101
|
Li H, Liu B, Lian L, Zhou J, Xiang S, Zhai Y, Chen Y, Ma X, Wu W, Hou L. High dose expression of heme oxigenase-1 induces retinal degeneration through ER stress-related DDIT3. Mol Neurodegener 2021; 16:16. [PMID: 33691741 PMCID: PMC7944639 DOI: 10.1186/s13024-021-00437-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxidative stress is a common cause of neurodegeneration and plays a central role in retinal degenerative diseases. Heme oxygenase-1 (HMOX1) is a redox-regulated enzyme that is induced in neurodegenerative diseases and acts against oxidative stress but can also promote cell death, a phenomenon that is still unexplained in molecular terms. Here, we test whether HMOX1 has opposing effects during retinal degeneration and investigate the molecular mechanisms behind its pro-apoptotic role. METHODS Basal and induced levels of HMOX1 in retinas are examined during light-induced retinal degeneration in mice. Light damage-independent HMOX1 induction at two different expression levels is achieved by intraocular injection of different doses of an adeno-associated virus vector expressing HMOX1. Activation of Müller glial cells, retinal morphology and photoreceptor cell death are examined using hematoxylin-eosin staining, TUNEL assays, immunostaining and retinal function are evaluated with electroretinograms. Downstream gene expression of HMOX1 is analyzed by RNA-seq, qPCR examination and western blotting. The role of one of these genes, the pro-apoptotic DNA damage inducible transcript 3 (Ddit3), is analyzed in a line of knockout mice. RESULTS Light-induced retinal degeneration leads to photoreceptor degeneration and concomitant HMOX1 induction. HMOX1 expression at low levels before light exposure prevents photoreceptor degeneration but expression at high levels directly induces photoreceptor degeneration even without light stress. Photoreceptor degeneration following high level expression of HMOX1 is associated with a mislocalization of rhodopsin in photoreceptors and an increase in the expression of DDIT3. Genetic deletion of Ddit3 in knockout mice prevents photoreceptor cell degeneration normally resulting from high level HMOX1 expression. CONCLUSION The results reveal that the expression levels determine whether HMOX1 is protective or deleterious in the retina. Furthermore, in contrast to the protective low dose of HMOX1, the deleterious high dose is associated with induction of DDIT3 and endoplasmic reticulum stress as manifested, for instance, in rhodopsin mislocalization. Hence, future applications of HMOX1 or its regulated targets in gene therapy approaches should carefully consider expression levels in order to avoid potentially devastating effects.
Collapse
Affiliation(s)
- Huirong Li
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and 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, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jiajia Zhou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Shengjin Xiang
- Eye Hospital of Wenzhou Medical University, Wenzhou, 325003 China
| | - Yifan Zhai
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Wencan Wu
- Eye Hospital of Wenzhou Medical University, Wenzhou, 325003 China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
102
|
He Y, Xu Y, Chen Z, He B, Quan Z, Zhang R, Ren Y. Protective Effect of Mitochondrially Targeted Peptide Against Oxidant Injury of Cone Photoreceptors Through Preventing Necroptosis Pathway. J Biomed Nanotechnol 2021; 17:279-290. [PMID: 33785098 DOI: 10.1166/jbn.2021.3017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Retinopathy is an eye disease caused by the death of retinal cells in the macular area and the surrounding choroid. As the retinal rod cell dysfunction and death lead to the loss of night vision, the disease will lead to visual dysfunction and blindness as the disease progresses. Because of the irreversible nature of cell death, gene therapy has become a research hotspot in the field of retinopathy. But the technology is still in animal studies or clinical trials, and more research is needed to prove its feasibility. In this study, oxidative damage cell model was established and divided into a control group, H₂O₂ group, SS31 +NEC1 group, SS31 +H₂O₂ group, and SS31 +NEC1 +H₂O₂ group, for different interventions. The cell survival rate of the H₂O₂ group was significantly increased compared with those of the SS31 + H₂O₂ group, SS31 +NEC1 +H₂O₂ group, and NEC1 +H₂O₂ group. Nec1 