1
|
Takita Y, Sugano E, Kitabayashi K, Tabata K, Saito A, Yokoyama T, Onoguchi R, Fukuda T, Ozaki T, Bai L, Tomita H. Evaluation of Local Retinal Function in Light-Damaged Rats Using Multifocal Electroretinograms and Multifocal Visual Evoked Potentials. Int J Mol Sci 2023; 24:16433. [PMID: 38003623 PMCID: PMC10670973 DOI: 10.3390/ijms242216433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Electroretinograms (ERGs) are often used to evaluate retinal function. However, assessing local retinal function can be challenging; therefore, photopic and scotopic ERGs are used to record whole-retinal function. This study evaluated focal retinal function in rats exposed to continuous light using a multifocal ERG (mfERG) system. The rats were exposed to 1000 lux of fluorescent light for 24 h to induce photoreceptor degeneration. After light exposure, the rats were reared under cyclic light conditions (12 h: 5 lux, 12 h: dark). Photopic and multifocal ERGs and single-flash and multifocal visual evoked potentials (mfVEPs) were recorded 7 days after light exposure. Fourteen days following light exposure, paraffin-embedded sections were prepared from the eyes for histological evaluation. The ERG and VEP responses dramatically decreased after 24 h of light exposure, and retinal area-dependent decreases were observed in mfERGs and mfVEPs. Histological assessment revealed severe damage to the superior retina and less damage to the inferior retina. Considering the recorded visual angles of mfERGs and mfVEPs, the degenerated area shown on the histological examinations correlates well with the responses from multifocal recordings.
Collapse
Grants
- 21-Ⅱ4001 Terumo (Japan)
- 22H00579 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
- 21K18278 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
- 22K09760 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
- 21K09713 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan; (Y.T.); (E.S.); (K.K.); (K.T.); (A.S.); (T.Y.); (R.O.); (T.F.); (T.O.); (L.B.)
| |
Collapse
|
2
|
Sirés A, Pazo-González M, López-Soriano J, Méndez A, de la Rosa EJ, de la Villa P, Comella JX, Hernández-Sánchez C, Solé M. The Absence of FAIM Leads to a Delay in Dark Adaptation and Hampers Arrestin-1 Translocation upon Light Reception in the Retina. Cells 2023; 12:cells12030487. [PMID: 36766830 PMCID: PMC9914070 DOI: 10.3390/cells12030487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/14/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
The short and long isoforms of FAIM (FAIM-S and FAIM-L) hold important functions in the central nervous system, and their expression levels are specifically enriched in the retina. We previously described that Faim knockout (KO) mice present structural and molecular alterations in the retina compatible with a neurodegenerative phenotype. Here, we aimed to study Faim KO retinal functions and molecular mechanisms leading to its alterations. Electroretinographic recordings showed that aged Faim KO mice present functional loss of rod photoreceptor and ganglion cells. Additionally, we found a significant delay in dark adaptation from early adult ages. This functional deficit is exacerbated by luminic stress, which also caused histopathological alterations. Interestingly, Faim KO mice present abnormal Arrestin-1 redistribution upon light reception, and we show that Arrestin-1 is ubiquitinated, a process that is abrogated by either FAIM-S or FAIM-L in vitro. Our results suggest that FAIM assists Arrestin-1 light-dependent translocation by a process that likely involves ubiquitination. In the absence of FAIM, this impairment could be the cause of dark adaptation delay and increased light sensitivity. Multiple retinal diseases are linked to deficits in photoresponse termination, and hence, investigating the role of FAIM could shed light onto the underlying mechanisms of their pathophysiology.
Collapse
Affiliation(s)
- Anna Sirés
- Cell Signaling and Apoptosis Group, Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Mateo Pazo-González
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
- Department of Systems Biology, Facultad de Medicina, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Joaquín López-Soriano
- Cell Signaling and Apoptosis Group, Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Ana Méndez
- Department of Physiological Sciences, School of Medicine, Campus Universitari de Bellvitge, University of Barcelona, 08907 Barcelona, Spain
- Institut de Neurociències, Campus Universitari de Bellvitge, University of Barcelona, 08907 Barcelona, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Campus Universitari de Bellvitge, University of Barcelona, 08907 Barcelona, Spain
| | - Enrique J. de la Rosa
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Pedro de la Villa
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
- Department of Systems Biology, Facultad de Medicina, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Joan X. Comella
- Cell Signaling and Apoptosis Group, Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Catalina Hernández-Sánchez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Montse Solé
- Cell Signaling and Apoptosis Group, Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
- Correspondence:
| |
Collapse
|
3
|
Glial cell response to constant low light exposure in rat retina. Vis Neurosci 2022; 39:E005. [PMID: 36164752 DOI: 10.1017/s0952523822000049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To study the macroglia and microglia and the immune role in long-time light exposure in rat eyes, we performed glial cell characterization along the time-course of retinal degeneration induced by chronic exposure to low-intensity light. Animals were exposed to light for periods of 2, 4, 6, or 8 days, and the retinal glial response was evaluated by immunohistochemistry, western blot and real-time reverse transcription polymerase chain reaction. Retinal cells presented an increased expression of the macroglia marker GFAP, as well as increased mRNA levels of microglia markers Iba1 and CD68 after 6 days. Also, at this time-point, we found a higher number of Iba1-positive cells in the outer nuclear layer area; moreover, these cells showed the characteristic activated-microglia morphology. The expression levels of immune mediators TNF, IL-6, and chemokines CX3CR1 and CCL2 were also significantly increased after 6 days. All the events of glial activation occurred after 5-6 days of constant light exposure, when the number of photoreceptor cells has already decreased significantly. Herein, we demonstrated that glial and immune activation are secondary to neurodegeneration; in this scenario, our results suggest that photoreceptor death is an early event that occurs independently of glial-derived immune responses.
Collapse
|
4
|
Zhuang X, Ma J, Xu S, Zhang M, Xu G, Sun Z. All-Trans Retinoic Acid Attenuates Blue Light-Induced Apoptosis of Retinal Photoreceptors by Upregulating MKP-1 Expression. Mol Neurobiol 2021; 58:4157-4168. [PMID: 33950345 DOI: 10.1007/s12035-021-02380-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/31/2021] [Indexed: 11/30/2022]
Abstract
The study investigated the antiapoptotic effects of all-trans retinoic acid (RA) on retinal degeneration caused by exposure to blue light. Sprague-Dawley rats received intraperitoneal injections of RA and, if necessary, the mitogen-activated protein kinase phosphotase-1(MKP-1) inhibitor, (E)-2-benzylidene-3-(cyclohexylamino)-2, 3-dihydro-1H-inden-1-one (BCI), or the retinoic acid receptor (RAR) antagonist, AGN 193109. Retinal damage was induced by 24 h of continuous exposure to blue light. Haematoxylin and eosin staining and electroretinography were performed to measure retinal thickness and retinal function before and at 3 days and 7 days after light exposure. The retinal protein expression levels of phosphorylated c-Jun N-terminal kinase (JNK), phosphorylated nuclear factor-κB, MKP-1, Bim, Bax, and cleaved caspase-3 were also measured. Terminal-deoxynucleotidyl-transferase-mediated deoxyuridine triphosphate-biotin nick end labelling (TUNEL) staining and immunofluorescent staining of cleaved caspase-3 were also performed to evaluate photoreceptor apoptosis. The administration of RA significantly mitigated retinal dysfunction and the decrease in the outer nuclear layer (ONL) thickness at 3 days and 7 days after light exposure. RA also reduced the percentage of TUNEL-positive nuclei in the ONL and cleaved caspase-3 immunofluorescence intensity at 3 days after light exposure. Light exposure increased the retinal expression of proapoptotic proteins (Bim, Bax, and cleaved caspase-3), which was attenuated by RA. Moreover, RA enhanced the expression of MKP-1 and inhibited the phosphorylation of JNK, which were attenuated by the inhibition of RAR. The inhibitory effects of RA on blue light-induced photoreceptor apoptosis were abrogated by the MKP-1inhibitor. Our results indicate that RA alleviates photoreceptor loss following blue light exposure, at least partly, by the MKP-1/JNK pathway, which may serve as a therapeutic target for relieving retinal degeneration.
Collapse
Affiliation(s)
- Xiaonan Zhuang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Jun Ma
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Sisi Xu
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Meng Zhang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Gezhi Xu
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Zhongcui Sun
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Li H, Lian L, Liu B, Chen Y, Yang J, Jian S, Zhou J, Xu Y, Ma X, Qu J, Hou L. KIT ligand protects against both light-induced and genetic photoreceptor degeneration. eLife 2020; 9:51698. [PMID: 32242818 PMCID: PMC7170656 DOI: 10.7554/elife.51698] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Photoreceptor degeneration is a major cause of blindness and a considerable health burden during aging but effective therapeutic or preventive strategies have not so far become readily available. Here, we show in mouse models that signaling through the tyrosine kinase receptor KIT protects photoreceptor cells against both light-induced and inherited retinal degeneration. Upon light damage, photoreceptor cells upregulate Kit ligand (KITL) and activate KIT signaling, which in turn induces nuclear accumulation of the transcription factor NRF2 and stimulates the expression of the antioxidant gene Hmox1. Conversely, a viable Kit mutation promotes light-induced photoreceptor damage, which is reversed by experimental expression of Hmox1. Furthermore, overexpression of KITL from a viral AAV8 vector prevents photoreceptor cell death and partially restores retinal function after light damage or in genetic models of human retinitis pigmentosa. Hence, application of KITL may provide a novel therapeutic avenue for prevention or treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Huirong Li
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Lili Lian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jinglei Yang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Shuhui Jian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiajia Zhou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ying Xu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Cebulla CM, Kim B, George V, Heisler-Taylor T, Hamadmad S, Reese AY, Kothari SS, Kusibati R, Wilson H, Abdel-Rahman MH. Oral Selumetinib Does Not Negatively Impact Photoreceptor Survival in Murine Experimental Retinal Detachment. Invest Ophthalmol Vis Sci 2019; 60:349-357. [PMID: 30682205 PMCID: PMC6348998 DOI: 10.1167/iovs.18-25405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Purpose Mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling is neuroprotective in some retinal damage models but its role in neuronal survival during retinal detachment (RD) is unclear. In addition, serous RDs are a prevalent side effect of MEK inhibitors (MEKi), blocking MAPK/ERK signaling for treatment of certain cancers. We tested the hypothesis that MEKi treatment in experimental RD would increase photoreceptor death. Methods The MEKi selumetinib was delivered daily to C57BL/6 mice at a clinically relevant dose (10 mg/mL) starting 1 day prior to creating RD with subretinal hyaluronic acid injection. Photoreceptor TUNEL and outer nuclear layer (ONL) thickness were analyzed. Phospho-ERK1/2 (pERK) distribution, glial fibrillary acidic protein (GFAP) accumulation, and Iba-1 (microglia/macrophages) were evaluated with immunofluorescence. Results pERK accumulated in the Müller glia in detached retinas, but this was effectively blocked by selumetinib. Selumetinib did not induce serous RDs at day 1 and did not increase TUNEL positive photoreceptors or further decrease ONL thickness compared to controls. Retinal gliosis was not altered, but selumetinib did block the increase in intraretinal microglia/macrophage Iba-1 fluorescence intensity and acquisition of amoeboid morphology. Conclusions MAPK/ERK is neuroprotective in some retinal damage models; in RD, selumetinib blocked Müller pERK accumulation and changed the retinal microglia/macrophage phenotype but did not alter photoreceptor survival. This is consistent with the relatively good visual acuity seen in patients developing transient retinal detachments on MEK inhibitor therapy. Compensation by other neuroprotective pathways in the retina during retinal detachment may occur in the presence of MEK inhibition.
