1
|
Schmetterer L, Tezel G, Schuman J. Neville Osborne - Editor-in-Chief of Progress in Retinal and Eye Research for 40 years. Prog Retin Eye Res 2023; 96:101194. [PMID: 37473797 DOI: 10.1016/j.preteyeres.2023.101194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore; Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria; Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland.
| | - Gülgün Tezel
- Department of Ophthalmology, Vagelos College of Physicians and Surgeons, Columbia University, New York, USA
| | - Joel Schuman
- Wills Eye Hospital, Department of Ophthalmology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
2
|
Stavropoulos D, Grewal MK, Petriti B, Chau KY, Hammond CJ, Garway-Heath DF, Lascaratos G. The Role of Mitophagy in Glaucomatous Neurodegeneration. Cells 2023; 12:1969. [PMID: 37566048 PMCID: PMC10417839 DOI: 10.3390/cells12151969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
This review aims to provide a better understanding of the emerging role of mitophagy in glaucomatous neurodegeneration, which is the primary cause of irreversible blindness worldwide. Increasing evidence from genetic and other experimental studies suggests that mitophagy-related genes are implicated in the pathogenesis of glaucoma in various populations. The association between polymorphisms in these genes and increased risk of glaucoma is presented. Reduction in intraocular pressure (IOP) is currently the only modifiable risk factor for glaucoma, while clinical trials highlight the inadequacy of IOP-lowering therapeutic approaches to prevent sight loss in many glaucoma patients. Mitochondrial dysfunction is thought to increase the susceptibility of retinal ganglion cells (RGCs) to other risk factors and is implicated in glaucomatous degeneration. Mitophagy holds a vital role in mitochondrial quality control processes, and the current review explores the mitophagy-related pathways which may be linked to glaucoma and their therapeutic potential.
Collapse
Affiliation(s)
- Dimitrios Stavropoulos
- Department of Ophthalmology, King’s College Hospital, London SE5 9RS, UK;
- Department of Ophthalmology, 417 Veterans Army Hospital (NIMTS), 11521 Athens, Greece
| | - Manjot K. Grewal
- NIHR Biomedical Research Center, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London EC1V 9EL, UK
- Division of Optometry and Visual Science, School of Health Sciences, City, University of London, London EC1V 0HB, UK
| | - Bledi Petriti
- NIHR Biomedical Research Center, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London EC1V 9EL, UK
- Department of Clinical & Movement Neurosciences, UCL Queens Square Institute of Neurology, London NW3 2PF, UK
| | - Kai-Yin Chau
- Department of Clinical & Movement Neurosciences, UCL Queens Square Institute of Neurology, London NW3 2PF, UK
| | - Christopher J. Hammond
- Section of Ophthalmology, School of Life Course Sciences, King’s College London, London SE1 7EH, UK
- Department of Ophthalmology, St Thomas’ Hospital, London SE1 7EH, UK
| | - David F. Garway-Heath
- NIHR Biomedical Research Center, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Gerassimos Lascaratos
- Department of Ophthalmology, King’s College Hospital, London SE5 9RS, UK;
- Section of Ophthalmology, School of Life Course Sciences, King’s College London, London SE1 7EH, UK
| |
Collapse
|
3
|
Ziółkowska N, Lewczuk B, Szyryńska N, Rawicka A, Vyniarska A. Low-Intensity Blue Light Exposure Reduces Melanopsin Expression in Intrinsically Photosensitive Retinal Ganglion Cells and Damages Mitochondria in Retinal Ganglion Cells in Wistar Rats. Cells 2023; 12:cells12071014. [PMID: 37048087 PMCID: PMC10093228 DOI: 10.3390/cells12071014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/18/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
This study investigated the effect of low-intensity blue light on the albino Wistar rat retina, including intrinsically photosensitive retinal ganglion cells (ipRGCs). Three groups of nine albino Wistar rats were used. One group was continuously exposed to blue light (150 lx) for 2 d (STE); one was exposed to 12 h of blue light and 12 h of darkness for 10 d (LTE); one was maintained in 12 h of white light (150 lx) and 12 h of darkness for 10 d (control). Melanopsin (Opn4) was immunolabelled on retinal whole-mounts. To count and measure Opn4-positive ipRGC somas and dendrites (including Sholl profiles), Neuron J was used. Retinal cryosections were immunolabeled for glial fibrillary acid protein (GFAP) and with terminal deoxynucleotidyl transferase dUTP nick-end labelling for apoptosis detection. LTE reduced the length of Opn4-positive ipRGC dendrites (p = 0.03) and decreased Opn4-immunoreactivity in ipRGC outer stratifying dendrites. LTE and STE decreased the complexity of dendritic arborization (Sholl profile; p < 0.001, p = 0.03, respectively), increased retinal GFAP immunoreactivity (p < 0.001, p = 0.002, respectively), and caused outer segment vesiculation and outer nuclear layer apoptosis. Ultrastructural analysis showed that LTE damaged mitochondria in retinal ganglion cells and in the inner plexiform layer. Thus, LTE to low-intensity blue light harms the retinas of albino Wistar rats.
Collapse
|
4
|
Tapia ML, Nascimento-Dos-Santos G, Park KK. Subtype-specific survival and regeneration of retinal ganglion cells in response to injury. Front Cell Dev Biol 2022; 10:956279. [PMID: 36035999 PMCID: PMC9411869 DOI: 10.3389/fcell.2022.956279] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a heterogeneous population of neurons that function synchronously to convey visual information through the optic nerve to retinorecipient target areas in the brain. Injury or disease to the optic nerve results in RGC degeneration and loss of visual function, as few RGCs survive, and even fewer can be provoked to regenerate their axons. Despite causative insults being broadly shared, regeneration studies demonstrate that RGC types exhibit differential resilience to injury and undergo selective survival and regeneration of their axons. While most early studies have identified these RGC types based their morphological and physiological characteristics, recent advances in transgenic and gene sequencing technologies have further enabled type identification based on unique molecular features. In this review, we provide an overview of the well characterized RGC types and identify those shown to preferentially survive and regenerate in various regeneration models. Furthermore, we discuss cellular characteristics of both the resilient and susceptible RGC types including the combinatorial expression of different molecular markers that identify these specific populations. Lastly, we discuss potential molecular mechanisms and genes found to be selectively expressed by specific types that may contribute to their reparative capacity. Together, we describe the studies that lay the important groundwork for identifying factors that promote neural regeneration and help advance the development of targeted therapy for the treatment of RGC degeneration as well as neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Mary L Tapia
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gabriel Nascimento-Dos-Santos
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
5
|
Ziólkowska N, Chmielewska-Krzesinska M, Vyniarska A, Sienkiewicz W. Exposure to Blue Light Reduces Melanopsin Expression in Intrinsically Photoreceptive Retinal Ganglion Cells and Damages the Inner Retina in Rats. Invest Ophthalmol Vis Sci 2022; 63:26. [PMID: 35060997 PMCID: PMC8787613 DOI: 10.1167/iovs.63.1.26] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose The purpose of this study was to investigative the effects of blue light on intrinsically photoreceptive retinal ganglion cells (ipRGCs). Methods Brown Norway rats were used. Nine rats were continuously exposed to blue light (light emitting diodes [LEDs]: 463 nm; 1000 lx) for 2 days (acute exposure [AE]); 9 rats were exposed to 12 hours of blue light and 12 hours of darkness for 10 days (long-term exposure [LTE]); 6 control rats were exposed to 12 hours of white fluorescent light (1000 lx) and 12 hours of darkness for 10 days. Whole-mount retinas were immunolabelled with melanopsin antibodies; melanopsin-positive (MP) ipRGC somas and processes were counted and measured with Neuron J. To detect apoptosis, retinal cryo-sections were stained with terminal deoxynucleotidyl transferase dUTP nick-end labeling. Ultra-thin sections were visualized with transmission electron microscopy. Results The number of MP ipRGC somas was significantly lower in retinas from AE and LTE rats than in those from control rats (P < 0.001 and = 0.002, respectively). The mean length of MP areas of processes was significantly lower in AE rats (P < 0.001). AE rats had severe retinal damage and massive apoptosis in the outer nuclear layer; their mitochondria were damaged in the axons and dendrites of the nerve fiber layer and the inner plexiform layer. Retinal ganglion cells (RGCs) in AE rats appeared to have reduced amounts of free ribosomes and rough endoplasmic reticulum. Conclusions AE to blue light reduces melanopsin expression and damages RGCs, likely including ipRGCs. Changes in the axons and dendrites of RGCs suggest possible disruption of intraretinal and extraretinal signal transmission.
Collapse
Affiliation(s)
- Natalia Ziólkowska
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Malgorzata Chmielewska-Krzesinska
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Alla Vyniarska
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Stepan Gzhytskyi National University of Veterinary and Biotechnologies, Lviv, Ukraine
| | - Waldemar Sienkiewicz
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
6
|
Kang EYC, Liu PK, Wen YT, Quinn PMJ, Levi SR, Wang NK, Tsai RK. Role of Oxidative Stress in Ocular Diseases Associated with Retinal Ganglion Cells Degeneration. Antioxidants (Basel) 2021; 10:1948. [PMID: 34943051 PMCID: PMC8750806 DOI: 10.3390/antiox10121948] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Ocular diseases associated with retinal ganglion cell (RGC) degeneration is the most common neurodegenerative disorder that causes irreversible blindness worldwide. It is characterized by visual field defects and progressive optic nerve atrophy. The underlying pathophysiology and mechanisms of RGC degeneration in several ocular diseases remain largely unknown. RGCs are a population of central nervous system neurons, with their soma located in the retina and long axons that extend through the optic nerve to form distal terminals and connections in the brain. Because of this unique cytoarchitecture and highly compartmentalized energy demand, RGCs are highly mitochondrial-dependent for adenosine triphosphate (ATP) production. Recently, oxidative stress and mitochondrial dysfunction have been found to be the principal mechanisms in RGC degeneration as well as in other neurodegenerative disorders. Here, we review the role of oxidative stress in several ocular diseases associated with RGC degenerations, including glaucoma, hereditary optic atrophy, inflammatory optic neuritis, ischemic optic neuropathy, traumatic optic neuropathy, and drug toxicity. We also review experimental approaches using cell and animal models for research on the underlying mechanisms of RGC degeneration. Lastly, we discuss the application of antioxidants as a potential future therapy for the ocular diseases associated with RGC degenerations.