combined treatment significantly reduced reactive oxygen species (ROS) production compared with that in the H₂O₂ group. The level of MDA in the SS31 group, Nec-1 group and combined treatment of SS31 +NEC1 group decreased significantly compared with the H₂O₂ group. The proportion of cells with decreased mitochondrial membrane potential in the H₂O₂ group significantly increased, and the rate of positivity for propidium iodide (PI) of 661W cells in the H₂O₂ group and the control group significantly increased. Nine hours after H₂O₂ treatment of 661W cells, the RIP3 expression level began to increase, and peaked at 24 h. The level of RIP3 in the H₂O₂ group was significantly increased, while this level was downregulated in the SS31 and NEC1 treatment groups. Therefore, this study suggests that SS31 has a partial protective effect on 661W cells by inhibiting necrosis, which has certain guiding significance for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Yuan He
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| | - Yun Xu
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| | - Zejun Chen
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| | - Beilei He
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| | - Zhuoya Quan
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| | - Ruixue Zhang
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| | - Yuan Ren
- Department of Ophthalmology, The Second Affiliated Hospital ofXi'an Medical University, Xi'an 710038, Shanxi, PR China
| |
Collapse
|
103
|
Retinal Neurodegeneration: Correlation between Nutraceutical Treatment and Animal Model. Nutrients 2021; 13:nu13030770. [PMID: 33673449 PMCID: PMC7997156 DOI: 10.3390/nu13030770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal diseases can be induced by a variety of factors, including gene mutations, environmental stresses and dysmetabolic processes. The result is a progressive deterioration of visual function, which sometimes leads to blindness. Many treatments are under investigation, though results are still mostly unsatisfactory and restricted to specific pathologies, particularly in the case of gene therapy. The majority of treatments have been tested in animal models, but very few have progressed to human clinical trials. A relevant approach is to study the relation between the type of treatments and the degenerative characteristics of the animal model to better understand the effectiveness of each therapy. Here we compare the results obtained from different animal models treated with natural compounds (saffron and naringenin) to anticipate the potentiality of a single treatment in different pathologies.
Collapse
|
104
|
Transcriptomic Changes Associated with Loss of Cell Viability Induced by Oxysterol Treatment of a Retinal Photoreceptor-Derived Cell Line: An In Vitro Model of Smith-Lemli-Opitz Syndrome. Int J Mol Sci 2021; 22:ijms22052339. [PMID: 33652836 PMCID: PMC7956713 DOI: 10.3390/ijms22052339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 11/17/2022] Open
Abstract
Smith–Lemli–Opitz Syndrome (SLOS) results from mutations in the gene encoding the enzyme DHCR7, which catalyzes conversion of 7-dehydrocholesterol (7DHC) to cholesterol (CHOL). Rats treated with a DHCR7 inhibitor serve as a SLOS animal model, and exhibit progressive photoreceptor-specific cell death, with accumulation of 7DHC and oxidized sterols. To understand the basis of this cell type specificity, we performed transcriptomic analyses on a photoreceptor-derived cell line (661W), treating cells with two 7DHC-derived oxysterols, which accumulate in tissues and bodily fluids of SLOS patients and in the rat SLOS model, as well as with CHOL (negative control), and evaluated differentially expressed genes (DEGs) for each treatment. Gene enrichment analysis and compilation of DEG sets indicated that endoplasmic reticulum stress, oxidative stress, DNA damage and repair, and autophagy were all highly up-regulated pathways in oxysterol-treated cells. Detailed analysis indicated that the two oxysterols exert their effects via different molecular mechanisms. Changes in expression of key genes in highlighted pathways (Hmox1, Ddit3, Trib3, and Herpud1) were validated by immunofluorescence confocal microscopy. The results extend our understanding of the pathobiology of retinal degeneration and SLOS, identifying potential new druggable targets for therapeutic intervention into these and other related orphan diseases.