Collapse
Affiliation(s)
- Colleen M Cebulla
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Bongsu Kim
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Valerie George
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Tyler Heisler-Taylor
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States.,Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, United States
| | - Sumaya Hamadmad
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Alana Y Reese
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Shaili S Kothari
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Rania Kusibati
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Hailey Wilson
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Mohamed H Abdel-Rahman
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States.,Division of Human Genetics, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
7
|
Rajala RVS. Therapeutic Benefits from Nanoparticles: The Potential Significance of Nanoscience in Retinal Degenerative Diseases. JOURNAL OF MOLECULAR BIOLOGY & THERAPEUTICS 2019; 1:44-55. [PMID: 34528026 PMCID: PMC8439377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Several nanotechnology podiums have gained remarkable attention in the area of medical sciences, including diagnostics and treatment. In the past decade, engineered multifunctional nanoparticles have served as drug and gene carriers. The most important aspect of translating nanoparticles from the bench to bedside is safety. These nanoparticles should not elicit any immune response and should not be toxic to humans or the environment. Lipid-based nanoparticles have been shown to be the least toxic for in vivo applications, and significant progress has been made in gene and drug delivery employing lipid-based nanoassemblies. Several excellent reviews and reports discuss the general use and application of lipid-based nanoparticles; our review focuses on the application of lipid-based nanoparticles for the treatment of ocular diseases, and recent advances in and updates on their use.
Collapse
Affiliation(s)
- Raju V S Rajala
- Departments of Ophthalmology, Physiology and Cell Biology, University of Oklahoma Health Sciences Center, Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Stone J, Mitrofanis J, Johnstone DM, Falsini B, Bisti S, Adam P, Nuevo AB, George-Weinstein M, Mason R, Eells J. Acquired Resilience: An Evolved System of Tissue Protection in Mammals. Dose Response 2018; 16:1559325818803428. [PMID: 30627064 PMCID: PMC6311597 DOI: 10.1177/1559325818803428] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
This review brings together observations on the stress-induced regulation of resilience mechanisms in body tissues. It is argued that the stresses that induce tissue resilience in mammals arise from everyday sources: sunlight, food, lack of food, hypoxia and physical stresses. At low levels, these stresses induce an organised protective response in probably all tissues; and, at some higher level, cause tissue destruction. This pattern of response to stress is well known to toxicologists, who have termed it hormesis. The phenotypes of resilience are diverse and reports of stress-induced resilience are to be found in journals of neuroscience, sports medicine, cancer, healthy ageing, dementia, parkinsonism, ophthalmology and more. This diversity makes the proposing of a general concept of induced resilience a significant task, which this review attempts. We suggest that a system of stress-induced tissue resilience has evolved to enhance the survival of animals. By analogy with acquired immunity, we term this system 'acquired resilience'. Evidence is reviewed that acquired resilience, like acquired immunity, fades with age. This fading is, we suggest, a major component of ageing. Understanding of acquired resilience may, we argue, open pathways for the maintenance of good health in the later decades of human life.
Collapse
Affiliation(s)
- Jonathan Stone
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - John Mitrofanis
- Discipline of Anatomy and Histology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Daniel M. Johnstone
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Benedetto Falsini
- Facolta’ di Medicina e Chirurgia, Fondazione Policlinico A. Gemelli, Universita’ Cattolica del S. Cuore, Rome, Italy
| | - Silvia Bisti
- Department of Biotechnical and Applied Clinical Sciences, Università degli Studi dell’Aquila, IIT Istituto Italiano di Tecnologia Genova and INBB Istituto Nazionale Biosistemi e Biostrutture, Rome, Italy
| | - Paul Adam
- School of Biological, Earth and Environmental Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Arturo Bravo Nuevo
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Mindy George-Weinstein
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Rebecca Mason
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Janis Eells
- College of Health Sciences, University of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
9
|
Mammone T, Chidlow G, Casson RJ, Wood JPM. Expression and activation of mitogen-activated protein kinases in the optic nerve head in a rat model of ocular hypertension. Mol Cell Neurosci 2018; 88:270-291. [PMID: 29408550 DOI: 10.1016/j.mcn.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 01/03/2018] [Accepted: 01/11/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glaucoma is a leading cause of irreversible blindness manifesting as an age-related, progressive optic neuropathy with associated retinal ganglion cell (RGC) loss. Mitogen-activated protein kinases (MAPKs: p42/44 MAPK, SAPK/JNK, p38 MAPK) are activated in various retinal disease models and likely contribute to the mechanisms of RGC death. Although MAPKs play roles in the development of retinal pathology, their action in the optic nerve head (ONH), where the initial insult to RGC axons likely resides in glaucoma, remains unexplored. METHODS An experimental paradigm representing glaucoma was established by induction of chronic ocular hypertension (OHT) via laser-induced coagulation of the trabecular meshwork in Sprague-Dawley rats. MAPKs were subsequently investigated over the following days for expression and activity alterations, using RT-PCR, immunohistochemistry and Western immunoblot. RESULTS p42/44 MAPK expression was unaltered after intraocular pressure (IOP) elevation, but there was a significant activation of this enzyme in ONH astrocytes after 6-24 h. Activated SAPK/JNK isoforms were present throughout healthy RGC axons but after IOP elevation or optic nerve crush, they both accumulated at the ONH, likely due to RGC axon transport disruption, and were subject to additional activation. p38 MAPK was expressed by a population of microglia which were significantly more populous following IOP elevation. However it was only significantly activated in microglia after 3 days, and then only in the ONH and optic nerve; in the retina it was solely activated in RGC perikarya. CONCLUSIONS In conclusion, each of the MAPKs showed a specific spatio-temporal expression and activation pattern in the retina, ONH and optic nerve as a result of IOP elevation. These findings likely reflect the roles of the individual enzymes, and the cells in which they reside, in the developing pathology following IOP elevation. These data have implications for understanding the mechanisms of ocular pathology in diseases such as glaucoma.
Collapse
Affiliation(s)
- Teresa Mammone
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Robert J Casson
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - John P M Wood
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
10
|
Gidday JM. Adaptive Plasticity in the Retina: Protection Against Acute Injury and Neurodegenerative Disease by Conditioning Stimuli. CONDITIONING MEDICINE 2018; 1:85-97. [PMID: 31423482 PMCID: PMC6696944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Although both preclinical and clinical conditioning studies in heart and brain lead the field of conditioning medicine, investigations of retinal conditioning still number more than 100. In this brief review, we highlight findings to date from animal and cell culture models of conditioning that provide demonstrated protection in acute and chronic retinal injury and disease models. The multitude of stimuli used to condition the retina, the signaling mediators and pathways identified, and the injury- and disease-resilient phenotypes documented are discussed herein, along with our recommendations for the kinds of studies needed to continue to advance this promising field. In our view, the robust protection afforded by these adaptive epigenetic responses to conditioning stress provides significant incentives for both furthering our investment in bench research and underwriting clinical trials, so that the full potential of this therapy can be realized.
Collapse
Affiliation(s)
- Jeffrey M Gidday
- Departments of Ophthalmology, Physiology, and the Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA 70112
| |
Collapse
|
11
|
Iliescu DA, Ciubotaru A, Ghiţă MA, Dumitru A, Zăgrean L. Effect of sevoflurane preconditioning on light-induced retinal damage in diabetic rats. Rom J Ophthalmol 2018; 62:24-33. [PMID: 29796431 PMCID: PMC5959021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2018] [Indexed: 11/29/2022] Open
Abstract
Hyperglycemia and bright light are powerful stress agents that produce an enhanced retinal damage, when simultaneously acting on retina. Previous studies have shown that preconditioning with sevoflurane anesthesia offers a certain degree of protection to retinal cells against light damage. The objective of this study was to explore the effect of sevoflurane anesthetic preconditioning on a model of light-induced retinal degeneration in diabetic rats. Wistar rats that were randomly divided into four groups: control (rats exposed to photostress), group 1 (rats exposed to photostress and sevoflurane preconditioning), group 2 (diabetic rats exposed to photostress), group 3 (diabetic rats exposed to photostress and sevoflurane preconditioning) were used for this experiment. We recorded basal electroretinogram (ERG), at 36 h and 14 days after photostress and performed histological analysis of the retina. Results showed that sevoflurane has a protective effect on light-induced neuroretinal degeneration proved by significantly less variations of the ERG before and after photostress. Diabetes appears to increase the damaging effect of photostress on retina and attenuate the protection provided by sevoflurane preconditioning.