Collapse
Affiliation(s)
- Eugene Yu-Chuan Kang
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Pei-Kang Liu
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 80424, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80424, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97403, Taiwan;
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; (P.M.J.Q.); (S.R.L.)
| | - Sarah R. Levi
- Jonas Children’s Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; (P.M.J.Q.); (S.R.L.)
| | - Nan-Kai Wang
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97403, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 97403, Taiwan
| |
Collapse
|
7
|
Córdova C, Lozano C, Rodríguez B, Marchant I, Zúñiga R, Ochova P, Olivero P, González-Arriagada WA. Optogenetic control of cancer cell survival in ChR2-transfected HeLa cells. Int J Exp Pathol 2021; 102:242-248. [PMID: 34791724 DOI: 10.1111/iep.12426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2021] [Accepted: 10/28/2021] [Indexed: 11/30/2022] Open
Abstract
Optogenetics is a molecular biological technique involving transfection of cells with photosensitive proteins and the subsequent study of their biological effects. The aim of this study was to evaluate the effect of blue light on the survival of HeLa cells, transfected with channelrhodopsin-2 (ChR2). HeLa wild-type cells were transfected with a plasmid that contained the gene for ChR2. Transfection and channel function were evaluated by real-time polymerase chain reaction (RT-PCR), fluorescence imaging using green fluorescent protein (GFP) and flow cytometry for intracellular calcium changes using a Fura Red probe. We developed a platform for optogenetic stimulation for use within the cell culture incubator. Different stimulation procedures using blue light (467 nm) were applied for up to 24 h. Cell survival was determined by flow cytometry using propidium iodide and rhodamine probes. Change in cell survival showed a statistically significant (p < 0.05) inverse association with the frequency and time of application of the light stimulus. This change seemed to be associated with the ChR2 cis-trans-isomerization cycle. Cell death was associated with high concentrations of calcium in the cytoplasm and stimulation intervals less than the period of isomerization. It is possible to transfect HeLa cells with ChR2 and control their survival under blue light stimulation. We suggest that this practice should be considered in the future development of optogenetic systems in biological or biomedical research.
Collapse
Affiliation(s)
- Claudio Córdova
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlo Lozano
- Servicio de Anatomía Patológica, Carlos Van Buren Hospital, Valparaíso, Chile
| | - Belén Rodríguez
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Ivanny Marchant
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigación Interoperativo en Ciencias Odontológicas y Médicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Rodrigo Zúñiga
- Servicio de Anatomía Patológica, Carlos Van Buren Hospital, Valparaíso, Chile
| | - Paola Ochova
- Servicio de Anatomía Patológica, Carlos Van Buren Hospital, Valparaíso, Chile
| | - Pablo Olivero
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigación Interoperativo en Ciencias Odontológicas y Médicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Wilfredo Alejandro González-Arriagada
- Oral and Maxillofacial Pathology, Facultad de Odontología, Universidad de Los Andes, Las Condes, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de Los Andes, Las Condes, Chile
| |
Collapse
|
8
|
Sato T, Takeuchi M, Karasawa Y, Ito M. Profiles of Cytokines Secreted by ARPE-19 Cells Exposed to Light and Incubated with Anti-VEGF Antibody. Biomedicines 2021; 9:biomedicines9101333. [PMID: 34680450 PMCID: PMC8533158 DOI: 10.3390/biomedicines9101333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
The retinal pigment epithelium (RPE) is the major source of cytokines in the retina regulating the intraocular immune environment, and a primary target of photodamage. Here, we examined 27 types of cytokines secreted by ARPE-19 cells exposed to visible light and incubated with aflibercept or ranibizumab, which are two anti-vascular endothelial growth factor (VEGF) antibodies. The cells were cultured for 24 h in the dark or under 2000 lux irradiation from a daylight-colored fluorescent lamp, and cytokine levels in the culture supernatant were measured. In the light-irradiated culture, the levels of IL-9, IL-17A and bFGF were higher, and the levels of IL-6, IL-7, IL-8 and MCP-1 were lower than those in the dark culture, while there was no significant difference with the VEGF-A level. In subgroup analyses of the light-irradiated culture, the bFGF level under 250 to 2000 lux irradiation was elevated in a light intensity-dependent manner. In culture exposed to blue, green or red light, the bFGF level was elevated by blue light and was high compared to that by green or red light. In culture with aflibercept or ranibizumab in the dark, the levels of IL-6, IL-8, bFGF and MCP-1 were increased, and the IL-12 level decreased synchronously with a reduction in the VEGF-A level. Our findings indicate that continuous irradiation of visible light and VEGF suppression may be an influential factor in expression patterns of inflammatory cytokines secreted by human RPE cells.
Collapse
Affiliation(s)
- Tomohito Sato
- Department of Ophthalmology, National Defense Medical College, Tokorozawa 359-8513, Japan; (T.S.); (Y.K.)
| | - Masaru Takeuchi
- Department of Ophthalmology, National Defense Medical College, Tokorozawa 359-8513, Japan; (T.S.); (Y.K.)
- Correspondence: ; Tel.: +81-4-2995-1683; Fax: +81-4-2993-5332
| | - Yoko Karasawa
- Department of Ophthalmology, National Defense Medical College, Tokorozawa 359-8513, Japan; (T.S.); (Y.K.)
| | - Masataka Ito
- Department of Developmental Anatomy and Regenerative Biology, National Defense Medical College, Tokorozawa 359-8513, Japan;
| |
Collapse
|
9
|
Establishing the ground squirrel as a superb model for retinal ganglion cell disorders and optic neuropathies. J Transl Med 2021; 101:1289-1303. [PMID: 34253851 PMCID: PMC8753557 DOI: 10.1038/s41374-021-00637-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/08/2023] Open
Abstract
Retinal ganglion cell (RGC) death occurs after optic nerve injury due to acute trauma or chronic degenerative conditions such as optic neuropathies (e.g., glaucoma). Currently, there are no effective therapies to prevent permanent vision loss resulting from RGC death, underlining the need for research on the pathogenesis of RGC disorders. Modeling human RGC/optic nerve diseases in non-human primates is ideal because of their similarity to humans, but has practical limitations including high cost and ethical considerations. In addition, many retinal degenerative disorders are age-related making the study in primate models prohibitively slow. For these reasons, mice and rats are commonly used to model RGC injuries. However, as nocturnal mammals, these rodents have retinal structures that differ from primates - possessing less than one-tenth of the RGCs found in the primate retina. Here we report the diurnal thirteen-lined ground squirrel (TLGS) as an alternative model. Compared to other rodent models, the number and distribution of RGCs in the TLGS retina are closer to primates. The TLGS retina possesses ~600,000 RGCs with the highest density along the equatorial retina matching the location of the highest cone density (visual streak). TLGS and primate retinas also share a similar interlocking pattern between RGC axons and astrocyte processes in the retina nerve fiber layer (RNFL). In addition, using TLGS we establish a new partial optic nerve injury model that precisely controls the extent of injury while sparing a portion of the retina as an ideal internal control for investigating the pathophysiology of axon degeneration and RGC death. Moreover, in vivo optical coherence tomography (OCT) imaging and ex vivo microscopic examinations of the retina in optic nerve injured TLGS confirm RGC loss precedes proximal axon degeneration, recapitulating human pathology. Thus, the TLGS retina is an excellent model, for translational research in neurodegeneration and therapeutic neuroprotection.
Collapse
|
10
|
Plasma Rich in Growth Factors Promotes Autophagy in ARPE19 Cells in Response to Oxidative Stress Induced by Blue Light. Biomolecules 2021; 11:biom11070954. [PMID: 34203504 PMCID: PMC8301887 DOI: 10.3390/biom11070954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) causes the degeneration of photoreceptors and retinal cells leading to vision loss in older subjects. Among possible exogenous risk factors, it has been recently proposed that long-term exposure to blue light could aggravate the course of AMD. In the search for therapeutic options, plasma rich in growth factors (PRGF) has been shown to enhance cell antioxidant pathways and protect photoreceptors against the harm produced by blue light, although its mechanism of action remains unknown. One possible mechanism, autophagy, is one of the most conservative cell renewal systems used in eukaryotes to destroy cellular components that have been damaged by some kind of insult. The oxidative stress of exposure to blue light is known to induce cell autophagy. In this study, we examined the combined effects on autophagy of blue light and PRGF in a retinal cell line, ARPE19. In response to treatment with both PRGF and blue light, we detected the modulated expression of autophagy markers such as NF-kB, p62/sqstm1, Atg5, LC3 and Beclin1, and inflammatory markers such as IL1B and IL18. Our findings suggest that PRGF promotes cell autophagy in response to exposure to blue light.
Collapse
|
11
|
Yang D, Rong R, Yang R, You M, Wang M, Li H, Ji D. CaMK II -induced Drp1 phosphorylation contributes to blue light-induced AIF-mediated necroptosis in retinal R28 cells. Biochem Biophys Res Commun 2021; 559:113-120. [PMID: 33940381 DOI: 10.1016/j.bbrc.2021.04.082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 11/17/2022]
Abstract
Retinal damage caused by blue light has become an important public health concern. Mitochondria have been found to play a key role in light-induced retinal cell death. In this study, we aimed to clarify the molecular mechanism involved in mitochondrion-related retinal cell damage caused by blue light, the major component of light-emitting diodes (LEDs). Our results show that blue light (450 nm, 300lux)-induced R28 cell death is caspase independent and can be attenuated by necrostatin-1. Apoptosis-inducing factor (AIF) cleavage and translocation to the nucleus are involved in the cell death progress. Blue light exposure causes mitochondrial fragmentation, which is mediated by phosphorylation at dynamin-related protein 1 (Drp1) Ser616 site, but it does not alter the protein levels of fission or fusion machinery. Knocking down Drp1 or treatment with Drp1 inhibitor Mdivi-1 protects R28 cells from blue light. Overproduction of reactive oxygen species (ROS) is induced by blue light. The ROS scavenger Trolox decreases Drp1 Ser616 phosphorylation level and mitochondrial fragmentation upon blue light exposure. Moreover, Calcium/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN93 blocks Drp1 phosphorylation and rescues mitochondrial fragmentation and AIF-mediated cell death caused by blue light. In conclusion, our data suggest that the CaMKII-Drp1 pathway plays a major role in blue light-induced AIF-mediated retinal cell damage.
