie Suzuki
- 1 Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Co., Ltd. , Kobe, Japan
| | | | | | | | | |
Collapse
|
48
|
Trager N, Smith A, Wallace Iv G, Azuma M, Inoue J, Beeson C, Haque A, Banik NL. Effects of a novel orally administered calpain inhibitor SNJ-1945 on immunomodulation and neurodegeneration in a murine model of multiple sclerosis. J Neurochem 2014; 130:268-79. [PMID: 24447070 DOI: 10.1111/jnc.12659] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/27/2013] [Accepted: 01/15/2014] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) pathology is marked by the massive infiltration of myelin-specific T cells into the CNS. Hallmarks of T helper (Th) cells during active disease are pro-inflammatory Th1/Th17 cells that predominate over immunoregulatory Th2/Treg cells. Neurodegeneration, a major factor in progressive MS, is often overlooked when considering drug prescription. Here, we show that oral dosing with SNJ-1945, a novel water-soluble calpain inhibitor, reduces experimental autoimmune encephalomyelitis clinical scores in vivo and has a two pronged effect via anti-inflammation and protection against neurodegeneration. We also show that SNJ-1945 treatment down-regulates Th1/Th17 inflammatory responses, and promotes regulatory T cells (Tregs) and myeloid-derived suppressor cells in vivo, which are known to have the capacity to suppress helper as well as cytotoxic T cell functions. Through analysis of spinal cord samples, we show a reduction in calpain expression, decreased infiltration of inflammatory cells, and signs of inhibition of neurodegeneration. We also show a marked reduction in neuronal cell death in spinal cord (SC) sections. These results suggest that calpain inhibition attenuates experimental autoimmune encephalomyelitis pathology by reducing both inflammation and neurodegeneration, and could be used in clinical settings to augment the efficacy of standard immunomodulatory agents used to treat MS. Multiple sclerosis (MS) pathology is marked by inflammation and infiltration of myelin-specific T cells into the central nervous system. Inflammation leads to neurodegeneration in progressive MS which also leads to epitope spreading, feedback looping to more inflammation. Calpain can play a role in both arms of the disease. Here, oral dosing with SNJ-1945, a novel water-soluble calpain inhibitor, reduces experimental autoimmune encephalomyelitis clinical scores in vivo and has a two-pronged effect via anti-inflammation and protection against neurodegeneration.
Collapse
Affiliation(s)
- Nicole Trager
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, USA; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Omodaka K, Murata T, Sato S, Takahashi M, Tatewaki Y, Nagasaka T, Doi H, Araie M, Takahashi S, Nakazawa T. Correlation of magnetic resonance imaging optic nerve parameters to optical coherence tomography and the visual field in glaucoma. Clin Exp Ophthalmol 2013; 42:360-8. [PMID: 24119065 DOI: 10.1111/ceo.12237] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 08/25/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND To determine the viability of using magnetic resonance imaging measurement of optic nerve morphology as an objective analysis of glaucomatous damage. DESIGN Retrospective study conducted at Tohoku University Hospital. PARTICIPANTS Thirty-eight eyes of 19 patients with open-angle glaucoma. METHODS Patients were scanned with T2-weighted and 3-T diffusion tensor magnetic resonance imaging, and parameters of the optic nerve, including fractional anisotropy, apparent diffusion coefficient and cross-sectional area, were determined. Conventional parameters of glaucomatous damage, including circumpapillary and macular retinal nerve fibre layer thickness, and mean deviation and average total deviation of the central 16 test points from the Humphrey Field Analyzer, were then compared with the magnetic resonance imaging-derived parameters. Spearman's coefficient of correlation was calculated to determine the significance of the correlation. MAIN OUTCOME MEASURE Correlation coefficient between the magnetic resonance imaging parameters and the parameters of glaucomatous damage. RESULTS Mean deviation was significantly correlated with all magnetic resonance imaging parameters (fractional anisotropy: r = 0.53, apparent diffusion coefficient: r = -0.44, cross-sectional area: r = 0.70). Circumpapillary retinal nerve fibre layer thickness was significantly correlated with fractional anisotropy (r = 0.60) and cross-sectional area (r = 0.47), but not apparent diffusion coefficient (r = -0.29). Central macular function and macular retinal nerve fibre layer thickness were also significantly correlated with magnetic resonance imaging parameters. CONCLUSIONS Optic nerve magnetic resonance imaging parameters were significantly correlated to glaucomatous damage. Magnetic resonance imaging analysis of the optic nerve may, thus, have value as an objective instrument to assess glaucomatous degeneration, including the function of the macula.
Collapse
Affiliation(s)
- Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Tohoku, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Li X, Kang Q, Gao S, Wei T, Liu Y, Chen X, Lv H. Transplantation with retinal progenitor cells repairs visual function in rats with retinal ischemia-reperfusion injury. Neurosci Lett 2013; 558:8-13. [PMID: 24076256 DOI: 10.1016/j.neulet.2013.09.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/20/2013] [Accepted: 09/07/2013] [Indexed: 01/01/2023]
Abstract
The retinal ischemia-reperfusion injury (RIR) is a common pathological process that leads to progressive visual loss and blindness in many retinal diseases such as retinal vascular occlusion disease, diabetic retinopathy, and acute glaucoma. Currently, there has been no effective therapy. The purpose of this study was to investigate the effects of transplantation of retinal progenitor cells (RPCs) into the subretinal space (SRS) and the superior colliculus (SC) in a rat model of RIR injury. We used cultured postnatal day 1 rat RPCs transfected with adeno-associated virus containing the cDNA encoding enhanced green fluorescence protein (EGFP) for transplantation. RIR injury was induced by increases in the intraocular pressure to 110 mmHg for 60 min. The effects of transplantation were evaluated by immunohistochemistry, electroretinography (ERG), and visual evoked potentials (VEP). We found that in rats with RIR injury, RPCs transplanted into the SRS and the SC survived for at least 8 weeks, migrated into surrounding tissues, and improved the ERG and VEP responses. Cells transplanted into the SC improved the VEP response more than those transplanted into the SRS. Our data suggest that transplantation of RPCs into the SRS and the SC may be a possible method for cell replacement therapy for retinal diseases.
Collapse
Affiliation(s)
- Xueying Li
- Department of Ophthalmology, The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China.
| | - Qianyan Kang
- Department of Ophthalmology, The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China.
| | - Shan Gao
- Department of Ophthalmology, The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Ting Wei
- Department of Ophthalmology, The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| |
Collapse
|