Collapse
Affiliation(s)
- Daniela Adriana Iliescu
- Physiology Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Ophthalmology Department, "Dr. Carol Davila" Central Military University Emergency Hospital, Bucharest, Romania
| | - Alexandra Ciubotaru
- Physiology Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihai Aurelian Ghiţă
- Physiology Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Ophthalmology Department, University Emergency Hospital, Bucharest, Romania
| | - Adrian Dumitru
- Pathology Department, University Emergency Hospital, Bucharest, Romania
| | - Leon Zăgrean
- Physiology Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
12
|
Retinal Pigment Epithelium and Photoreceptor Preconditioning Protection Requires Docosanoid Signaling. Cell Mol Neurobiol 2017; 38:901-917. [PMID: 29177613 PMCID: PMC5882642 DOI: 10.1007/s10571-017-0565-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 11/03/2017] [Indexed: 01/10/2023]
Abstract
Omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) are necessary for functional cell integrity. Preconditioning (PC), as we define it, is an acquired protection or resilience by a cell, tissue, or organ to a lethal stimulus enabled by a previous sublethal stressor or stimulus. In this study, we provide evidence that the omega-3 fatty acid docosahexaenoic acid (DHA) and its derivatives, the docosanoids 17-hydroxy docosahexaenoic acid (17-HDHA) and neuroprotectin D1 (NPD1), facilitate cell survival in both in vitro and in vivo models of retinal PC. We also demonstrate that PC requires the enzyme 15-lipoxygenase-1 (15-LOX-1), which synthesizes 17-HDHA and NPD1, and that this is specific to docosanoid signaling despite the concomitant release of the omega-6 arachidonic acid and eicosanoid synthesis. These findings advocate that DHA and docosanoids are protective enablers of PC in photoreceptor and retinal pigment epithelial cells.
Collapse
|
13
|
Fabiani C, Zulueta A, Bonezzi F, Casas J, Ghidoni R, Signorelli P, Caretti A. 2-Acetyl-5-tetrahydroxybutyl imidazole (THI) protects 661W cells against oxidative stress. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:741-751. [DOI: 10.1007/s00210-017-1374-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
|
14
|
de la Barca JMC, Huang NT, Jiao H, Tessier L, Gadras C, Simard G, Natoli R, Tcherkez G, Reynier P, Valter K. Retinal metabolic events in preconditioning light stress as revealed by wide-spectrum targeted metabolomics. Metabolomics 2017; 13:22. [PMID: 28706468 PMCID: PMC5486622 DOI: 10.1007/s11306-016-1156-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/20/2016] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Light is the primary stimulus for vision, but may also cause damage to the retina. Pre-exposing the retina to sub-lethal amount of light (or preconditioning) improves chances for retinal cells to survive acute damaging light stress. OBJECTIVES This study aims at exploring the changes in retinal metabolome after mild light stress and identifying mechanisms that may be involved in preconditioning. METHODS Retinas from 12 rats exposed to mild light stress (1000 lux × for 12 h) and 12 controls were collected one and seven days after light stress (LS). One retina was used for targeted metabolomics analysis using the Biocrates p180 kit while the fellow retina was used for histological and immunohistochemistry analysis. RESULTS Immunohistochemistry confirmed that in this experiment, a mild LS with retinal immune response and minimal photoreceptor loss occurred. Compared to controls, LS induced an increased concentration in phosphatidylcholines. The concentration in some amino acids and biogenic amines, particularly those related to the nitric oxide pathway (like asymmetric dimethylarginine (ADMA), arginine and citrulline) also increased 1 day after LS. 7 days after LS, the concentration in two sphingomyelins and phenylethylamine was found to be higher. We further found that in controls, retina metabolome was different between males and females: male retinas had an increased concentration in tyrosine, acetyl-ornithine, phosphatidylcholines and (acyl)-carnitines. CONCLUSIONS Besides retinal sexual metabolic dimorphism, this study shows that preconditioning is mostly associated with re-organisation of lipid metabolism and changes in amino acid composition, likely reflecting the involvement of arginine-dependent NO signalling.
Collapse
Affiliation(s)
- Juan Manuel Chao de la Barca
- 0000 0001 2248 3363grid.7252.2PREMMi/Pôle de Recherche et d’Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d’Angers, 49933 Angers, France
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
| | - Nuan-Ting Huang
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
| | - Haihan Jiao
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
| | - Lydie Tessier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Cédric Gadras
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Gilles Simard
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
- 0000 0001 2248 3363grid.7252.2INSERM U1063, Université d’Angers, 49933 Angers, France
| | - Riccardo Natoli
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
- 0000 0001 2180 7477grid.1001.0Medical School, Australian National University, Canberra, ACT 2601 Australia
| | - Guillaume Tcherkez
- 0000 0001 2180 7477grid.1001.0Research School of Biology, College of Medicine, Biology and Environment, Australian National University, Canberra, ACT 2601 Australia
| | - Pascal Reynier
- 0000 0001 2248 3363grid.7252.2PREMMi/Pôle de Recherche et d’Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d’Angers, 49933 Angers, France
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Krisztina Valter
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
- 0000 0001 2180 7477grid.1001.0Medical School, Australian National University, Canberra, ACT 2601 Australia
| |
Collapse
|
15
|
Gu X, Reagan AM, McClellan ME, Elliott MH. Caveolins and caveolae in ocular physiology and pathophysiology. Prog Retin Eye Res 2016; 56:84-106. [PMID: 27664379 DOI: 10.1016/j.preteyeres.2016.09.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/15/2016] [Accepted: 09/20/2016] [Indexed: 12/14/2022]
Abstract
Caveolae are specialized, invaginated plasma membrane domains that are defined morphologically and by the expression of signature proteins called, caveolins. Caveolae and caveolins are abundant in a variety of cell types including vascular endothelium, glia, and fibroblasts where they play critical roles in transcellular transport, endocytosis, mechanotransduction, cell proliferation, membrane lipid homeostasis, and signal transduction. Given these critical cellular functions, it is surprising that ablation of the caveolae organelle does not result in lethality suggesting instead that caveolae and caveolins play modulatory roles in cellular homeostasis. Caveolar components are also expressed in ocular cell types including retinal vascular cells, Müller glia, retinal pigment epithelium (RPE), conventional aqueous humor outflow cells, the corneal epithelium and endothelium, and the lens epithelium. In the eye, studies of caveolae and other membrane microdomains (i.e., "lipid rafts") have lagged behind what is a substantial body of literature outside vision science. However, interest in caveolae and their molecular components has increased with accumulating evidence of important roles in vision-related functions such as blood-retinal barrier homeostasis, ocular inflammatory signaling, pathogen entry at the ocular surface, and aqueous humor drainage. The recent association of CAV1/2 gene loci with primary open angle glaucoma and intraocular pressure has further enhanced the need to better understand caveolar functions in the context of ocular physiology and disease. Herein, we provide the first comprehensive review of literature on caveolae, caveolins, and other membrane domains in the context of visual system function. This review highlights the importance of caveolae domains and their components in ocular physiology and pathophysiology and emphasizes the need to better understand these important modulators of cellular function.
Collapse
Affiliation(s)
- Xiaowu Gu
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alaina M Reagan
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mark E McClellan
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael H Elliott
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
16
|
Kong L, Liu B, Zhang C, Wang B, Wang H, Song X, Yang Y, Ren X, Yin L, Kong H, Ma H. The therapeutic potential of sulforaphane on light-induced photoreceptor degeneration through antiapoptosis and antioxidant protection. Neurochem Int 2016; 100:52-61. [PMID: 27567738 DOI: 10.1016/j.neuint.2016.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 08/18/2016] [Accepted: 08/23/2016] [Indexed: 01/26/2023]
Abstract
Oxidative stress due to excessive light exposure can exacerbate a variety of human retinal diseases by accelerating photoreceptor cell death. The thioredoxin (Trx) system is considered to play a crucial role in reduction/oxidation (redox) regulation of signal transduction and in cell defense against oxidative stresses. Sulforaphane (SF) protects cells from oxidative damage through nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which is responsible for multiple detoxification processes, including elevating the expression of Trx. This study sought to demonstrate whether SF increased Trx expression in retinal tissues in vivo and whether it could preserve the photoreceptors from degeneration induced by oxidative stress. Our data clearly showed that pretreatment with SF abated photoreceptor cell loss, in association with increased expression of Nrf2 and Trx, subsequently activating the Ras/Raf1/Erk signaling pathway and decreasing the expression of Bak1, Cyt-c release and the activity of caspase-3 in light-induced mouse retinas. These data suggested that the therapeutic potential of SF in retinal degeneration due to oxidative stress might partially involve anti-caspase and antioxidant protection mediated by Trx.
Collapse
Affiliation(s)
- Li Kong
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Bo Liu
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Chenghong Zhang
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Bing Wang
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Hongfei Wang
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Xiaoxia Song
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Yang Yang
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Xiang Ren
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China
| | - Liangwei Yin
- Department of Oncology, Dalian Central Hospital, 116033, Dalian, Liaoning, China
| | - Hui Kong
- Department of Otorhinolaryngology, The Second Hospital of Dalian Medical University, 116023, Dalian, Liaoning, China.
| | - Haiying Ma
- Department of Histoembryology of Dalian Medical University, 116044, Dalian, Liaoning, China.
| |
Collapse
|
17
|
Krishnamoorthy V, Cherukuri P, Poria D, Goel M, Dagar S, Dhingra NK. Retinal Remodeling: Concerns, Emerging Remedies and Future Prospects. Front Cell Neurosci 2016; 10:38. [PMID: 26924962 PMCID: PMC4756099 DOI: 10.3389/fncel.2016.00038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/01/2016] [Indexed: 12/21/2022] Open
Abstract
Deafferentation results not only in sensory loss, but also in a variety of alterations in the postsynaptic circuitry. These alterations may have detrimental impact on potential treatment strategies. Progressive loss of photoreceptors in retinal degenerative diseases, such as retinitis pigmentosa and age-related macular degeneration, leads to several changes in the remnant retinal circuitry. Müller glial cells undergo hypertrophy and form a glial seal. The second- and third-order retinal neurons undergo morphological, biochemical and physiological alterations. A result of these alterations is that retinal ganglion cells (RGCs), the output neurons of the retina, become hyperactive and exhibit spontaneous, oscillatory bursts of spikes. This aberrant electrical activity degrades the signal-to-noise ratio in RGC responses, and thus the quality of information they transmit to the brain. These changes in the remnant retina, collectively termed “retinal remodeling”, pose challenges for genetic, cellular and bionic approaches to restore vision. It is therefore crucial to understand the nature of retinal remodeling, how it affects the ability of remnant retina to respond to novel therapeutic strategies, and how to ameliorate its effects. In this article, we discuss these topics, and suggest that the pathological state of the retinal output following photoreceptor loss is reversible, and therefore, amenable to restorative strategies.