Collapse
Affiliation(s)
- Dawei Yang
- The School of Life Sciences, Central South University, Changsha, 410078, Hunan Province, China; Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China
| | - Rong Rong
- Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China
| | - Rongliang Yang
- Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China
| | - Mengling You
- Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China
| | - Mengxiao Wang
- Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China
| | - Haibo Li
- Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China.
| | - Dan Ji
- Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
12
|
Jin M, Li X, Yan F, Chen W, Jiang L, Zhang X. The effects of low-color-temperature dual-primary-color light-emitting diodes on three kinds of retinal cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 214:112099. [PMID: 33285486 DOI: 10.1016/j.jphotobiol.2020.112099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/03/2020] [Accepted: 11/23/2020] [Indexed: 12/20/2022]
Abstract
Long-term illumination of the retina with blue-light-excited phosphor-converted light-emitting diodes (LEDs) may result in decreased retinal function, even if the levels of blue light emitted are low. New low-color-temperature dual-primary-color LEDs have been developed that are composed of only two LED chips: a red chip and a yellow chip. These LEDs are expected to become a new type of healthy lighting source because they do not emit blue light, they lack phosphor, and they solve the problem of low efficiency encountered with phosphor-converted low-color-temperature LEDs. Many studies have indicated that these new low-color-temperature LEDs are likely to have therapeutic effects. However, the biological safety of these LEDs needs to be explored before the therapeutic effects are explored. Therefore, this experiment was conducted to investigate the effects of the new low-color-temperature LEDs and fluorescent white LEDs on three types of retinal cells. We observed that the viability and numbers of retinal cells decreased gradually with increasing LED color temperature. The new low-color-temperature LEDs caused less death and adverse effects on proliferation than the fluorescent white LEDs. After irradiation with high-color-temperature LEDs, the expression of Zonula Occludens-1 (ZO-1) was decreased and discontinuous in ARPE-19 cells; the stress protein hemeoxygenase-1 (HO-1) was upregulated in R28 cells; and glial fibrillary acidic protein (GFAP) and vimentin were upregulated in rMC-1 cells. We therefore conclude that the new white LEDs cause almost no damage to retinal cells and reduce the potential human health risks of chronic exposure to fluorescent white LEDs.
Collapse
Affiliation(s)
- Ming Jin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, Jiangxi 330006, China
| | - Xiongfeng Li
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, Jiangxi 330006, China
| | - Feng Yan
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, Jiangxi 330006, China
| | - Weixin Chen
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, Jiangxi 330006, China
| | - Lei Jiang
- Queen Mary School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi 330046, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
13
|
Zhao Y, Shen Y. Light-Induced Retinal Ganglion Cell Damage and the Relevant Mechanisms. Cell Mol Neurobiol 2020; 40:1243-1252. [PMID: 32107750 PMCID: PMC11448955 DOI: 10.1007/s10571-020-00819-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/18/2020] [Indexed: 12/21/2022]
Abstract
While light is the basic element for inducing vision and modulating circadian rhythms, excessive light has been reported to have a negative effect on the survival of various types of retinal cells. Among them photoreceptors and retinal pigment epithelial (RPE) cells degeneration after light exposure is widely observed, but light-induced retinal ganglion cell (RGC) damage achieves relatively little attention. The purpose of this article is to summarize the experimental evidence for the possible negative effects of excessive light on RGCs. By searching the database, twenty-six related articles have been included. Taken together, excessive light may insult RGCs through the three main ways: (i) directly action on RGC mitochondria, as well as DNA, resulting in an upregulation of reactive oxygen species (ROS) and subsequently caspase-dependent or -independent cell death; (ii) mediation in gliotransmitters or relevant receptors of retinal glial cells; and (iii) a secondary event to photoreceptors and RPE cells degeneration and subsequent retinal remodeling. So RGCs can certainly be injured by excessive light, especially when they are already energetically compromised in some diseases. And more attentions should be paid to this topic to take timely measures to protect these frail RGCs from being damaged by excessive light.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Ophthalmology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
14
|
Baudouin C, Kolko M, Melik-Parsadaniantz S, Messmer EM. Inflammation in Glaucoma: From the back to the front of the eye, and beyond. Prog Retin Eye Res 2020; 83:100916. [PMID: 33075485 DOI: 10.1016/j.preteyeres.2020.100916] [Citation(s) in RCA: 235] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 02/08/2023]
Abstract
The pathophysiology of glaucoma is complex, multifactorial and not completely understood. Elevated intraocular pressure (IOP) and/or impaired retinal blood flow may cause initial optic nerve damage. In addition, age-related oxidative stress in the retina concurrently with chronic mechanical and vascular stress is crucial for the initiation of retinal neurodegeneration. Oxidative stress is closely related to cell senescence, mitochondrial dysfunction, excitotoxicity, and neuroinflammation, which are involved in glaucoma progression. Accumulating evidence from animal glaucoma models and from human ocular samples suggests a dysfunction of the para-inflammation in the retinal ganglion cell layer and the optic nerve head. Moreover, quite similar mechanisms in the anterior chamber could explain the trabecular meshwork dysfunction and the elevated IOP in primary open-angle glaucoma. On the other hand, ocular surface disease due to topical interventions is the most prominent and visible consequence of inflammation in glaucoma, with a negative impact on filtering surgery failure, topical treatment efficacy, and possibly on inflammation in the anterior segment. Consequently, glaucoma appears as an outstanding eye disease where inflammatory changes may be present to various extents and consequences along the eye structure, from the ocular surface to the posterior segment, and the visual pathway. Here we reviewed the inflammatory processes in all ocular structures in glaucoma from the back to the front of the eye and beyond. Our approach was to explain how para-inflammation is necessary to maintain homoeostasis, and to describe abnormal inflammatory findings observed in glaucomatous patients or in animal glaucoma models, supporting the hypothesis of a dysregulation of the inflammatory balance toward a pro-inflammatory phenotype. Possible anti-inflammatory therapeutic approaches in glaucoma are also discussed.
Collapse
Affiliation(s)
- Christophe Baudouin
- Quinze-Vingts National Ophthalmology Hospital, INSERM-DGOS CIC 1423, IHU Foresight, Paris, France; Sorbonne Université, INSERM, CNRS, Institut de La Vision, Paris, France; Department of Ophthalmology, Ambroise Paré Hospital, APHP, Université de Versailles Saint-Quentin en Yvelines, Boulogne-Billancourt, France.
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| | | | | |
Collapse
|
15
|
Plasma Rich in Growth Factors Enhances Cell Survival after in Situ Retinal Degeneration. Int J Mol Sci 2020; 21:ijms21207442. [PMID: 33050198 PMCID: PMC7590176 DOI: 10.3390/ijms21207442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The purpose of this study was to examine the effect of plasma rich in growth factors (PRGFs) under blue light conditions in an in vivo model of retinal degeneration. METHODS Male Wistar rats were exposed to dark/blue light conditions for 9 days. On day 7, right eyes were injected with saline and left eyes with PRGF. Electroretinography (ERG) and intraocular pressure (IoP) measurements were performed before and after the experiment. After sacrifice, retinal samples were collected. Hematoxylin and eosin staining was performed to analyze the structure of retinal sections. Immunofluorescence for brain-specific homeobox/POU domain protein 3A (Brn3a), choline acetyltransferase (ChAT), rhodopsin, heme oxygenase-1 (HO-1), and glial fibrillary acidic protein (GFAP) was performed to study the retinal conditions. RESULTS Retinal signaling measured by ERG was reduced by blue light and recovered with PRGF; however, IoP measurements did not show significant differences among treatments. Blue light reduced the expression for Brn3a, ChAT, and rhodopsin. Treatment with PRGF showed a recovery in their expressions. HO-1 and GFAP results showed that blue light increased their expression but the use of PRGF reduced the effect of light. CONCLUSIONS Blue light causes retinal degeneration. PRGF mitigated the injury, restoring the functionality of these cells and maintaining the tissue integrity.
Collapse
|
16
|
Abstract
Since the original ICNIRP Statement was published in 2000, there have been significant improvements in the efficiency and radiance (i.e., optical radiation emission) of LEDs. The most important improvement is the development of 'white' LEDs that can be used as general lighting sources, which are more efficient than traditional lighting sources. LEDs emitting in the ultraviolet wavelength region have also become available and have made their way into consumer products. All these changes have led to a rise in concern for the safety of the optical radiation emissions from LEDs. Several in vitro and animal studies have been conducted, which indicate that blue and white LEDs can potentially cause retinal cell damage under high irradiance and lengthy exposure conditions. However, these studies cannot be directly extrapolated to normal exposure conditions for humans, and equivalent effects can also be caused by the optical radiation from other light sources under extreme exposure conditions. Acute damage to the human retina from typical exposure to blue or white LEDs has not been demonstrated. Concern for potential long-term effects, e.g. age-related macular degeneration (AMD), remains based on epidemiological studies indicating a link between high levels of exposure to sunlight and AMD. When evaluating the optical radiation safety of LEDs, it has now been established that published safety standards for lamps, not lasers, should be applied. Thus far, the only clear, acute adverse health effects from LEDs are those due to temporal light modulation (including flicker). Glare can also create visual disturbances when LED light fixtures are not properly designed. Further research is needed on potential health effects from short- and long-term exposure to new and emerging lighting technologies.