Collapse
|
105
|
Murenu E, Kostidis S, Lahiri S, Geserich AS, Imhof A, Giera M, Michalakis S. Metabolic Analysis of Vitreous/Lens and Retina in Wild Type and Retinal Degeneration Mice. Int J Mol Sci 2021; 22:ijms22052345. [PMID: 33652907 PMCID: PMC7956175 DOI: 10.3390/ijms22052345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Photoreceptors are the light-sensing cells of the retina and the major cell type affected in most inherited retinal degenerations. Different metabolic pathways sustain their high energetic demand in physiological conditions, particularly aerobic glycolysis. The principal metabolome of the mature retina has been studied, but only limited information is available on metabolic adaptations in response to key developmental events, such as eye opening. Moreover, dynamic metabolic changes due to retinal degeneration are not well understood. Here, we aimed to explore and map the ocular metabolic dynamics induced by eye opening in healthy (wild type) or Pde6b-mutant (retinal degeneration 1, Rd1) mice, in which photoreceptors degenerate shortly after eye opening. To unravel metabolic differences emerging before and after eye opening under physiological and pathophysiological conditions, we performed nuclear magnetic resonance (NMR) spectroscopy-based metabolome analysis of wild type and Rd1 retina and vitreous/lens. We show that eye opening is accompanied by changes in the concentration of selected metabolites in the retina and by alterations in the vitreous/lens composition only in the retinal degeneration context. As such, we identify NAcetylaspartate as a potential novel vitreous/lens marker reflecting progressive retinal degeneration. Thus, our data can help elucidating mechanisms underlying key events in retinal physiology and reveal changes occurring in pathology, while highlighting the importance of the vitreous/lens in the characterization of retinal diseases.
Collapse
Affiliation(s)
- Elisa Murenu
- Department of Ophthalmology, Ludwig-Maximilians-Universität München, Mathildenstraße 8, 80336 Munich, Germany;
- Department of Pharmacy, Ludwig-Maximilians Universität München, Butenandtstr. 7, 81377 Munich, Germany;
| | - Sarantos Kostidis
- Leiden University Medical Center, Center for Proteomics & Metabolomics, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.K.); (M.G.)
| | - Shibojyoti Lahiri
- Biomedical Center Munich-Molecular Biology, Ludwig-Maximilians-Universität München, Großhaderner Strasse 9, 82152 Planegg-Martinsried, Germany; (S.L.); (A.I.)
| | - Anna S. Geserich
- Department of Pharmacy, Ludwig-Maximilians Universität München, Butenandtstr. 7, 81377 Munich, Germany;
| | - Axel Imhof
- Biomedical Center Munich-Molecular Biology, Ludwig-Maximilians-Universität München, Großhaderner Strasse 9, 82152 Planegg-Martinsried, Germany; (S.L.); (A.I.)
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics & Metabolomics, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.K.); (M.G.)
| | - Stylianos Michalakis
- Department of Ophthalmology, Ludwig-Maximilians-Universität München, Mathildenstraße 8, 80336 Munich, Germany;
- Department of Pharmacy, Ludwig-Maximilians Universität München, Butenandtstr. 7, 81377 Munich, Germany;
- Correspondence: ; Tel.: +49-89-2180-77325
| |
Collapse
|
106
|
Koyanagi Y, Akiyama M, Nishiguchi KM, Momozawa Y, Kamatani Y, Takata S, Inai C, Iwasaki Y, Kumano M, Murakami Y, Komori S, Gao D, Kurata K, Hosono K, Ueno S, Hotta Y, Murakami A, Terasaki H, Wada Y, Nakazawa T, Ishibashi T, Ikeda Y, Kubo M, Sonoda KH. Regional differences in genes and variants causing retinitis pigmentosa in Japan. Jpn J Ophthalmol 2021; 65:338-343. [PMID: 33629268 DOI: 10.1007/s10384-021-00824-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE To investigate the regional differences in the genes and variants causing retinitis pigmentosa (RP) in Japan STUDY DESIGN: Retrospective multicenter study METHODS: In total, 1204 probands of each pedigree clinically diagnosed with nonsyndromic RP were enrolled from 5 Japanese facilities. The regions were divided into the Tohoku region, the Kanto and Chubu regions, and the Kyushu region according to the location of the hospitals where the participants were enrolled. We compared the proportions of the causative genes and the distributions of the pathogenic variants among these 3 regions. RESULTS The proportions of genetically solved cases were 29.4% in the Tohoku region (n = 500), 29.6% in the Kanto and Chubu regions (n = 196), and 29.7% in the Kyushu region (n = 508), which did not differ statistically (P = .99). No significant regional differences in the proportions of each causative gene in genetically solved patients were observed after correction by multiple testing. Among the 29 pathogenic variants detected in all 3 regions, only p.(Pro347Leu) in RHO was an autosomal dominant variant; the remaining 28 variants were found in autosomal recessive genes. Conversely, 78.6% (275/350) of the pathogenic variants were detected only in a single region, and 6 pathogenic variants (p.[Asn3062fs] in EYS, p.[Ala315fs] in EYS, p.[Arg872fs] in RP1, p.[Ala126Val] in RDH12, p.[Arg41Trp] in CRX, and p.[Gly381fs] in PRPF31) were frequently found in ≥ 4 patients in the single region. CONCLUSION We observed region-specific pathogenic variants in the Japanese population. Further investigations of causative genes in multiple regions in Japan will contribute to the expansion of the catalog of genetic variants causing RP.