Collapse
Affiliation(s)
| | - Pitchaiah Cherukuri
- Developmental Neurobiology Laboratory, European Neuroscience Institute Göttingen Göttingen, Germany
| | - Deepak Poria
- National Brain Research Centre Manesar, Haryana, India
| | - Manvi Goel
- National Brain Research Centre Manesar, Haryana, India
| | - Sushma Dagar
- Institute of Neuro- and Sensory Physiology, Heinrich-Heine University Düsseldorf, Germany
| | | |
Collapse
|
18
|
Retinal Cell Degeneration in Animal Models. Int J Mol Sci 2016; 17:ijms17010110. [PMID: 26784179 PMCID: PMC4730351 DOI: 10.3390/ijms17010110] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/25/2015] [Accepted: 01/08/2016] [Indexed: 01/01/2023] Open
Abstract
The aim of this review is to provide an overview of various retinal cell degeneration models in animal induced by chemicals (N-methyl-d-aspartate- and CoCl2-induced), autoimmune (experimental autoimmune encephalomyelitis), mechanical stress (optic nerve crush-induced, light-induced) and ischemia (transient retinal ischemia-induced). The target regions, pathology and proposed mechanism of each model are described in a comparative fashion. Animal models of retinal cell degeneration provide insight into the underlying mechanisms of the disease, and will facilitate the development of novel effective therapeutic drugs to treat retinal cell damage.
Collapse
|
19
|
Song D, Wilson B, Zhao L, Bhuyan R, Bandyopadhyay M, Lyubarsky A, Yu C, Li Y, Kanu L, Miwa T, Song WC, Finnemann SC, Rohrer B, Dunaief JL. Retinal Pre-Conditioning by CD59a Knockout Protects against Light-Induced Photoreceptor Degeneration. PLoS One 2016; 11:e0166348. [PMID: 27893831 PMCID: PMC5125596 DOI: 10.1371/journal.pone.0166348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/27/2016] [Indexed: 11/25/2022] Open
Abstract
Complement dysregulation plays a key role in the pathogenesis of age-related macular degeneration (AMD), but the specific mechanisms are incompletely understood. Complement also potentiates retinal degeneration in the murine light damage model. To test the retinal function of CD59a, a complement inhibitor, CD59a knockout (KO) mice were used for light damage (LD) experiments. Retinal degeneration and function were compared in WT versus KO mice following light damage. Gene expression changes, endoplasmic reticulum (ER) stress, and glial cell activation were also compared. At baseline, the ERG responses and rhodopsin levels were lower in CD59aKO compared to wild-type (WT) mice. Following LD, the ERG responses were better preserved in CD59aKO compared to WT mice. Correspondingly, the number of photoreceptors was higher in CD59aKO retinas than WT controls after LD. Under normal light conditions, CD59aKO mice had higher levels than WT for GFAP immunostaining in Müller cells, mRNA and protein levels of two ER-stress markers, and neurotrophic factors. The reduction in photon capture, together with the neurotrophic factor upregulation, may explain the structural and functional protection against LD in the CD59aKO.
Collapse
Affiliation(s)
- Delu Song
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Brooks Wilson
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC
| | - Liangliang Zhao
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin, China
| | - Rupak Bhuyan
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | | | - Arkady Lyubarsky
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Chen Yu
- Center for Cancer, Genetic Diseases, and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Yafeng Li
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Levi Kanu
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Takashi Miwa
- Department of Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Wen-Chao Song
- Department of Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases, and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC
- * E-mail: (JLD); (BR)
| | - Joshua L. Dunaief
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- * E-mail: (JLD); (BR)
| |
Collapse
|
20
|
Levkovitch-Verbin H. Retinal ganglion cell apoptotic pathway in glaucoma: Initiating and downstream mechanisms. PROGRESS IN BRAIN RESEARCH 2015; 220:37-57. [PMID: 26497784 DOI: 10.1016/bs.pbr.2015.05.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Apoptosis of retinal ganglion cells (RGCs) in glaucoma causes progressive visual field loss, making it the primary cause of irreversible blindness worldwide. Elevated intraocular pressure and aging, the main risk factors for glaucoma, accelerate RGC apoptosis. Numerous pathways and mechanisms were found to be involved in RGC death in glaucoma. Neurotrophic factors deprivation is an early event. Oxidative stress, mitochondrial dysfunction, inflammation, glial cell dysfunction, and activation of apoptotic pathways and prosurvival pathways play a significant role in RGC death in glaucoma. The most important among the involved pathways are the MAP-kinase pathway, PI-3 kinase/Akt pathway, Bcl-2 family, caspase family, and IAP family.
Collapse
Affiliation(s)
- Hani Levkovitch-Verbin
- Glaucoma Service, Goldschleger Eye Institute, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.
| |
Collapse
|
21
|
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: 2.2] [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
|
22
|
|
23
|
Mueller BH, Park Y, Ma HY, Dibas A, Ellis DZ, Clark AF, Yorio T. Sigma-1 receptor stimulation protects retinal ganglion cells from ischemia-like insult through the activation of extracellular-signal-regulated kinases 1/2. Exp Eye Res 2014; 128:156-69. [PMID: 25305575 DOI: 10.1016/j.exer.2014.10.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/18/2014] [Accepted: 10/07/2014] [Indexed: 11/20/2022]
Abstract
Sigma-1 receptor (σ-1) activation and mitogen-activated protein kinases (MAPKs) have been shown to protect retinal ganglion cells (RGCs) from cell death. The purpose of this study was to determine if σ-1 receptor stimulation with pentazocine could promote neuroprotection under conditions of an ischemia-like insult (oxygen glucose deprivation (OGD)) through the phosphorylation of extracellular signal regulated kinase (pERK)1/2. Primary RGCs were isolated from P3-P7 Sprague-Dawley rats and purified by sequential immunopanning using Thy1.1 antibodies. RGCs were cultured for 7 days before subjecting the cells to an OGD insult (0.5% oxygen in glucose-free medium) for 6 h. During the OGD, RGCs were treated with pentazocine (σ-1 receptor agonist) with or without BD 1047 (σ-1 receptor antagonist). In other experiments, primary RGCs were treated with pentazocine in the presence or absence of an MEK1/2 inhibitor, PD098059. Cell survival/death was assessed by staining with the calcein-AM/ethidium homodimer reagent. Levels of pERK1/2, total ERK1/2, and beta tubulin expression were determined by immunoblotting and immunofluorescence staining. RGCs subjected to OGD for 6 h induced 50% cell death in primary RGCs (p < 0.001) and inhibited pERK1/2 expression by 65% (p < 0.001). Cell death was attenuated when RGCs were treated with pentazocine under OGD (p < 0.001) and pERK1/2 expression was increased by 1.6 fold (p < 0.05) compared to OGD treated RGCs without pentazocine treatment. The co-treatment of PD098059 (MEK1/2 inhibitor) with pentazocine significantly abolished the protective effects of pentazocine on the RGCs during this OGD insult. Activation of the σ-1 receptor is a neuroprotective target that can protect RGCs from an ischemia-like insult. These results also established a direct relationship between σ-1 receptor stimulation and the neuroprotective effects of the ERK1/2 pathway in purified RGCs subjected to OGD. These findings suggest that activation of the σ-1 receptor may be a therapeutic target for neuroprotection particularly relevant to ocular neurodegenerative diseases that effect RGCs.
Collapse
Affiliation(s)
- Brett H Mueller
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | - Yong Park
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Hai-Ying Ma
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Adnan Dibas
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Dorette Z Ellis
- College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Abbot F Clark
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Thomas Yorio
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
24
|
Mathews MK, Guo Y, Langenberg P, Bernstein SL. Ciliary neurotrophic factor (CNTF)-mediated ganglion cell survival in a rodent model of non-arteritic anterior ischaemic optic neuropathy (NAION). Br J Ophthalmol 2014; 99:133-7. [PMID: 25336580 DOI: 10.1136/bjophthalmol-2014-305969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Ciliary neurotrophic factor (CNTF) has been shown to protect retinal ganglion cells (RGCs) in traumatic optic nerve injury. We sought to evaluate this neuroprotective effect of CNTF after an ischaemic event using rodent anterior ischaemic optic neuropathy (rAION), a mouse model of non-arteritic anterior ischaemic optic neuropathy (NAION). METHODS We induced rAION in Thy1-cyan fluorescent protein (CFP) transgenic mice by exposing the optic nerve to frequency doubled neodymium yttrium aluminium garnet laser pulses following intravenous rose bengal injection. One day after rAION induction, an intravitreal injection of 0.75 μg CNTF or vehicle (sham injection) was given. Animals were euthanised on day 15 after induction, tissues isolated and CFP cells in the RGC layer were counted using stereology in flat-mounted retina. The average number of CFP-positive (CFP+) cells was determined for each study group and the percentages of RGC loss were compared between the different groups. RESULTS Two weeks after rAION induction, significantly more (CFP+) cells were preserved in CNTF-treated eyes than in sham-injected controls. Sham-treated animals showed a 58% loss of CFP+ cells. In contrast, CFP+ cell density in CNTF-treated eyes decreased by only 10%, when compared with untreated control eyes. This increased survival was statistically significant (p<0.05). CONCLUSIONS CNTF exerts a neuroprotective effect in ischaemic optic nerve injury and promotes RGC survival, suggesting that CNTF may be effective in the clinical treatment of human NAION.