Collapse
|
17
|
Fasciani I, Petragnano F, Aloisi G, Marampon F, Rossi M, Coppolino MF, Rossi R, Longoni B, Scarselli M, Maggio R. A New Threat to Dopamine Neurons: The Downside of Artificial Light. Neuroscience 2020; 432:216-228. [PMID: 32142863 DOI: 10.1016/j.neuroscience.2020.02.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022]
Abstract
Growing awareness of adverse impacts of artificial light on human health has led to recognize light pollution as a significant global environmental issue. Despite, a large number of studies in rodent and monkey models of Parkinson's disease have reported that near infrared light has neuroprotective effects on dopaminergic neurons, recent findings have shown that prolonged exposure of rodents and birds to fluorescent artificial light results in an increase of neuromelanin granules in substantia nigra and loss of dopaminergic neurons. The observed detrimental effect seems to be dependent on a direct effect of light on the substantia nigra rather than a secondary effect of the alterations of circadian rhythms. Moreover, inferences from animal models to human studies have shown a positive correlation between the prevalence of Parkinson's disease and light pollution. The present article discusses experimental evidence supporting a potentially deleterious impact of light on dopaminergic neurons and highlights the mechanisms whereby light might damage neuronal tissue. Moreover, it analyses epidemiological evidence that suggests light pollution to be an environmental risk factor for Parkinson's disease.
Collapse
Affiliation(s)
- Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Petragnano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Gabriella Aloisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Mario Rossi
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Maria Francesca Coppolino
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Rossi
- Ph D Programme in Neuroscience, University Tor Vergata, Rome, Italy
| | - Biancamaria Longoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
18
|
Antioxidant Role of PRGF on RPE Cells after Blue Light Insult as a Therapy for Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21031021. [PMID: 32033116 PMCID: PMC7037919 DOI: 10.3390/ijms21031021] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress has a strong impact on the development of retinal diseases such as age-related macular degeneration (AMD). Plasma rich in growth factors (PRGF) is a novel therapeutic approach in ophthalmological pathologies. The aim of this study was to analyze the antioxidant effect of PRGF in retinal epithelial cells (EPR) in in vitro and ex vivo retinal phototoxicity models. In vitro analyses were performed on ARPE19 human cell line. Viability and mitochondrial status were assessed in order to test the primary effects of PRGF. GSH level, and protein and gene expression of the main antioxidant pathway (Keap1, Nrf2, GCL, HO-1, and NQO1) were also studied. Ex vivo analyses were performed on rat RPE, and HO-1 and Nrf2 gene and protein expression were evaluated. The results show that PRGF reduces light insult by stimulating the cell response against oxidative damage and modulates the antioxidant pathway. We conclude that PRGF’s protective effect could prove useful as a new therapy for treating neurodegenerative disorders such as AMD.
Collapse
|
19
|
Spectral Evaluation of Eyeglass Blocking Efficiency of Ultraviolet/High-energy Visible Blue Light for Ocular Protection. Optom Vis Sci 2020; 96:513-522. [PMID: 31274740 PMCID: PMC6615932 DOI: 10.1097/opx.0000000000001393] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Supplemental digital content is available in the text. SIGNIFICANCE We investigated, for safety and awareness, ultraviolet and high-energy violet light–blocking protection provided by assorted types of eyewear. Ultraviolet and high-energy violet light–filtering efficiency varied and did not correlate with price or advertised claims. Standardization of methods and specifications for lens spectral transmission evaluation is recommended. PURPOSE Studies have linked exposure of high-energy visible blue light to effect and damage on retinal epithelial cells, photoreceptors, and ganglion cells. “Blue light” is more accurately differentiated into “high-energy visible blue-violet light” and “circadian rhythm blue-turquoise light.” This study measured and compared spectral transmission of ultraviolet and high-energy violet light of low-, medium-, and high-priced sunglasses. METHODS Sunglasses and lens blanks were obtained from the University of Texas Medical Branch Optical Shop and vendors. Groups were based on promotional, retail, designer sunglasses, or “blue blocker” lenses. The percent transmittance of ultraviolet/visible spectral scans (800 to 350 nm) was measured using an Agilent Cary 50 spectrophotometer. High-energy violet/blue light was defined as 400 to 450 nm. RESULTS Promotional sunglasses (tinted polycarbonate) blocked 100% ultraviolet and 67 to 99.8% high-energy violet blue light. Retail sunglasses filtered out 95 to 100% ultraviolet A and 67% high-energy violet light. The tested designer sunglasses varied widely in their optical transmissibility with respect to their ultraviolet A and high-energy violet light–blocking properties, with some not blocking ultraviolet A. Clear and colorless Kodak Total Blue provided maximal high-energy violet protection, whereas clear Essilor Crizal Prevencia provided less high-energy violet blocking between 400 and 450 nm. CONCLUSIONS The ultraviolet and high-energy violet (400 to 450 nm) light–filtering efficiency varied between sunglasses and clear lenses and did not correlate with price or advertised claims. Standardization of methods and specifications for lens spectral transmission evaluation is recommended.
Collapse
|
20
|
Tao JX, Zhou WC, Zhu XG. Mitochondria as Potential Targets and Initiators of the Blue Light Hazard to the Retina. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6435364. [PMID: 31531186 PMCID: PMC6721470 DOI: 10.1155/2019/6435364] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/18/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022]
Abstract
Commercially available white light-emitting diodes (LEDs) have an intense emission in the range of blue light, which has raised a range of public concerns about their potential risks as retinal hazards. Distinct from other visible light components, blue light is characterized by short wavelength, high energy, and strong penetration that can reach the retina with relatively little loss in damage potential. Mitochondria are abundant in retinal tissues, giving them relatively high access to blue light, and chromophores, which are enriched in the retina, have many mitochondria able to absorb blue light and induce photochemical effects. Therefore, excessive exposure of the retina to blue light tends to cause ROS accumulation and oxidative stress, which affect the structure and function of the retinal mitochondria and trigger mitochondria-involved death signaling pathways. In this review, we highlight the essential roles of mitochondria in blue light-induced photochemical damage and programmed cell death in the retina, indicate directions for future research and preventive targets in terms of the blue light hazard to the retina, and suggest applying LED devices in a rational way to prevent the blue light hazard.
Collapse
Affiliation(s)
- Jin-Xin Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Clinical Medicine, The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Chuan Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Clinical Medicine, The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Xin-Gen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
21
|
Bapary MAJ, Takano J, Soma S, Sankai T. Effect of blue light‐emitting diode light and antioxidant potential in a somatic cell. Cell Biol Int 2019; 43:1296-1306. [DOI: 10.1002/cbin.11131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/16/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Mohammad A. J. Bapary
- Tsukuba Primate Research CenterNational Institutes of Biomedical Innovation, Health and Nutrition 305‐0843 Ibaraki Japan
- Department of Fisheries Technology and Quality ControlFaculty of Fisheries, Sylhet Agricultural University 3100 Sylhet Bangladesh
| | - Jun‐ichiro Takano
- Tsukuba Primate Research CenterNational Institutes of Biomedical Innovation, Health and Nutrition 305‐0843 Ibaraki Japan
| | - Shogo Soma
- Tsukuba Primate Research CenterNational Institutes of Biomedical Innovation, Health and Nutrition 305‐0843 Ibaraki Japan
| | - Tadashi Sankai
- Tsukuba Primate Research CenterNational Institutes of Biomedical Innovation, Health and Nutrition 305‐0843 Ibaraki Japan
| |
Collapse
|
22
|
Sun YJ, Ma S, Fan B, Wang Y, Wang SR, Li GY. Therapeutic hypothermia protects photoreceptors through activating Cirbp pathway. Neurochem Int 2019; 126:86-95. [DOI: 10.1016/j.neuint.2019.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 02/02/2023]
|
23
|
Enhancement of corneal epithelium cell survival, proliferation and migration by red light: Relevance to corneal wound healing. Exp Eye Res 2019; 180:231-241. [DOI: 10.1016/j.exer.2019.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/13/2018] [Accepted: 01/02/2019] [Indexed: 02/04/2023]
|
24
|
Núñez-Álvarez C, Suárez-Barrio C, del Olmo Aguado S, Osborne NN. Blue light negatively affects the survival of ARPE19 cells through an action on their mitochondria and blunted by red light. Acta Ophthalmol 2019; 97:e103-e115. [PMID: 30198155 DOI: 10.1111/aos.13812] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/08/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE To ascertain whether red light, known to enhance mitochondrial function, can blunt a blue light insult to ARPE19 cells in culture. METHODS Semi-confluent ARPE19 cells cultured in 10% FBS were subjected to various regimes of treatment with blue (465-475 nm, 800 lux, 26 W/m2 ) and red (625-635 nm, 950 lux, 6.5 W/m2 ) light, as well as with toxins that inactivate specific enzymes associated with mitochondrial oxidative phosphorylation. Cultures were then analysed for cell viability (MTT assay), mitochondrial status (JC-1), ROS formation, immunocytochemistry and the activation of specific proteins by electrophoresis/Western blotting. In addition, ARPE19 cells were cultured in polycarbonate membrane inserts in culture medium containing 1% FBS. Such cultures were exposed to cycles of red, blue or a combination of red and blue light for up to 6 weeks. Culture medium was changed and the trans-epithelium membrane resistance (TER) of the inserts-containing cells was measured twice weekly. RESULTS ARPE19 cells in culture are affected negatively when exposed to blue light. This is indicated by a loss of viability, a depolarization of their mitochondria and a stimulation of ROS. Moreover, blue light causes an up-regulation of HO-1 and phospho-p-38-MAPK and a cleavage of apoptosis inhibitory factor, proteins which are all known to be activated during cell death. All of these negative effects of blue light are significantly blunted by the red light administered after the blue light insult in each case. ARPE19 cell loss of viability and mitochondrial potential caused by toxins that inhibit specific mitochondrial enzyme complexes was additive to an insult delivered by blue light in each case. After a time, ARPE19 cells in culture express the tight junction protein ZO-1, which is affected by blue light. The development of tight junctions between ARPE19 cells grown in inserts reached a steady peak of resistance after about 40 days and then increased very slightly over the next 40 days when still in darkness. However, maximum resistance was significantly attenuated, when cultures were treated with cycles of blue light after the initial 40 days in the dark and counteracted significantly when the blue light cycle insult was combined with red light. CONCLUSION Blue light affects mitochondrial function and also the development tight junctions between ARPE19 cells, which results in a loss of cell viability. Importantly, red light delivered after a blue light insult is significantly blunted. These findings argue for the therapeutic use of red light as a noninvasive procedure to attenuate insults caused by blue light and other insults to retinal pigment epithelial cell mitochondria that are likely to occur in age-related macular degeneration.