Collapse
Affiliation(s)
- Yoshito Koyanagi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Masato Akiyama
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. .,Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan. .,Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.,Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Sadaaki Takata
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Chihiro Inai
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Yusuke Iwasaki
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Mikako Kumano
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shiori Komori
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dan Gao
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kentaro Kurata
- Department of Ophthalmology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Katsuhiro Hosono
- Department of Ophthalmology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shinji Ueno
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Hotta
- Department of Ophthalmology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroko Terasaki
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Toru Nakazawa
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuro Ishibashi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuhiro Ikeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
107
|
Retinal Inflammation, Cell Death and Inherited Retinal Dystrophies. Int J Mol Sci 2021; 22:ijms22042096. [PMID: 33672611 PMCID: PMC7924201 DOI: 10.3390/ijms22042096] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a group of retinal disorders that cause progressive and severe loss of vision because of retinal cell death, mainly photoreceptor cells. IRDs include retinitis pigmentosa (RP), the most common IRD. IRDs present a genetic and clinical heterogeneity that makes it difficult to achieve proper treatment. The progression of IRDs is influenced, among other factors, by the activation of the immune cells (microglia, macrophages, etc.) and the release of inflammatory molecules such as chemokines and cytokines. Upregulation of tumor necrosis factor alpha (TNFα), a pro-inflammatory cytokine, is found in IRDs. This cytokine may influence photoreceptor cell death. Different cell death mechanisms are proposed, including apoptosis, necroptosis, pyroptosis, autophagy, excessive activation of calpains, or parthanatos for photoreceptor cell death. Some of these cell death mechanisms are linked to TNFα upregulation and inflammation. Therapeutic approaches that reduce retinal inflammation have emerged as useful therapies for slowing down the progression of IRDs. We focused this review on the relationship between retinal inflammation and the different cell death mechanisms involved in RP. We also reviewed the main anti-inflammatory therapies for the treatment of IRDs.
Collapse
|
108
|
Maguire AM, Bennett J, Aleman EM, Leroy BP, Aleman TS. Clinical Perspective: Treating RPE65-Associated Retinal Dystrophy. Mol Ther 2021; 29:442-463. [PMID: 33278565 PMCID: PMC7854308 DOI: 10.1016/j.ymthe.2020.11.029] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/16/2020] [Accepted: 11/25/2020] [Indexed: 12/25/2022] Open
Abstract
Until recently, there was no approved treatment for a retinal degenerative disease. Subretinal injection of a recombinant adeno-associated virus (AAV) delivering the normal copy of the human RPE65 cDNA led to reversal of blindness first in animal models and then in humans. This led to the first US Food and Drug Administration (FDA)-approved gene therapy product for a genetic disease, voretigene neparvovec-rzyl (Luxturna). Luxturna was then approved by the European Medicines Association and is now available in the US through Spark Therapeutics and worldwide through Novartis. Not only has treatment with Luxturna changed the lives of people previously destined to live a life of blindness, but it has fueled interest in developing additional gene therapy reagents targeting numerous other genetic forms of inherited retinal disease. This review describes many of the considerations for administration of Luxturna and describes how lessons from experience with Luxturna could lead to additional gene-based treatments of blindness.
Collapse
Affiliation(s)
- Albert M Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Elena M Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bart P Leroy
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Ophthalmology and Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Tomas S Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.