Collapse
Affiliation(s)
- Michaela K Mathews
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yan Guo
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Patricia Langenberg
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Steven L Bernstein
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Effects of a conventional photocoagulator and a 3-ns pulse laser on preconditioning responses and retinal ganglion cell survival after optic nerve crush. Exp Eye Res 2014; 127:77-90. [DOI: 10.1016/j.exer.2014.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/07/2014] [Accepted: 07/10/2014] [Indexed: 11/22/2022]
|
26
|
Kucharska J, Del Río P, Arango-Gonzalez B, Gorza M, Feuchtinger A, Hauck SM, Ueffing M. Cyr61 activates retinal cells and prolongs photoreceptor survival in rd1 mouse model of retinitis pigmentosa. J Neurochem 2014; 130:227-40. [PMID: 24593181 DOI: 10.1111/jnc.12704] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/27/2014] [Accepted: 02/27/2014] [Indexed: 01/08/2023]
Abstract
Subretinal injections with glial cell line-derived neurotrophic factor (GDNF) rescue morphology as well as function of rod cells in mouse and rat animal models of retinitis pigmentosa. At the same time, it is postulated that this effect is indirect, mediated by activation of retinal Müller glial (RMG) cells. Here, we show that Cyr61/CCN1, one of the secreted proteins up-regulated in primary RMG after glial cell line-derived neurotrophic factor stimulation, provides neuroprotective and pro-survival capacities: Recombinant Cyr61 significantly reduced photoreceptor (PR) cells death in organotypic cultures of Pde6b(rd1) retinas. To identify stimulated pathways in the retina, we treated Pde6b(rd1) retinal explants with Cyr61 and observed an overall increase in activated Erk1/2 and Stat3 signalling molecules characterized by activation-site-specific phosphorylation. To identify Cyr61 retinal target cells, we isolated primary porcine PR, RMG and retinal pigment epithelium (RPE) cells and exposed them separately to Cyr61. Here, RMG as well as RPE cells responded with induced phosphorylation of Erk1/2, Stat3 and Akt. In PR, no increase in phosphorylation in any of the studied proteins was detected, suggesting an indirect neuroprotective effect of Cyr61. Cyr61 may thus act as an endogenous pro-survival factor for PR, contributing to the complex repertoire of neuroprotective activities generated by RMG and RPE cells. We propose the following model of Cyr61 neuroprotection within the retina: Cyr61 stimulates retinal Müller glial (RMG) and retinal pigment epithelium (RPE) cells and activates PI3K/Akt, mitogen-activated protein kinase(MAPK)/Erk and Janus kinase(JAK)/Stat-signalling pathways in these cells. Phosphorylated Stat3 and Erk1/2 presumably translocate to the nucleus, induce transcriptional changes, which increase secretion of neuroprotective agents that protect photoreceptors (PR) from mutation-induced death.
Collapse
Affiliation(s)
- Joanna Kucharska
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Research Unit Protein Science, Helmholtz Zentrum München, Neuherberg, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Trophic factors in the pathogenesis and therapy for retinal degenerative diseases. Surv Ophthalmol 2014; 59:134-65. [PMID: 24417953 DOI: 10.1016/j.survophthal.2013.09.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 09/11/2013] [Accepted: 09/17/2013] [Indexed: 12/27/2022]
Abstract
Trophic factors are endogenously secreted proteins that act in an autocrine and/or paracrine fashion to affect vital cellular processes such as proliferation, differentiation, and regeneration, thereby maintaining overall cell homeostasis. In the eye, the major contributors of these molecules are the retinal pigment epithelial (RPE) and Müller cells. The primary paracrine targets of these secreted proteins include the photoreceptors and choriocapillaris. Retinal degenerative diseases such as age-related macular degeneration and retinitis pigmentosa are characterized by aberrant function and/or eventual death of RPE cells, photoreceptors, choriocapillaris, and other retinal cells. We discuss results of in vitro and in vivo animal studies in which candidate trophic factors, either singly or in combination, were used in an attempt to ameliorate photoreceptor and/or retinal degeneration. We also examine current trophic factor therapies as they relate to the treatment of retinal degenerative diseases in clinical studies.
Collapse
|
28
|
Marco FD, Romeo S, Nandasena C, Purushothuman S, Adams C, Bisti S, Stone J. The time course of action of two neuroprotectants, dietary saffron and photobiomodulation, assessed in the rat retina. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:208-20. [PMID: 24093084 PMCID: PMC3783833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/05/2013] [Indexed: 06/02/2023]
Abstract
BACKGROUND Dietary saffron and photobiomodulation (low-level infrared radiation, PBM) are emerging as therapeutically promising protectants for neurodegenerative conditions, such as the retinal dystrophies. In animal models, saffron and PBM, given in limited daily doses, protect retina and brain from toxin- or light-induced stress. This study addresses the rate at which saffron and PBM, given in daily doses, induce neuroprotection, using a light damage model of photoreceptor degeneration in Sprague Dawley (SD) rats. RESULTS Rats were raised in dim cyclic (12 h 5 lux, 12 h dark) illumination, treated with saffron or PBM for 2-10 d, and then exposed to bright damaging light (1,000 lux for 24 h). After 1 week survival, the retina was assessed for photoreceptor death (using the TUNEL reaction), for surviving photoreceptor damage (thickness of the outer nuclear layer) and for the expression of a stress-related protein GFAP, using immunohistochemistry. Preconditioning the retina with saffron or PBM reduced photoreceptor death, preserved the population of surviving photoreceptors and reduced the upregulation of GFAP in Müller cells. At the daily dose of saffron used (1 mg/kg), protection was detectable at 2 d, increasing to 10 d. At the daily dose of PBM used (5 J/cm(2) at 670 nm) protection was detectable at 5 d, increasing to 7-10 d. CONCLUSIONS The results provide time parameters for exploration of the mechanisms and durability of the protection provided by saffron and PBM.
Collapse
Affiliation(s)
- Fabiana Di Marco
- Department of Biotechnology and Applied Clinical Science, University of L’AquilaItaly
| | - Stefania Romeo
- Department of Biotechnology and Applied Clinical Science, University of L’AquilaItaly
| | - Charith Nandasena
- Discipline of Physiology and Bosch Institute, University of Sydney and ARC Centre of Excellence in Vision Science, University of SydneyAustralia
| | - Sivaraman Purushothuman
- Discipline of Physiology and Bosch Institute, University of Sydney and ARC Centre of Excellence in Vision Science, University of SydneyAustralia
| | - Charean Adams
- Discipline of Physiology and Bosch Institute, University of Sydney and ARC Centre of Excellence in Vision Science, University of SydneyAustralia
| | - Silvia Bisti
- Department of Biotechnology and Applied Clinical Science, University of L’AquilaItaly
| | - Jonathan Stone
- Discipline of Physiology and Bosch Institute, University of Sydney and ARC Centre of Excellence in Vision Science, University of SydneyAustralia
| |
Collapse
|
29
|
Basic fibroblast growth factor contributes to a shift in the angioregulatory activity of retinal glial (Müller) cells. PLoS One 2013; 8:e68773. [PMID: 23861940 PMCID: PMC3701643 DOI: 10.1371/journal.pone.0068773] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Accepted: 06/05/2013] [Indexed: 01/13/2023] Open
Abstract
Basic fibroblast growth factor (bFGF) is a pleiotropic cytokine with pro-angiogenic and neurotrophic effects. The angioregulatory role of this molecule may become especially significant in retinal neovascularization, which is a hallmark of a number of ischemic eye diseases. This study was undertaken to reveal expression characteristics of bFGF, produced by retinal glial (Müller) cells, and to determine conditions under which glial bFGF may stimulate the proliferation of retinal microvascular endothelial cells. Immunofluorescence labeling detected bFGF in Müller cells of the rat retina and in acutely isolated Müller cells with bFGF levels, which increased after ischemia-reperfusion in postischemic retinas. In patients with proliferative diabetic retinopathy or myopia, the immunoreactivity of bFGF co-localized to glial fibrillary acidic protein (GFAP)-positive cells in surgically excised retinal tissues. RT-PCR and ELISA analyses indicated that cultured Müller cells produce bFGF, which is elevated under hypoxia or oxidative stress, as well as under stimulation with various growth factors and cytokines, including pro-inflammatory factors. When retinal endothelial cells were cultured in the presence of media from hypoxia (0.2%)-conditioned Müller cells, a distinct picture of endothelial cell proliferation emerged. Media from 24-h cultured Müller cells inhibited proliferation, whereas 72-h conditioned media elicited a stimulatory effect. BFGF-neutralizing antibodies suppressed the enhanced endothelial cell proliferation to a similar extent as anti-VEGF antibodies. Furthermore, phosphorylation of extracellular signal-regulated kinases (ERK−1/−2) in retinal endothelial cells was increased when the cells were cultured in 72-h conditioned media, while neutralizing bFGF attenuated the activation of this signaling pathway. These data provide evidence that retinal (glial) Müller cells are major sources of bFGF in the ischemic retina. Müller cells under physiological conditions or transient hypoxia seem to provide an anti-angiogenic environment, but long-lasting hypoxia causes the release of bFGF, which might significantly co-stimulate neovascularization in the retina.
Collapse
|
30
|
Dy GK, Adjei AA. Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA Cancer J Clin 2013; 63:249-79. [PMID: 23716430 DOI: 10.3322/caac.21184] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/21/2013] [Accepted: 02/25/2013] [Indexed: 12/11/2022] Open
Abstract
Answer questions and earn CME/CNE Advances in genomics and molecular biology have identified aberrant proteins in cancer cells that are attractive targets for cancer therapy. Because these proteins are overexpressed or dysregulated in cancer cells compared with normal cells, it was assumed that their inhibitors will be narrowly targeted and relatively nontoxic. However, this hope has not been achieved. Current targeted agents exhibit the same frequency and severity of toxicities as traditional cytotoxic agents, with the main difference being the nature of the toxic effects. Thus, the classical chemotherapy toxicities of alopecia, myelosuppression, mucositis, nausea, and vomiting have been generally replaced by vascular, dermatologic, endocrine, coagulation, immunologic, ocular, and pulmonary toxicities. These toxicities need to be recognized, prevented, and optimally managed.