Collapse
|
25
|
Retinal Neuron Is More Sensitive to Blue Light-Induced Damage than Glia Cell Due to DNA Double-Strand Breaks. Cells 2019; 8:cells8010068. [PMID: 30669263 PMCID: PMC6356720 DOI: 10.3390/cells8010068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/06/2019] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Blue light is a major component of visible light and digital displays. Over-exposure to blue light could cause retinal damage. However, the mechanism of its damage is not well defined. Here, we demonstrate that blue light (900 lux) impairs cell viability and induces cell apoptosis in retinal neurocytes in vitro. A DNA electrophoresis assay shows severe DNA damage in retinal neurocytes at 2 h after blue light treatment. γ-H2AX foci, a specific marker of DNA double-strand breaks (DSBs), is mainly located in the Map2-posotive neuron other than the glia cell. After assaying the expression level of proteins related to DNA repair, Mre11, Ligase IV and Ku80, we find that Ku80 is up-regulated in retinal neurocytes after blue light treatment. Interestingly, Ku80 is mainly expressed in glia fibrillary acidic protein (GFAP)-positive glia cells. Moreover, following blue light exposure in vivo, DNA DSBs are shown in the ganglion cell layer and only observed in Map2-positive cells. Furthermore, long-term blue light exposure significantly thinned the retina in vivo. Our findings demonstrate that blue light induces DNA DSBs in retinal neurons, and the damage is more pronounced compared to glia cells. Thus, this study provides new insights into the mechanisms of the effect of blue light on the retina.
Collapse
|
26
|
Li JY, Zhang K, Xu D, Zhou WT, Fang WQ, Wan YY, Yan DD, Guo MY, Tao JX, Zhou WC, Yang F, Jiang LP, Han XJ. Mitochondrial Fission Is Required for Blue Light-Induced Apoptosis and Mitophagy in Retinal Neuronal R28 Cells. Front Mol Neurosci 2018; 11:432. [PMID: 30538621 PMCID: PMC6277708 DOI: 10.3389/fnmol.2018.00432] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/06/2018] [Indexed: 11/13/2022] Open
Abstract
Light emitting diodes (LEDs) are widely used to provide illumination due to their low energy requirements and high brightness. However, the LED spectrum contains an intense blue light component which is phototoxic to the retina. Recently, it has been reported that blue light may directly impinge on mitochondrial function in retinal ganglion cells (RGCs). Mitochondria are high dynamic organelles that undergo frequent fission and fusion events. The aim of our study was to elucidate the role of mitochondrial dynamics in blue light-induced damage in retinal neuronal R28 cells. We found that exposure to blue light (450 nm, 1000 lx) for up to 12 h significantly up-regulated the expression of mitochondrial fission protein Drp1, while down-regulating the expression of mitochondrial fusion protein Mfn2 in cells. Mitochondrial fission was simultaneously stimulated by blue light irradiation. In addition, exposure to blue light increased the production of reactive oxygen species (ROS), disrupted mitochondrial membrane potential (MMP), and induced apoptosis in R28 cells. Notably, Drp1 inhibitor Mdivi-1 and Drp1 RNAi not only attenuated blue light-induced mitochondrial fission, but also alleviated blue light-induced ROS production, MMP disruption and apoptosis in cells. Compared with Mdivi-1 and Drp1 RNAi, the antioxidant N-acetyl-L-cysteine (NAC) only slightly inhibited mitochondrial fission, while significantly alleviating apoptosis after blue light exposure. Moreover, we examined markers for mitophagy, which is responsible for the clearance of dysfunctional mitochondria. It was found that blue light stimulated the conversion of LC3B-I to LC3B-II as well as the expression of PINK1 in R28 cells. Mdivi-1 or Drp1 RNAi efficiently inhibited the blue light-induced expression of PINK1 and co-localization of LC3 with mitochondria. Thus, our data suggest that mitochondrial fission is required for blue light-induced mitochondrial dysfunction and apoptosis in RGCs.
Collapse
Affiliation(s)
- Jia-Yu Li
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Kun Zhang
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Dan Xu
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Wen-Tian Zhou
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Wen-Qing Fang
- National Engineering Technology Research Center for LED on Silicon Substrate, Nanchang University, Nanchang, China
| | - Yu-Ying Wan
- Department of Intra-Hospital Infection Management, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dan-Dan Yan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Miao-Yu Guo
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Jin-Xin Tao
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Wen-Chuan Zhou
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Fan Yang
- National Engineering Technology Research Center for LED on Silicon Substrate, Nanchang University, Nanchang, China
| | - Li-Ping Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xiao-Jian Han
- Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
27
|
Cheung LTY, Manthey AL, Lai JSM, Chiu K. Targeted Delivery of Mitochondrial Calcium Channel Regulators: The Future of Glaucoma Treatment? Front Neurosci 2017; 11:648. [PMID: 29213227 PMCID: PMC5702640 DOI: 10.3389/fnins.2017.00648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/07/2017] [Indexed: 11/18/2022] Open
Affiliation(s)
- Leanne T Y Cheung
- Department of Ophthalmology, University of Hong Kong, Hong Kong, China
| | - Abby L Manthey
- Department of Ophthalmology, University of Hong Kong, Hong Kong, China
| | - Jimmy S M Lai
- Department of Ophthalmology, University of Hong Kong, Hong Kong, China
| | - Kin Chiu
- Department of Ophthalmology, University of Hong Kong, Hong Kong, China
| |
Collapse
|
28
|
Ba-Ali S, Lund-Andersen H. Pupillometric evaluation of the melanopsin containing retinal ganglion cells in mitochondrial and non-mitochondrial optic neuropathies. Mitochondrion 2017; 36:124-129. [PMID: 28716667 DOI: 10.1016/j.mito.2017.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/29/2017] [Accepted: 07/13/2017] [Indexed: 01/08/2023]
Abstract
In recent years, chromatic pupillometry is used in humans to evaluate the activity of melanopsin expressing intrinsic photosensitive retinal ganglion cells (ipRGCs). Blue light is used to stimulate the ipRGCs and red light activates the rod/cone photoreceptors. The late re-dilation phase of pupillary light reflex is primarily driven by the ipRGCs. Optic neuropathies i.e. Leber hereditary optic neuropathy (LHON), autosomal dominant optic atrophy (ADOA), nonarteritic anterior ischemic optic neuropathy (NAION), glaucoma, optic neuritis and idiopathic intracranial hypertension (IIH) are among the diseases, which have been subject to pupillometric studies. The ipRGCs are differentially affected in these various optic neuropathies. In mitochondrial optic neuropathies, the ipRGCs are protected against degeneration, whereas in glaucoma, NAION, optic neuritis and IIH the ipRGCs are damaged. Here, we will review the results of pupillometric, histopathological and animal studies evaluating the ipRGCs in mitochondrial and non-mitochondrial optic neuropathies.
Collapse
Affiliation(s)
- Shakoor Ba-Ali
- Department of Ophthalmology, Rigshospitalet, Glostrup, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Henrik Lund-Andersen
- Department of Ophthalmology, Rigshospitalet, Glostrup, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Liu B, McNally S, Kilpatrick JI, Jarvis SP, O'Brien CJ. Aging and ocular tissue stiffness in glaucoma. Surv Ophthalmol 2017; 63:56-74. [PMID: 28666629 DOI: 10.1016/j.survophthal.2017.06.007] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/27/2022]
Abstract
Glaucoma is a progressive and chronic neurodegenerative disorder characterized by damage to the inner layers of the retina and deformation of the optic nerve head. The degeneration of retinal ganglion cells and their axons results in an irreversible loss of vision and is correlated with increasing age. Extracellular matrix changes related to natural aging generate a stiffer extracellular environment throughout the body. Altered age-associated ocular tissue stiffening plays a major role in a significant number of ophthalmic pathologies. In glaucoma, both the trabecular meshwork and the optic nerve head undergo extensive extracellular matrix remodeling, characterized by fibrotic changes associated with cellular and molecular events (including myofibroblast activation) that drive further tissue fibrosis and stiffening. Here, we review the literature concerning the role of age-related ocular stiffening in the trabecular meshwork, lamina cribrosa, sclera, cornea, retina, and Bruch membrane/choroid and discuss their potential role in glaucoma progression. Because both trabecular meshwork and lamina cribrosa cells are mechanosensitive, we then describe molecular mechanisms underlying tissue stiffening and cell mechanotransduction and how these cellular activities can drive further fibrotic changes within ocular tissues. An improved understanding of the interplay between age-related tissue stiffening and biological responses in the trabecular meshwork and optic nerve head could potentially lead to novel therapeutic strategies for glaucoma treatment.
Collapse
Affiliation(s)
- Baiyun Liu
- School of Physics, Conway Institute, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Sara McNally
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Jason I Kilpatrick
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Suzanne P Jarvis
- School of Physics, Conway Institute, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Colm J O'Brien
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
30
|
Shang YM, Wang GS, Sliney DH, Yang CH, Lee LL. Light-emitting-diode induced retinal damage and its wavelength dependency in vivo. Int J Ophthalmol 2017; 10:191-202. [PMID: 28251076 DOI: 10.18240/ijo.2017.02.03] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/21/2016] [Indexed: 02/07/2023] Open
Abstract
AIM To examine light-emitting-diode (LED)-induced retinal neuronal cell damage and its wavelength-driven pathogenic mechanisms. METHODS Sprague-Dawley rats were exposed to blue LEDs (460 nm), green LEDs (530 nm), and red LEDs (620 nm). Electroretinography (ERG), Hematoxylin and eosin (H&E) staining, transmission electron microscopy (TEM), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and immunohistochemical (IHC) staining, Western blotting (WB) and the detection of superoxide anion (O2-·), hydrogen peroxide (H2O2), total iron, and ferric (Fe3+) levels were applied. RESULTS ERG results showed the blue LED group induced more functional damage than that of green or red LED groups. H&E staining, TUNEL, IHC, and TEM revealed apoptosis and necrosis of photoreceptors and RPE, which indicated blue LED also induced more photochemical injury. Free radical production and iron-related molecular marker expressions demonstrated that oxidative stress and iron-overload were associated with retinal injury. WB assays correspondingly showed that defense gene expression was up-regulated after the LED light exposure with a wavelength dependency. CONCLUSION The study results indicate that LED blue-light exposure poses a great risk of retinal injury in awake, task-oriented rod-dominant animals. The wavelength-dependent effect should be considered carefully when switching to LED lighting applications.