Collapse
Affiliation(s)
- Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | | |
Collapse
|
31
|
Reichenbach A, Bringmann A. Cell Biology of the Müller Cell. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00017-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
32
|
Bai S, Sheline CR, Zhou Y, Sheline CT. A reduced zinc diet or zinc transporter 3 knockout attenuate light induced zinc accumulation and retinal degeneration. Exp Eye Res 2012; 108:59-67. [PMID: 23274584 DOI: 10.1016/j.exer.2012.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/20/2012] [Accepted: 12/12/2012] [Indexed: 01/05/2023]
Abstract
Our previous study on retinal light exposure suggests the involvement of zinc (Zn(2+)) toxicity in the death of RPE and photoreceptors (LD) which could be attenuated by pyruvate and nicotinamide, perhaps through restoration of NAD(+) levels. In the present study, we examined Zn(2+) toxicity, and the effects of NAD(+) restoration in primary retinal cultures. We then reduced Zn(2+) levels in rodents by reducing Zn(2+) levels in the diet, or by genetics and measured LD. Sprague Dawley albino rats were fed 2, or 61 mg Zn(2+)/kg of diet for 3 weeks, and exposed to 18 kLux of white light for 4 h. We light exposed (70 kLux of white light for 50 h) Zn(2+) transporter 3 knockout (ZnT3-KO, no synaptic Zn(2+)), or RPE65 knockout mice (RPE65-KO, lack rhodopsin cycling), or C57/BI6/J controls and determined light damage and Zn(2+) staining. Retinal Zn(2+) staining was examined at 1 h and 4 h after light exposure. Retinas were examined after 7 d by optical coherence tomography and histology. After LD, rats fed the reduced Zn(2+) diet showed less photoreceptor Zn(2+) staining and degeneration compared to a normal Zn(2+) diet. Similarly, ZnT3-KO and RPE65-KO mice showed less Zn(2+) staining, NAD(+) loss, and RPE or photoreceptor death than C57/BI6/J control mice. Dietary or ZnT3-dependent Zn(2+) stores, and intracellular Zn(2+) release from rhodopsin recycling are suggested to be involved in light-induced retinal degeneration. These results implicate novel rhodopsin-mediated mechanisms and therapeutic targets for LD. Our companion manuscript demonstrates that pharmacologic, circadian, or genetic manipulations which maintain NAD(+) levels reduce LD.
Collapse
Affiliation(s)
- Shi Bai
- Department of Ophthalmology and The Neuroscience Center of Excellence, LSU Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
33
|
Chucair-Elliott AJ, Elliott MH, Wang J, Moiseyev GP, Ma JX, Politi LE, Rotstein NP, Akira S, Uematsu S, Ash JD. Leukemia inhibitory factor coordinates the down-regulation of the visual cycle in the retina and retinal-pigmented epithelium. J Biol Chem 2012; 287:24092-102. [PMID: 22645143 DOI: 10.1074/jbc.m112.378240] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukemia inhibitory factor (LIF), an interleukin-6 family neurocytokine, is up-regulated in response to different types of retinal stress and has neuroprotective activity through activation of the gp130 receptor/STAT3 pathway. We observed that LIF induces rapid, robust, and sustained activation of STAT3 in both the retina and retinal pigmented epithelium (RPE). Here, we tested whether LIF-induced STAT3 activation within the RPE can down-regulate RPE65, the central enzyme in the visual cycle that provides the 11-cis-retinal chromophore to photoreceptors in vivo. We generated conditional knock-out mice to specifically delete STAT3 or gp130 in RPE, retina, or both RPE and retina. After intravitreal injection of LIF, we analyzed the expression levels of visual cycle genes and proteins, isomerase activity of RPE65, levels of rhodopsin protein, and the rates of dark adaptation and rhodopsin regeneration. We found that RPE65 protein levels and isomerase activity were reduced and recovery of bleachable rhodopsin was delayed in LIF-injected eyes. In mice with functional gp130/STAT3 signaling in the retina, rhodopsin protein was also reduced by LIF. However, the LIF-induced down-regulation of RPE65 required a functional gp130/STAT3 cascade intrinsic to RPE. Our data demonstrate that a single cytokine, LIF, can simultaneously and independently affect both RPE and photoreceptors through the same signaling cascade to reduce the generation and utilization of 11-cis-retinal.
Collapse
Affiliation(s)
- Ana J Chucair-Elliott
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vander S, Levkovitch-Verbin H. Regulation of cell death and survival pathways in secondary degeneration of the optic nerve - a long-term study. Curr Eye Res 2012; 37:740-8. [PMID: 22631427 DOI: 10.3109/02713683.2012.673679] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To investigate cell death and survival pathways in secondary degeneration of the optic nerve (ON) and retina over a period of 6 months. METHODS A partial transection model of the ON that morphologically separates primary and secondary degeneration was applied unilaterally in 89 Wistar rat eyes. The upper third of the retinas were analyzed for primary degeneration, while the lower third of the retinas were analyzed for secondary degeneration. The involvement of members of the mitogen-activated protein (MAP) kinase pathway and the PI-3-kinase/Akt pathway were evaluated in primary and secondary degeneration in multiple time points over a period of 6 months using immunohistochemistry and western blotting. Results were compared to corresponding areas from control fellow eyes. RESULTS All investigated members of the MAP kinase pathway were significantly activated in primary degeneration, secondary degeneration or both. P-SAPK/JNK and P-ERK were activated in primary degeneration without a concomitant activation in secondary degeneration. The prosurvival protein p-Akt, a member of the PI-3-kinase survival pathway, was significantly activated in secondary but not in primary degeneration. P-c-jun and p-ATF were significantly activated in both primary and secondary degeneration. The time-dependent pattern of activation was different for each protein and in secondary degeneration the activation of these proteins was usually short termed. CONCLUSIONS The significant involvement of the MAP kinase pathway and the PI-3-kinase survival pathway in secondary degeneration of the ON and retina is short termed despite continuous retinal ganglion cells (RGCs)apoptosis for at least 6 months.
Collapse
Affiliation(s)
- Shelly Vander
- Sam Rothberg Ophthalmic Molecular Biology Laboratory, Goldschleger Eye Institute, Sheba Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel
| | | |
Collapse
|
35
|
Bordone MP, Lanzani MF, López-Costa JJ, Chianelli MS, Franco P, Sáenz DA, Rosenstein RE. Bacterial lipopolysaccharide protects the retina from light-induced damage. J Neurochem 2012; 122:392-403. [DOI: 10.1111/j.1471-4159.2012.07767.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Yi H, Patel AK, Sodhi CP, Hackam DJ, Hackam AS. Novel role for the innate immune receptor Toll-like receptor 4 (TLR4) in the regulation of the Wnt signaling pathway and photoreceptor apoptosis. PLoS One 2012; 7:e36560. [PMID: 22615780 PMCID: PMC3355158 DOI: 10.1371/journal.pone.0036560] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 04/07/2012] [Indexed: 01/01/2023] Open
Abstract
Recent evidence has implicated innate immunity in regulating neuronal survival in the brain during stroke and other neurodegenerations. Photoreceptors are specialized light-detecting neurons in the retina that are essential for vision. In this study, we investigated the role of the innate immunity receptor TLR4 in photoreceptors. TLR4 activation by lipopolysaccharide (LPS) significantly reduced the survival of cultured mouse photoreceptors exposed to oxidative stress. With respect to mechanism, TLR4 suppressed Wnt signaling, decreased phosphorylation and activation of the Wnt receptor LRP6, and blocked the protective effect of the Wnt3a ligand. Paradoxically, TLR4 activation prior to oxidative injury protected photoreceptors, in a phenomenon known as preconditioning. Expression of TNFα and its receptors TNFR1 and TNFR2 decreased during preconditioning, and preconditioning was mimicked by TNFα antagonists, but was independent of Wnt signaling. Therefore, TLR4 is a novel regulator of photoreceptor survival that acts through the Wnt and TNFα pathways.
Collapse
Affiliation(s)
- Hyun Yi
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Amit K. Patel
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Chhinder P. Sodhi
- Department of Surgery (Pediatric), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - David J. Hackam
- Department of Surgery (Pediatric), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Abigail S. Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
37
|
Albarracin R, Valter K. 670 nm Red Light Preconditioning Supports Müller Cell Function: Evidence from the White Light-induced Damage Model in the Rat Retina†. Photochem Photobiol 2012; 88:1418-27. [DOI: 10.1111/j.1751-1097.2012.01130.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
38
|
Thomas JL, Nelson CM, Luo X, Hyde DR, Thummel R. Characterization of multiple light damage paradigms reveals regional differences in photoreceptor loss. Exp Eye Res 2012; 97:105-16. [PMID: 22425727 DOI: 10.1016/j.exer.2012.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 11/16/2022]
Abstract
Zebrafish provide an attractive model to study the retinal response to photoreceptor apoptosis due to its remarkable ability to spontaneously regenerate retinal neurons following damage. There are currently two widely-used light-induced retinal degeneration models to damage photoreceptors in the adult zebrafish. One model uses constant bright light, whereas the other uses a short exposure to extremely intense ultraviolet light. Although both models are currently used, it is unclear whether they differ in regard to the extent of photoreceptor damage or the subsequent regeneration response. Here we report a thorough analysis of the photoreceptor damage and subsequent proliferation response elicited by each individual treatment, as well as by the concomitant use of both treatments. We show a differential loss of rod and cone photoreceptors with each treatment. Additionally, we show that the extent of proliferation observed in the retina directly correlates with the severity of photoreceptor loss. We also demonstrate that both the ventral and posterior regions of the retina are partially protected from light damage. Finally, we show that combining a short ultraviolet exposure followed by a constant bright light treatment largely eliminates the neuroprotected regions, resulting in widespread loss of rod and cone photoreceptors and a robust regenerative response throughout the retina.
Collapse
Affiliation(s)
- Jennifer L Thomas
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 501 E. Canfield Ave., Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
39
|
Hunter JJ, Morgan JIW, Merigan WH, Sliney DH, Sparrow JR, Williams DR. The susceptibility of the retina to photochemical damage from visible light. Prog Retin Eye Res 2012; 31:28-42. [PMID: 22085795 PMCID: PMC3242847 DOI: 10.1016/j.preteyeres.2011.11.001] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 10/28/2011] [Accepted: 11/01/2011] [Indexed: 02/08/2023]
Abstract
The photoreceptor/RPE complex must maintain a delicate balance between maximizing the absorption of photons for vision and retinal image quality while simultaneously minimizing the risk of photodamage when exposed to bright light. We review the recent discovery of two new effects of light exposure on the photoreceptor/RPE complex in the context of current thinking about the causes of retinal phototoxicity. These effects are autofluorescence photobleaching in which exposure to bright light reduces lipofuscin autofluorescence and, at higher light levels, RPE disruption in which the pattern of autofluorescence is permanently altered following light exposure. Both effects occur following exposure to visible light at irradiances that were previously thought to be safe. Photopigment, retinoids involved in the visual cycle, and bisretinoids in lipofuscin have been implicated as possible photosensitizers for photochemical damage. The mechanism of RPE disruption may follow either of these paths. On the other hand, autofluorescence photobleaching is likely an indicator of photooxidation of lipofuscin. The permanent changes inherent in RPE disruption might require modification of the light safety standards. AF photobleaching recovers after several hours although the mechanisms by which this occurs are not yet clear. Understanding the mechanisms of phototoxicity is all the more important given the potential for increased susceptibility in the presence of ocular diseases that affect either the visual cycle and/or lipofuscin accumulation. In addition, knowledge of photochemical mechanisms can improve our understanding of some disease processes that may be influenced by light exposure, such as some forms of Leber's congenital amaurosis, and aid in the development of new therapies. Such treatment prior to intentional light exposures, as in ophthalmic examinations or surgeries, could provide an effective preventative strategy.
Collapse
Affiliation(s)
- Jennifer J Hunter
- Flaum Eye Institute, University of Rochester, Box 314, 601Elmwood Ave, Rochester, NY 14642, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Neurons of the central nervous system (CNS) of adult mammals can be damaged in a variety of ways. Most neurons rapidly die after injury. Even if the injured CNS neurons do not die in a short time, the neurons eventually die because they are not able to regenerate their axons to reconnect with their normal targets. In addition, neurons are normally not replaced. Therefore, much work has been directed toward understanding of the molecular regulation of the CNS degeneration following injury, and different experimental strategies are being used to try to protect the damaged neurons. Following axonal lesion, the neurons not only need to survive but also to reconnect to be functionally relevant, and efforts are directed toward not only survival but also axonal regeneration and proper rewiring of injured neurons. Recent experimental data suggest that electrical activity, endogenous or exogenous, can enhance neuronal survival and regeneration in vitro and in vivo. This chapter reviews the evidence that have been obtained on the role of neuronal electrical activity on neuroprotection. We will develop perspectives toward neuroprotection and regeneration of adult lesioned CNS neurons based on electrical activity-dependent cell survival that may be applicable to various diseases of the CNS.
Collapse
|
41
|
Caprara C, Grimm C. From oxygen to erythropoietin: relevance of hypoxia for retinal development, health and disease. Prog Retin Eye Res 2011; 31:89-119. [PMID: 22108059 DOI: 10.1016/j.preteyeres.2011.11.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/01/2011] [Accepted: 11/07/2011] [Indexed: 12/20/2022]
Abstract
Photoreceptors and other cells of the retina consume large quantities of energy to efficiently convert light information into a neuronal signal understandable by the brain. The necessary energy is mainly provided by the oxygen-dependent generation of ATP in the numerous mitochondria of retinal cells. To secure the availability of sufficient oxygen for this process, the retina requires constant blood flow through the vasculature of the retina and the choroid. Inefficient supply of oxygen and nutrients, as it may occur in conditions of disturbed hemodynamics or vascular defects, results in tissue ischemia or hypoxia. This has profound consequences on retinal function and cell survival, requiring an adaptational response by cells to cope with the reduced oxygen tension. Central to this response are hypoxia inducible factors, transcription factors that accumulate under hypoxic conditions and drive the expression of a large variety of target genes involved in angiogenesis, cell survival and metabolism. Prominent among these factors are vascular endothelial growth factor and erythropoietin, which may contribute to normal angiogenesis during development, but may also cause neovascularization and vascular leakage under pathologically reduced oxygen levels. Since ischemia and hypoxia may have a role in various retinal diseases such as diabetic retinopathy and retinopathy of prematurity, studying the cellular and molecular response to reduced tissue oxygenation is of high relevance. In addition, the concept of preconditioning with ischemia or hypoxia demonstrates the capacity of the retina to activate endogenous survival mechanisms, which may protect cells against a following noxious insult. Part of these mechanisms is the local production of protective factors such as erythropoietin. Due to its plethora of effects in the retina including neuro- and vaso-protective activities, erythropoietin has gained strong interest as potential therapeutic factor for retinal degenerative diseases.
Collapse
Affiliation(s)
- Christian Caprara
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
42
|
Bringmann A, Wiedemann P. Müller glial cells in retinal disease. ACTA ACUST UNITED AC 2011; 227:1-19. [PMID: 21921569 DOI: 10.1159/000328979] [Citation(s) in RCA: 289] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 04/27/2011] [Indexed: 11/19/2022]
Abstract
Virtually all pathogenic stimuli activate Müller cells. Reactive Müller cells exert protective and toxic effects on photoreceptors and neurons. They contribute to oxidative stress and glutamate toxicity due to malfunctions of glutamate uptake and glutathione synthesis. Downregulation of potassium conductance disrupts transcellular potassium and water transport, resulting in neuronal hyperexcitability and edema. Protective effects of reactive Müller cells include upregulation of adenosine 5'-triphosphate (ATP)-degrading ectoenzymes, which enhances the extracellular availability of the neuroprotectant adenosine, abrogation of the osmotic release of ATP, which might protect retinal ganglion cells from apoptosis, and the release of antioxidants and neurotrophic factors. The dedifferentiation of reactive Müller cells to progenitor-like cells might have an impact on future therapeutic approaches. A better understanding of the gliotic mechanisms will be helpful in developing efficient therapeutic strategies aiming at increased protective and regenerative properties and decreased toxicity of reactive Müller cells.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
43
|
Doonan F, O’Driscoll C, Kenna P, Cotter TG. Enhancing survival of photoreceptor cells in vivo using the synthetic progestin Norgestrel. J Neurochem 2011; 118:915-27. [DOI: 10.1111/j.1471-4159.2011.07354.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Yamashita H, Yamada-Nakayama C, Sugihara K, Tsuji S, Sakurai T, Ban Y, Tsutsumi S, Sato Y. Functional and morphological effects of β-estradiol in eyes with N-methyl-D-Aspartate-induced retinal neurotoxicity in rats. Exp Eye Res 2011; 93:75-81. [PMID: 21600896 DOI: 10.1016/j.exer.2011.04.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/07/2011] [Accepted: 04/12/2011] [Indexed: 11/27/2022]
Abstract
Glutamate-mediated excitotoxicity, mainly induced by N-methyl-d-aspartate (NMDA) receptors, is known to cause retinal ganglion cell death in retinal ischemia, glaucoma, and several other retinal diseases. We evaluated the effects of β-estradiol (E2) against a single intravitreal injection of NMDA using a functional and morphological approach. Male rats were randomly divided into 3 treatment groups: (1) Control; (2) NMDA (intravitreal injection of 5 mM NMDA); and (3) NMDA + E2 (intravitreal injection of 5 mM NMDA and pretreatment with subcutaneous E2 implantation). Seven days after NMDA injection, full-field electroretinograms (ERGs) and quantitative morphological analyses using transverse sections of the retina were conducted. In the NMDA group, full-field ERGs showed reductions in the amplitudes of the negative-scotopic threshold response, rod response b-wave, oscillatory potentials, flicker response second b-wave and cone response b-wave. Morphological evaluations of transverse sections of the retina demonstrated a reduction in the thickness of the inner plexiform layer, increases in the thickness of the outer plexiform and outer nuclear layers, and a loss of cells in the ganglion cell layer. In the NMDA + E2 group, pretreatment with E2 prevented the aggravations in the amplitudes of the ERGs except for oscillatory potential 2 (OP2); however, no morphological differences between the NMDA and NMDA + E2 groups were seen. These findings indicate that E2 can protect retinal function against NMDA-induced neurotoxicity. In addition, these indications suggested that the effect of E2 may have therapeutic benefits in NMDA related diseases, such as retinal ischemia and glaucoma.
Collapse
Affiliation(s)
- Haruhiro Yamashita
- Drug Safety Laboratory, Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi 331-9530, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Nanoceria extend photoreceptor cell lifespan in tubby mice by modulation of apoptosis/survival signaling pathways. Neurobiol Dis 2011; 42:514-23. [PMID: 21396448 DOI: 10.1016/j.nbd.2011.03.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 02/28/2011] [Accepted: 03/02/2011] [Indexed: 01/10/2023] Open
Abstract
Cerium oxide nanoparticles, nanoceria, are inorganic antioxidants that have catalytic activities which mimic those of the neuroprotective enzymes superoxide dismutase and catalase. We have previously shown that nanoceria preserve retinal morphology and prevent loss of retinal function in a rat light damage model. In this study, the homozygous tubby mutant mouse, which exhibits inherited early progressive cochlear and retinal degeneration, was used as a model to test the ability of nanoceria to slow the progression of retinal degeneration. Tubby mice were injected systemically, intracardially, with 20 μl of 1mM nanoceria in saline, at postnatal day 10 and subsequently at P20 and P30 whereas saline injected and uninjected wild type (or heterozygous tubby) served as injected and uninjected controls, respectively. Assays for retinal function, morphology and signaling pathway gene expression were performed on P34 mice. Our data demonstrate that nanoceria protect the retina by decreasing Reactive Oxygen Species (ROS), up-regulating the expression of neuroprotection-associated genes; down-regulating apoptosis signaling pathways and/or up-regulating survival signaling pathways to slow photoreceptor degeneration. These data suggest that nanoceria have significant potential as global agents for therapeutic treatment of inherited retinal degeneration and most types of ocular diseases.