Collapse
Affiliation(s)
- Yu-Man Shang
- Institute of Environmental Health, National Taiwan University, Taipei 10051, Taiwan, China
| | - Gen-Shuh Wang
- Institute of Environmental Health, National Taiwan University, Taipei 10051, Taiwan, China
| | - David H Sliney
- Army Medical Department, Consulting Medical Physicist, Aberdeen Proving Ground, Maryland, MD 21010-5403, USA
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University School of Medicine, Taipei 10051, Taiwan, China; Department of Ophthalmology, National Taiwan University Hospital, Taipei 10051, Taiwan, China
| | - Li-Ling Lee
- Green Energy and Environment Research Laboratories, Industrial Technology Research Institute, Chutung, Hsinchu 31040, Taiwan, China
| |
Collapse
|
31
|
Dominant optic atrophy: updates on the pathophysiology and clinical manifestations of the optic atrophy 1 mutation. Curr Opin Ophthalmol 2016; 27:475-480. [PMID: 27585216 DOI: 10.1097/icu.0000000000000314] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW Review recent advances in clinical and experimental studies of dominant optic atrophy (DOA) to better understand the complexities of pathophysiology caused by the optic atrophy 1 (OPA1) mutation. RECENT FINDINGS DOA is the most commonly diagnosed inherited optic atrophy, causing progressive bilateral visual loss that begins early in life. During the past 25 years, there has been substantial progress in the understanding of the clinical, genetic, and pathophysiological basis of this disease. The histopathological hallmark of DOA is the primary degeneration of retinal ganglion cells, preferentially in the papillomacular bundle, which results temporal optic disc pallor and cecocentral scotomata in patients with DOA. Loss of OPA1 protein function by OPA1 gene mutations causes mitochondrial dysfunction because of the loss of mitochondrial fusion, impaired mitochondrial oxidative phosphorylation, increases in reactive oxygen species, and altered calcium homeostasis. These factors lead to apoptosis of retinal ganglion cells by a haploinsufficiency mechanism. SUMMARY Improved understanding of the pathophysiology of DOA provides insights that can be used to develop therapeutic approaches to the DOA.
Collapse
|
32
|
Visual light effects on mitochondria: The potential implications in relation to glaucoma. Mitochondrion 2016; 36:29-35. [PMID: 27890822 DOI: 10.1016/j.mito.2016.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/02/2016] [Accepted: 11/23/2016] [Indexed: 12/18/2022]
Abstract
Light of different wave-lengths have the potential to interact with four major mitochondrial protein complexes that are involved in the generation of ATP. Neurones of the central nervous system have an absolute dependence on mitochondrial generated ATP. Laboratory studies show that short-wave or blue light (400-480nm) that impinges on the retina affect flavin and cytochrome constituents associated with mitochondria to decrease the rate of ATP formation, stimulate ROS and results in cell death. This suggests that blue light could potentially have a negative influence on retinal ganglion cell (RGC) mitochondria that are abundant and not shielded by macular pigments as occurs for photoreceptor mitochondria. This might be of significance in glaucoma where it is likely that RGC mitochondria are already affected and therefore be more susceptible to blue light. Thus simply filtering out some natural blue light from entering the eye might be beneficial for the treatment of glaucoma. Long-wave or red light (650-800nm) affects mitochondrial complex IV or cytochrome oxidase to increase the rate of formation of ATP and ROS causing the generation of a number of beneficial factors. Significantly, laboratory studies show that increasing the normal amount of natural red light reaching rat RGC mitochondria in situ, subjected to ischemia, proved to be beneficial. A challenge now is to test whether extra red light delivered to the human retina can slow-down RGC loss in glaucoma. Such a methodology has also the advantage of being non-invasive. One very exciting possibility might be in the production of a lens where solar UV light is convertes to add to the amount of natural red light entering the eye.
Collapse
|
33
|
del Olmo-Aguado S, Núñez-Álvarez C, Osborne NN. Red light of the visual spectrum attenuates cell death in culture and retinal ganglion cell death in situ. Acta Ophthalmol 2016; 94:e481-91. [PMID: 26928988 DOI: 10.1111/aos.12996] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022]
Abstract
PURPOSE To ascertain whether red light, known to enhance mitochondrial function, can blunt chemical insults to cell cultures and ischaemic insults to the rat retina. METHODS Raised intraocular pressure (IOP, 140 mmHg, 60 min) or ischaemia was delivered in complete darkness or in the presence of low intensity red light (16.5 watts/m(2) , 3000 lux, 625-635 nm) to one eye of each rat. Animals were killed at specific times after ischemia and retinas analysis for ganglion cell numbers, the localization of specific antigens or for changes in defined RNAs. RGC-5 cell cultures were also exposed to various chemical insults in the presence or absence of red light. Significant differences were determined by t-test and anova. RESULTS Elevation of IOP causes changes in the localization of glial fibrillary acid protein (GFAP), calretinin, calbindin, choline acetyltransferase, ganglion cell numbers and an elevation (GFAP, vimentin, HO-1 and mTORC1) or reduction (Thy-1 and Brn3a) of mRNAs in the rat retina. These negative effects to the rat retina caused by ischaemia are reduced by red light. Moreover, chemical insults to cell cultures are blunted by red light. CONCLUSIONS Low, non-toxic levels of red light focussed on the retina for a short period of time are sufficient to attenuate an insult of raised IOP to the rat retina. Since mitochondrial dysfunctions are thought to play a major role in ganglion cell death in glaucoma, we propose the potential use of red light therapy for the treatment of the disease.
Collapse
|
34
|
Blue Light Action on Mitochondria Leads to Cell Death by Necroptosis. Neurochem Res 2016; 41:2324-35. [DOI: 10.1007/s11064-016-1946-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/28/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
|
35
|
Hopkins SL, Siewert B, Askes SHC, Veldhuizen P, Zwier R, Heger M, Bonnet S. An in vitro cell irradiation protocol for testing photopharmaceuticals and the effect of blue, green, and red light on human cancer cell lines. Photochem Photobiol Sci 2016; 15:644-53. [PMID: 27098927 PMCID: PMC5044800 DOI: 10.1039/c5pp00424a] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/30/2016] [Indexed: 12/21/2022]
Abstract
Traditionally, ultraviolet light (100-400 nm) is considered an exogenous carcinogen while visible light (400-780 nm) is deemed harmless. In this work, a LED irradiation system for in vitro photocytotoxicity testing is described. The LED irradiation system was developed for testing photopharmaceutical drugs, but was used here to determine the basal level response of human cancer cell lines to visible light of different wavelengths, without any photo(chemo)therapeutic. The effects of blue (455 nm, 10.5 mW cm(-2)), green (520 nm, 20.9 mW cm(-2)), and red light (630 nm, 34.4 mW cm(-2)) irradiation was measured for A375 (human malignant melanoma), A431 (human epidermoid carcinoma), A549 (human lung carcinoma), MCF7 (human mammary gland adenocarcinoma), MDA-MB-231 (human mammary gland adenocarcinoma), and U-87 MG (human glioblastoma-grade IV) cell lines. In response to a blue light dose of 19 J cm(-2), three cell lines exhibited a minimal (20%, MDA-MB-231) to moderate (30%, A549 and 60%, A375) reduction in cell viability, compared to dark controls. The other cell lines were not affected. Effective blue light doses that produce a therapeutic response in 50% of the cell population (ED50) compared to dark conditions were found to be 10.9 and 30.5 J cm(-2) for A375 and A549 cells, respectively. No adverse effects were observed in any of the six cell lines irradiated with a 19 J cm(-2) dose of 520 nm (green) or 630 nm (red) light. The results demonstrate that blue light irradiation can have an effect on the viability of certain human cancer cell types and controls should be used in photopharmaceutical testing, which uses high-energy (blue or violet) visible light activation.
Collapse
Affiliation(s)
- S. L. Hopkins
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| | - B. Siewert
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| | - S. H. C. Askes
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| | - P. Veldhuizen
- Leiden Institute of Physics , Leiden University , Niels Bohrweg 2 , 2333CA Leiden , The Netherlands
| | - R. Zwier
- Leiden Institute of Physics , Leiden University , Niels Bohrweg 2 , 2333CA Leiden , The Netherlands
| | - Michal Heger
- Department of Experimental Surgery , Academic Medical Center , University of Amsterdam , Meibergdreef 9 , 1105 AZ Amsterdam , The Netherlands
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| |
Collapse
|
36
|
Angebault C, Guichet PO, Talmat-Amar Y, Charif M, Gerber S, Fares-Taie L, Gueguen N, Halloy F, Moore D, Amati-Bonneau P, Manes G, Hebrard M, Bocquet B, Quiles M, Piro-Mégy C, Teigell M, Delettre C, Rossel M, Meunier I, Preising M, Lorenz B, Carelli V, Chinnery PF, Yu-Wai-Man P, Kaplan J, Roubertie A, Barakat A, Bonneau D, Reynier P, Rozet JM, Bomont P, Hamel CP, Lenaers G. Recessive Mutations in RTN4IP1 Cause Isolated and Syndromic Optic Neuropathies. Am J Hum Genet 2015; 97:754-60. [PMID: 26593267 DOI: 10.1016/j.ajhg.2015.09.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/25/2015] [Indexed: 12/28/2022] Open
Abstract
Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.