Collapse
|
46
|
Natoli R, Zhu Y, Valter K, Bisti S, Eells J, Stone J. Gene and noncoding RNA regulation underlying photoreceptor protection: microarray study of dietary antioxidant saffron and photobiomodulation in rat retina. Mol Vis 2010; 16:1801-22. [PMID: 20844572 PMCID: PMC2932490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 08/31/2010] [Indexed: 11/28/2022] Open
Abstract
PURPOSE To identify the genes and noncoding RNAs (ncRNAs) involved in the neuroprotective actions of a dietary antioxidant (saffron) and of photobiomodulation (PBM). METHODS We used a previously published assay of photoreceptor damage, in which albino Sprague Dawley rats raised in dim cyclic illumination (12 h 5 lux, 12 h darkness) were challenged by 24 h exposure to bright (1,000 lux) light. Experimental groups were protected against light damage by pretreatment with dietary saffron (1 mg/kg/day for 21 days) or PBM (9 J/cm(2) at the eye, daily for 5 days). RNA from one eye of four animals in each of the six experimental groups (control, light damage [LD], saffron, PBM, saffronLD, and PBMLD) was hybridized to Affymetrix rat genome ST arrays. Quantitative real-time PCR analysis of 14 selected genes was used to validate the microarray results. RESULTS LD caused the regulation of 175 entities (genes and ncRNAs) beyond criterion levels (p<0.05 in comparison with controls, fold-change >2). PBM pretreatment reduced the expression of 126 of these 175 LD-regulated entities below criterion; saffron pretreatment reduced the expression of 53 entities (50 in common with PBM). In addition, PBM pretreatment regulated the expression of 67 entities not regulated by LD, while saffron pretreatment regulated 122 entities not regulated by LD (48 in common with PBM). PBM and saffron, given without LD, regulated genes and ncRNAs beyond criterion levels, but in lesser numbers than during their protective action. A high proportion of the entities regulated by LD (>90%) were known genes. By contrast, ncRNAs were prominent among the entities regulated by PBM and saffron in their neuroprotective roles (73% and 62%, respectively). CONCLUSIONS Given alone, saffron and (more prominently) PBM both regulated significant numbers of genes and ncRNAs. Given before retinal exposure to damaging light, thus while exerting their neuroprotective action, they regulated much larger numbers of entities, among which ncRNAs were prominent. Further, the downregulation of known genes and of ncRNAs was prominent in the protective actions of both neuroprotectants. These comparisons provide an overview of gene expression induced by two neuroprotectants and provide a basis for the more focused study of their mechanisms.
Collapse
Affiliation(s)
- Riccardo Natoli
- Division of Biomedical Sciences & Biochemistry, Research School of Biology, Australian National University; Sydney, Australia,ARC Centre of Excellence in Vision Science, Sydney, Australia
| | - Yuan Zhu
- ARC Centre of Excellence in Vision Science, Sydney, Australia,Bosch Institute, Discipline of Physiology and Save Sight Institute, University of Sydney, Sydney, Australia
| | - Krisztina Valter
- Division of Biomedical Sciences & Biochemistry, Research School of Biology, Australian National University; Sydney, Australia,ARC Centre of Excellence in Vision Science, Sydney, Australia
| | - Silvia Bisti
- ARC Centre of Excellence in Vision Science, Sydney, Australia,Department of Science and Biomedical Technology, University of L’Aquila, Coppito II, Via Vetoio, L’Aquila, Italy,Bosch Institute, Discipline of Physiology and Save Sight Institute, University of Sydney, Sydney, Australia
| | - Janis Eells
- ARC Centre of Excellence in Vision Science, Sydney, Australia,Department of Biomedical Sciences University of Wisconsin Milwaukee, Milwaukee, WI
| | - Jonathan Stone
- ARC Centre of Excellence in Vision Science, Sydney, Australia,Bosch Institute, Discipline of Physiology and Save Sight Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
47
|
Kirsch M, Trautmann N, Ernst M, Hofmann HD. Involvement of gp130-associated cytokine signaling in Müller cell activation following optic nerve lesion. Glia 2010; 58:768-79. [PMID: 20091786 DOI: 10.1002/glia.20961] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ciliary neurotrophic factor (CNTF) and the related cytokine leukemia inhibitory factor (LIF) have been implicated in regulating astrogliosis following CNS lesions. Application of the factors activates astrocytes in vivo and in vitro, and their expression as well as their receptors is upregulated after brain injury. Here, we investigated their function by studying Müller cell activation induced by optic nerve crush in CNTF- and LIF-deficient mice, and in animals with deficiencies in cytokine signaling pathways. In the retina of CNTF(-/-) mice, basal GFAP expression was reduced, but unexpectedly, injury-induced upregulation in activated Müller cells was increased during the first 3 days after lesion as compared to wild-type animals and this corresponded with higher phosphorylation level of STAT3, an indicator of cytokine signaling. The observation that LIF expression was strongly upregulated in CNTF(-/-) mice but not in wild-type animals following optic nerve lesion provided a possible explanation. In fact, additional ablation of the LIF gene in CNTF/LIF double knockout mice almost completely abolished early lesion-induced GFAP upregulation in Müller cells and STAT3 phosphorylation. Early Müller cell activation was also eliminated in LIF(-/-) mice, despite normal CNTF levels, as well as in mutants deficient in gp130/JAK/STAT signaling and in conditional STAT3 knockout mice. Our results demonstrate that LIF signaling via the gp130/JAK/STAT3 pathway is required for the initiation of the astrogliosis-like reaction of retinal Müller cells after optic nerve injury. A potential role of CNTF was possibly masked by a compensatory increase in LIF signaling in the absence of CNTF.
Collapse
Affiliation(s)
- Matthias Kirsch
- Institute of Anatomy and Cell Biology, University of Freiburg, Albertstr. 23, Freiburg, Germany
| | | | | | | |
Collapse
|
48
|
Exogenous modulation of intrinsic optic nerve neuroprotective activity. Graefes Arch Clin Exp Ophthalmol 2010; 248:1105-16. [PMID: 20229104 DOI: 10.1007/s00417-010-1336-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 02/06/2010] [Accepted: 02/14/2010] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND To characterize the molecular and functional status of the rat retina and optic nerve after acute elevation of intraocular pressure (IOP). METHODS Retinal ischemia was induced in rats by increasing the IOP (110 mmHg/60 minutes). Microarray analysis, quantitative RT-PCR (qRT-PCR) and immunohistochemistry were used to characterize retinal tissue. PLGA microspheres containing neurotrophic factors (BDNF, GDNF, or CNTF) or empty microspheres were injected into the vitreous of operated animals 1 day after elevation of IOP. Pupil light reflex (PLR) parameters and electroretinograms (ERG) were monitored at multiple time points during the 60-day postoperative recovery period. RESULTS Molecular analysis showed a significant intrinsic up-regulation of CNTF at 10 and 25 days after induction of the acute ocular hypertension (p = 0.0067). Molecular tissue analysis of GDNF and its receptors (GDNFR1, GDNFR2), and BDNF and its receptor (trkB) showed no change in expression. Animals that received CNTF microspheres had no significant functional recovery compared to animals which received blank microspheres (p > 0.05). Animals that received GDNF or BDNF microspheres showed significant PLR recovery (p < 0.05 and p < 0.001 respectively) compared to non-treated animals. CONCLUSIONS Continuous release of neurotrophic growth factors (NGFs) significantly protects optic nerve function in the experimental model of retinal ischemia observed by PLR analysis.
Collapse
|
49
|
Near-infrared light protect the photoreceptor from light-induced damage in rats. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 664:365-74. [PMID: 20238037 DOI: 10.1007/978-1-4419-1399-9_42] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND A project originally developed for NASA plant growth experiments in space demonstrating the Light-Emitting Diode (LED) could promote the wound healing. Further study showed that the LED's could protect cells by stimulating the basic energy processes in the mitochondria of each cell. OBJECTIVE The purpose of this study was to assess the effects of 670 nm LED to protect the photoreceptor from the light-induced damage in a rodent model. METHODS SD rats were randomly assigned to one of eight groups: untreated control group, the LED-treated control group, three light-induced damage groups, and three LED-protected groups. The rats were exposed to constant light for 3 h of different illuminations of 900, 1,800 and 2,700 lux, respectively. The LED treatment (50 mW) were done for 30 min, 3 h before the light damage and 0, 24 and 48 h after the light damage. Using the electroretinogram as a sensitive indicator of retinal function, and the histopathologic change was showed as a proof of the protective effect of LED treatment. RESULTS The 900 lux illumination for 3 h did not cause damage to the retina of rats, however, the 1,800 lux illumination for 3 h caused significant damage to ONL of an approximate half retina, which caused the swing of ERG b wave to be 431 muV. With the LED protection: the damage of ONL was near 1/6 of retina, which was significantly reduced than the ones without LED protection (P < 0.01); and the swing of ERG b wave was recorded to be 1,011 muV, which was increased significantly than the ones without LED protection (P < 0.01). The illumination of 2,700 lux for 3 h caused severe damage to the rats' retinas and the LED could not protect them significantly in both of morphology and function (P > 0.05, P > 0.05). CONCLUSIONS 670 nm LED treatment has an evident protective effect on retinal cells against light-induced damage, which may be an innovative and non-invasive therapeutic approach to prevent or to delay age-related macular degeneration.
Collapse
|
50
|
Abstract
By its action on rhodopsin, light triggers the well-known visual transduction cascade, but can also induce cell damage and death through phototoxic mechanisms - a comprehensive understanding of which is still elusive despite more than 40 years of research. Herein, we integrate recent experimental findings to address several hypotheses of retinal light damage, premised in part on the close anatomical and metabolic relationships between the photoreceptors and the retinal pigment epithelium. We begin by reviewing the salient features of light damage, recently joined by evidence for retinal remodeling which has implications for the prognosis of recovery of function in retinal degenerations. We then consider select factors that influence the progression of the damage process and the extent of visual cell loss. Traditional, genetically modified, and emerging animal models are discussed, with particular emphasis on cone visual cells. Exogenous and endogenous retinal protective factors are explored, with implications for light damage mechanisms and some suggested avenues for future research. Synergies are known to exist between our long term light environment and photoreceptor cell death in retinal disease. Understanding the molecular mechanisms of light damage in a variety of animal models can provide valuable insights into the effects of light in clinical disorders and may form the basis of future therapies to prevent or delay visual cell loss.
Collapse
|