Collapse
Affiliation(s)
- Claire Angebault
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Pierre-Olivier Guichet
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Yasmina Talmat-Amar
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Majida Charif
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - Sylvie Gerber
- INSERM U1163, Hôpital Necker Enfants-Malades, 75015 Paris, France
| | - Lucas Fares-Taie
- INSERM U1163, Hôpital Necker Enfants-Malades, 75015 Paris, France
| | - Naig Gueguen
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - François Halloy
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - David Moore
- Institute of Genetic Medicine, Centre for Life, Newcastle University and Wellcome Trust Centre for Mitochondrial Research, NE1 3BZ Newcastle upon Tyne, UK
| | - Patrizia Amati-Bonneau
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - Gael Manes
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Maxime Hebrard
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Béatrice Bocquet
- Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Mélanie Quiles
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Camille Piro-Mégy
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Marisa Teigell
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Cécile Delettre
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Mireille Rossel
- INSERM U710, Laboratoire MMDN EPHE, 34090 Montpellier, France
| | - Isabelle Meunier
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Markus Preising
- Department of Ophthalmology, Justus-Liebig University, 35392 Giessen, Germany
| | - Birgit Lorenz
- Department of Ophthalmology, Justus-Liebig University, 35392 Giessen, Germany
| | - Valerio Carelli
- IRCCS, Institute of Neurological Sciences of Bologna, Bellaria Hospital, 40139 Bologna, Italy; Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40139 Bologna, Italy
| | - Patrick F Chinnery
- Institute of Genetic Medicine, Centre for Life, Newcastle University and Wellcome Trust Centre for Mitochondrial Research, NE1 3BZ Newcastle upon Tyne, UK
| | - Patrick Yu-Wai-Man
- Institute of Genetic Medicine, Centre for Life, Newcastle University and Wellcome Trust Centre for Mitochondrial Research, NE1 3BZ Newcastle upon Tyne, UK; Newcastle Eye Centre, Royal Victoria Infirmary, NE1 4LP Newcastle upon Tyne, UK
| | - Josseline Kaplan
- INSERM U1163, Hôpital Necker Enfants-Malades, 75015 Paris, France
| | - Agathe Roubertie
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Abdelhamid Barakat
- Laboratoire de Génétique Moléculaire Humaine, Département de Recherche Scientifique, Institut Pasteur du Maroc, 20360 Casablanca, Morocco
| | - Dominique Bonneau
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - Pascal Reynier
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | | | - Pascale Bomont
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Christian P Hamel
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Guy Lenaers
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France.
| |
Collapse
|
37
|
German OL, Agnolazza DL, Politi LE, Rotstein NP. Light, lipids and photoreceptor survival: live or let die? Photochem Photobiol Sci 2015. [PMID: 26204250 DOI: 10.1039/c5pp00194c] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Due to its constant exposure to light and its high oxygen consumption the retina is highly sensitive to oxidative damage, which is a common factor in inducing the death of photoreceptors after light damage or in inherited retinal degenerations. The high content of docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, has been suggested to contribute to this sensitivity. DHA is crucial for developing and preserving normal visual function. However, further roles of DHA in the retina are still controversial. Current data support that it can tilt the scale either towards degeneration or survival of retinal cells. DHA peroxidation products can be deleterious to the retina and might lead to retinal degeneration. However, DHA has also been shown to act as, or to be the source of, a survival molecule that protects photoreceptors and retinal pigment epithelium cells from oxidative damage. We have established that DHA protects photoreceptors from oxidative stress-induced apoptosis and promotes their differentiation in vitro. DHA activates the retinoid X receptor (RXR) and the ERK/MAPK pathway, thus regulating the expression of anti and pro-apoptotic proteins. It also orchestrates a diversity of signaling pathways, modulating enzymatic pathways that control the sphingolipid metabolism and activate antioxidant defense mechanisms to promote photoreceptor survival and development. A deeper comprehension of DHA signaling pathways and context-dependent behavior is required to understand its dual functions in retinal physiology.
Collapse
Affiliation(s)
- Olga Lorena German
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
38
|
Siu AW, Shan SW, Li KK, Lam HY, Fung MY, Li KK, To CH, Do CW. Glutathione attenuates nitric oxide-induced retinal lipid and protein changes. Ophthalmic Physiol Opt 2015; 35:135-46. [DOI: 10.1111/opo.12198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 12/30/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Andrew W. Siu
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Sze Wan Shan
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - King Kit Li
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Hiu Yan Lam
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Man Yee Fung
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Ka Ki Li
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| | - Chi Ho To
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou China
| | - Chi Wai Do
- Laboratory of Experimental Optometry; School of Optometry; The Hong Kong Polytechnic University; Hong Kong China
| |
Collapse
|
39
|
Ong Q, Guo S, Zhang K, Cui B. U0126 protects cells against oxidative stress independent of its function as a MEK inhibitor. ACS Chem Neurosci 2015; 6:130-7. [PMID: 25544156 PMCID: PMC4304487 DOI: 10.1021/cn500288n] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
![]()
U0126 is a potent
and selective inhibitor of MEK1 and MEK2 kinases.
It has been widely used as an inhibitor for the Ras/Raf/MEK/ERK signaling
pathway with over 5000 references on the NCBI PubMed database. In
particular, U0126 has been used in a number of studies to show that
inhibition of the Raf/MEK/ERK pathway protects neuronal cells against
oxidative stress. Here, we report that U0126 can function as an antioxidant
that protects PC12 cells against a number of different oxidative-stress
inducers. This protective effect of U0126 is independent of its function
as a MEK inhibitor, as several other MEK inhibitors failed to show
similar protective effects. U0126 reduces reactive oxygen species
(ROS) in cells. We further demonstrate that U0126 is a direct ROS
scavenger in vitro, and the oxidation products of U0126 exhibit fluorescence.
Our finding that U0126 is a strong antioxidant signals caution for
its future usage as a MEK inhibitor and for interpreting some previous
results.
Collapse
Affiliation(s)
- Qunxiang Ong
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
| | - Shunling Guo
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
| | - Kai Zhang
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
- Department
of Biochemistry, University of Illinois at Urbana−Champaign, 600 South Mathews, Urbana, Illinois 61801, United States
| | - Bianxiao Cui
- Department
of Chemistry, Stanford University, 380 Roth Way, Palo Alto, California 94305, United States
| |
Collapse
|
40
|
Osborne NN, Núñez-Álvarez C, del Olmo-Aguado S. The effect of visual blue light on mitochondrial function associated with retinal ganglions cells. Exp Eye Res 2014; 128:8-14. [DOI: 10.1016/j.exer.2014.08.012] [Citation(s) in RCA: 305] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/18/2014] [Accepted: 08/20/2014] [Indexed: 11/16/2022]
|
41
|
Osborne NN, Álvarez CN, del Olmo Aguado S. Targeting mitochondrial dysfunction as in aging and glaucoma. Drug Discov Today 2014; 19:1613-22. [DOI: 10.1016/j.drudis.2014.05.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 04/16/2014] [Accepted: 05/20/2014] [Indexed: 12/21/2022]
|
42
|
Xiao JR, Do CW, To CH. Potential Therapeutic Effects of Baicalein, Baicalin, and Wogonin in Ocular Disorders. J Ocul Pharmacol Ther 2014; 30:605-14. [DOI: 10.1089/jop.2014.0074] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Jing-Ru Xiao
- Laboratory of Experimental Optometry, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, Hong Kong
| | - Chi-Wai Do
- Laboratory of Experimental Optometry, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, Hong Kong
| | - Chi-Ho To
- Laboratory of Experimental Optometry, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, Hong Kong
- State Key Laboratory of Ophthalmology, Department of Ophthalmology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
43
|
Wang Z, Pan X, Wang D, Sun H, Han F, Lv C, Zhang X. Protective effects of protocatechuic acid on retinal ganglion cells from oxidative damage induced by H2O2. Neurol Res 2014; 37:159-66. [DOI: 10.1179/1743132814y.0000000421] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
44
|
Froger N, Moutsimilli L, Cadetti L, Jammoul F, Wang QP, Fan Y, Gaucher D, Rosolen SG, Neveux N, Cynober L, Sahel JA, Picaud S. Taurine: the comeback of a neutraceutical in the prevention of retinal degenerations. Prog Retin Eye Res 2014; 41:44-63. [PMID: 24721186 DOI: 10.1016/j.preteyeres.2014.03.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 12/21/2022]
Abstract
Taurine is the most abundant amino acid in the retina. In the 1970s, it was thought to be involved in retinal diseases with photoreceptor degeneration, because cats on a taurine-free diet presented photoreceptor loss. However, with the exception of its introduction into baby milk and parenteral nutrition, taurine has not yet been incorporated into any commercial treatment with the aim of slowing photoreceptor degeneration. Our recent discovery that taurine depletion is involved in the retinal toxicity of the antiepileptic drug vigabatrin has returned taurine to the limelight in the field of neuroprotection. However, although the retinal toxicity of vigabatrin principally involves a deleterious effect on photoreceptors, retinal ganglion cells (RGCs) are also affected. These findings led us to investigate the possible role of taurine depletion in retinal diseases with RGC degeneration, such as glaucoma and diabetic retinopathy. The major antioxidant properties of taurine may influence disease processes. In addition, the efficacy of taurine is dependent on its uptake into retinal cells, microvascular endothelial cells and the retinal pigment epithelium. Disturbances of retinal vascular perfusion in these retinal diseases may therefore affect the retinal uptake of taurine, resulting in local depletion. The low plasma taurine concentrations observed in diabetic patients may further enhance such local decreases in taurine concentration. We here review the evidence for a role of taurine in retinal ganglion cell survival and studies suggesting that this compound may be involved in the pathophysiology of glaucoma or diabetic retinopathy. Along with other antioxidant molecules, taurine should therefore be seriously reconsidered as a potential treatment for such retinal diseases.
Collapse
Affiliation(s)
- Nicolas Froger
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France.
| | - Larissa Moutsimilli
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Lucia Cadetti
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Firas Jammoul
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Qing-Ping Wang
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Yichao Fan
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - David Gaucher
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France; Nouvel hôpital civil, hôpitaux universitaires de Strasbourg and Laboratoire de Bactériologie (EA-7290), Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, France
| | - Serge G Rosolen
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Nathalie Neveux
- Department of Nutrition, Faculty of Pharmacy, Paris Descartes University, Paris, France; Clinical Chemistry, Hôtel-Dieu-Cochin Hospitals, AP-HP, Paris, France
| | - Luc Cynober
- Department of Nutrition, Faculty of Pharmacy, Paris Descartes University, Paris, France; Clinical Chemistry, Hôtel-Dieu-Cochin Hospitals, AP-HP, Paris, France
| | - José-Alain Sahel
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France; Institute of Ophthalmology, University College of London, UK; Fondation Ophtalmologique Adolphe de Rothschild, Paris, France; French Academy of Sciences, Paris, France
| | - Serge Picaud
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France; Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.
| |
Collapse
|
45
|
Long-term blue light exposure induces RGC-5 cell death in vitro: involvement of mitochondria-dependent apoptosis, oxidative stress, and MAPK signaling pathways. Apoptosis 2014; 19:922-32. [DOI: 10.1007/s10495-014-0983-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Li GY, Fan B, Jiao YY. Rapamycin attenuates visible light-induced injury in retinal photoreceptor cells via inhibiting endoplasmic reticulum stress. Brain Res 2014; 1563:1-12. [PMID: 24607296 DOI: 10.1016/j.brainres.2014.02.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/19/2014] [Accepted: 02/08/2014] [Indexed: 01/08/2023]
Abstract
An extended exposure of the retina to visible light may lead to photochemical damage in retinal photoreceptor cells. The exact mechanism of retinal light damage remains unknown, and an effective therapy is still unavailable. Here, we demonstrated that rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), markedly protected 661W photoreceptor cells from visible light exposure-induced damage at the nanomolar level. We also observed by transmission electron microscopy that light exposure led to severe endoplasmic reticulum (ER) stress in 661W cells as well as abnormal endomembranes and ER membranes. In addition, obvious upregulated ER stress markers were monitored by western blot at the protein level and by quantitative reverse transcription-polymerase chain reaction (RT-PCR) at the mRNA level. Interestingly, rapamycin pretreatment significantly suppressed light-induced ER stress and all three major branches of the unfolded protein response (UPR), including the RNA-dependent protein kinase-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6) pathways both at the protein and mRNA levels. Additionally, the inhibition of ER stress by rapamycin was further confirmed with a dithiothreitol (DTT; a classical ER stress inducer)-damaged 661W cell model. Meanwhile, our results also revealed that rapamycin was able to remarkably inhibit the activation of mTOR and its downstream factors eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1), p-4EBP1, p70, p-p70, and phosphorylated ribosomal protein S6 kinase (p-S6K) in the light-injured 661W cells. Thus, these data indicate that visible light induces ER stress in 661W cells; whereas the mTOR inhibitor, rapamycin, effectively protects 661W cells from light injury through suppressing the ER stress pathway.
Collapse
Affiliation(s)
- Guang-Yu Li
- Department of Ophthalmology, Second Hospital of JiLin University, ChangChun 130041, China.
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of JiLin University, ChangChun 130041, China.
| | - Ying-Ying Jiao
- Department of Ophthalmology, Second Hospital of JiLin University, ChangChun 130041, China
| |
Collapse
|
47
|
Xie C, Li X, Tong J, Gu Y, Shen Y. Effects of white light-emitting diode (LED) light exposure with different correlated color temperatures (CCTs) on human lens epithelial cells in culture. Photochem Photobiol 2014; 90:853-9. [PMID: 24483628 DOI: 10.1111/php.12250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/21/2014] [Indexed: 11/30/2022]
Abstract
Cataract is the major cause for legal blindness in the world. Oxidative stress on the lens epithelial cells (hLECs) is the most important factor in cataract formation. Cumulative light-exposure from widely used light-emitting diodes (LEDs) may pose a potential oxidative threat to the lens epithelium, due to the high-energy blue light component in the white-light emission from diodes. In the interest of perfecting biosafety standards for LED domestic lighting, this study analyzed the photobiological effect of white LED light with different correlated color temperatures (CCTs) on cultured hLECs. The hLECs were cultured and cumulatively exposed to multichromatic white LED light with CCTs of 2954, 5624, and 7378 K. Cell viability of hLECs was measured by Cell Counting Kit-8 (CCK-8) assay. DNA damage was determined by alkaline comet assay. Intracellular reactive oxygen species (ROS) generation, cell cycle, and apoptosis were quantified by flow cytometry. Compared with 2954 and 5624 K LED light, LED light having a CCT of 7378 K caused overproduction of intracellular ROS and severe DNA damage, which triggered G2 /M arrest and apoptosis. These results indicate that white LEDs with a high CCT could cause significant photobiological damage to hLECs.
Collapse
Affiliation(s)
- Chen Xie
- Clinical Research Center, The First Affiliate Hospital, School Of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Roehlecke C, Schumann U, Ader M, Brunssen C, Bramke S, Morawietz H, Funk RHW. Stress reaction in outer segments of photoreceptors after blue light irradiation. PLoS One 2013; 8:e71570. [PMID: 24039718 PMCID: PMC3770596 DOI: 10.1371/journal.pone.0071570] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 07/07/2013] [Indexed: 12/26/2022] Open
Abstract
The retina is prone to oxidative stress from many factors which are also involved in the pathogenesis of degenerative diseases. In this study, we used the application of blue light as a physiological stress factor. The aim of this study was to identify the major source of intracellular ROS that mediates blue light-induced detrimental effects on cells which may lead to cytotoxicity. We hypothesized that outer segments are the major source of blue light induced ROS generation. In photoreceptors, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) enzymes and the recently found respiratory chain complexes may represent a major source for reactive oxygen species (ROS), beside mitochondria and chromophores. Therefore, we investigated this hypothesis and analysed the exact localization of the ROS source in photoreceptors in an organotypic culture system for mouse retinas. Whole eyeball cultures were irradiated with visible blue light (405 nm) with an output power of 1 mW/cm2. Blue light impingement lead to an increase of ROS production (detected by H2DCFDA in live retinal explants), which was particularly strong in the photoreceptor outer segments. Nox-2 and Nox-4 proteins are sources of ROS in blue light irradiated photoreceptors; the Nox inhibitor apocynin decreased ROS stimulated by blue light. Concomitantly, enzyme SOD-1, a member of the antioxidant defense system, indicator molecules of protein oxidation (CML) and lipid oxidation (MDA and 4-HNE) were also increased in the outer segments. Interestingly, outer segments showed a mitochondrial-like membrane potential which was demonstrated using two dyes (JC-1 and TMRE) normally exclusively associated with mitochondria. As in mitochondria, these dyes indicated a decrease of the membrane potential in hypoxic states or cell stress situations. The present study demonstrates that ROS generation and oxidative stress occurs directly in the outer segments of photoreceptors after blue light irradiation.
Collapse
Affiliation(s)
- Cora Roehlecke
- Institute of Anatomy, Technische Universität (TU) Dresden, Dresden, Germany
- * E-mail:
| | - Ulrike Schumann
- Institute of Anatomy, Technische Universität (TU) Dresden, Dresden, Germany
| | - Marius Ader
- Center for Regenerative Therapies Dresden (CRTD) DFG – Cluster of Excellence, Biotechnology Center, Dresden, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, TU Dresden, Dresden, Germany
| | - Silvia Bramke
- Institute of Anatomy, Technische Universität (TU) Dresden, Dresden, Germany
| | - Henning Morawietz
- Center for Regenerative Therapies Dresden (CRTD) DFG – Cluster of Excellence, Biotechnology Center, Dresden, Germany
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, TU Dresden, Dresden, Germany
| | - Richard H. W. Funk
- Institute of Anatomy, Technische Universität (TU) Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD) DFG – Cluster of Excellence, Biotechnology Center, Dresden, Germany
| |
Collapse
|
50
|
del Olmo-Aguado S, Núñez-Álvarez C, Ji D, Manso AG, Osborne NN. RTP801 immunoreactivity in retinal ganglion cells and its down-regulation in cultured cells protect them from light and cobalt chloride. Brain Res Bull 2013; 98:132-44. [PMID: 23978538 DOI: 10.1016/j.brainresbull.2013.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/13/2013] [Accepted: 08/14/2013] [Indexed: 10/26/2022]
Abstract
RTP801, a stress-related protein, is activated by adverse environmental conditions and inhibits the activity of mammalian target of rapamycin (mTOR) in promoting oxidative stress-dependent cell death. RTP801 exists both in the mammalian retina and the lens of the eye. Here, we observed RTP801 immunoreactivity in some retinal ganglion cells. Intravitreal injection of cobalt chloride (CoCl2) to mimick hypoxia influenced retinal GFAP (glial fibrillary acidic protein) and heme oxygenase-1 (HO-1) levels, but did not affect RTP801 immunoreactivity or mRNA content relative to GAPDH. However, RTP801 mRNA was elevated when compared with Brn3a mRNA, suggesting that RTP801 is activated in stressed Brn3a retinal ganglion cells. In cultures of RGC-5 cells, RTP801 immunoreactivity was located in the cytoplasm and partly present in the mitochondria. An insult of blue light or CoCl2 increased RTP801 expression, which was accompanied by cell death. However, in cultures where RTP801 mRNA was down-regulated, the negative influence of blue light and CoCl2 was blunted. Rapamycin nullified the CoCl2-induced up-regulation of RTP801 and attenuated cell death. Moreover, rapamycin was non-toxic to RGC-5 cells, even at a high concentration (10μM). The protective effect of rapamycin on RGC-5 cells caused by the inhibition of RTP801 suggests that rapamycin might attenuate retinal ganglion cell death in situ, as in glaucoma.
Collapse
Affiliation(s)
- Susana del Olmo-Aguado
- Fundación de Investigación Oftalmológica, Avda. Doctores Fernández-Vega 34, E-33012 Oviedo, Asturias, Spain
| | | | | | | | | |
Collapse
|