| |
Collapse
|
109
|
Wu DM, Ji X, Ivanchenko MV, Chung M, Piper M, Rana P, Wang SK, Xue Y, West E, Zhao SR, Xu H, Cicconet M, Xiong W, Cepko CL. Nrf2 overexpression rescues the RPE in mouse models of retinitis pigmentosa. JCI Insight 2021; 6:145029. [PMID: 33491671 PMCID: PMC7934854 DOI: 10.1172/jci.insight.145029] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Nrf2, a transcription factor that regulates the response to oxidative stress, has been shown to rescue cone photoreceptors and slow vision loss in mouse models of retinal degeneration (rd). The retinal pigment epithelium (RPE) is damaged in these models, but whether it also could be rescued by Nrf2 has not been previously examined. We used an adeno-associated virus (AAV) with an RPE-specific (Best1) promoter to overexpress Nrf2 in the RPE of rd mice. Control rd mice showed disruption of the regular array of the RPE, as well as loss of RPE cells. Cones were lost in circumscribed regions within the cone photoreceptor layer. Overexpression of Nrf2 specifically in the RPE was sufficient to rescue the RPE, as well as the disruptions in the cone photoreceptor layer. Electron microscopy showed compromised apical microvilli in control rd mice but showed preserved microvilli in Best1-Nrf2–treated mice. The rd mice treated with Best1-Nrf2 had slightly better visual acuity. Transcriptome profiling showed that Nrf2 upregulates multiple oxidative defense pathways, reversing declines seen in the glutathione pathway in control rd mice. In summary, Nrf2 overexpression in the RPE preserves RPE morphology and survival in rd mice, and it is a potential therapeutic for diseases involving RPE degeneration, including age-related macular degeneration (AMD).
Collapse
Affiliation(s)
- David M Wu
- Massachusetts Eye and Ear Infirmary Retina Service, Department of Ophthalmology.,Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Xuke Ji
- Massachusetts Eye and Ear Infirmary Retina Service, Department of Ophthalmology.,Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Maryna V Ivanchenko
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle Chung
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Mary Piper
- Department of Bioinformatics, T.H. Chan Harvard School of Public Health, Boston, Massachusetts, USA
| | - Parimal Rana
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Sean K Wang
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Yunlu Xue
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Emma West
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Sophia R Zhao
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and
| | - Hongbin Xu
- Departments of Genetics and Ophthalmology, Blavatnik Institute, and.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Marcelo Cicconet
- Image and Data Analysis Core, Harvard Medical School, Boston, Massachusetts, USA
| | - Wenjun Xiong
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Constance L Cepko
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
110
|
Wang J, Xiao H, Barwick S, Liu Y, Smith SB. Optimal timing for activation of sigma 1 receptor in the Pde6b rd10/J (rd10) mouse model of retinitis pigmentosa. Exp Eye Res 2021; 202:108397. [PMID: 33310057 PMCID: PMC7808329 DOI: 10.1016/j.exer.2020.108397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/24/2020] [Accepted: 12/07/2020] [Indexed: 11/18/2022]
Abstract
Sigma 1 Receptor (Sig1R), a pluripotent modulator of cell survival, is a promising target for treatment of retinal degenerative diseases. Previously, we reported that administration of the high-affinity, high-specificity Sig1R ligand (+)-pentazocine, ((+)-PTZ) beginning at post-natal day 14 (P14) and continuing every other day improves visual acuity and delays loss of photoreceptor cells (PRCs) in the Pde6βrd10/J (rd10) mouse model of retinitis pigmentosa. Whether administration of (+)-PTZ, at time points concomitant with (P18) or following (P21, P24) onset of PRC death, would prove neuroprotective was investigated in this study. Rd10 mice were administered (+)-PTZ intraperitoneally [0.5 mg/kg], starting at either P14, P18, P21 or P24. Injections continued every other day through P42. Visual acuity was assessed using the optokinetic tracking response (OKR). Rd10 mice treated with (+)-PTZ beginning at P14 retained visual acuity for the duration of the study (~0.33 c/d at P21, ~0.38 c/d at P28, ~0.32 c/d at P35, ~0.32 c/d at P42), whereas mice injected beginning at P18, P21, P24 showed a decline in acuity when tested at P35 and P42. Their acuity was only slightly better than rd10-non-treated mice. Electrophysiologic function was assessed using scotopic and photopic electroretinography (ERG) to assess rod and cone function, respectively. Photopic a- and b-wave amplitudes were significantly greater in rd10 mice treated with (+)-PTZ beginning at P14 compared with non-treated mice and those in the later-onset (+)-PTZ injection groups. Retinal architecture was visualized in living mice using spectral domain-optical coherence tomography (SD-OCT) allowing measurement of the total retinal thickness, the inner retina and the outer retina (the area most affected in rd10 mice). The outer retina measured ~35 μm in rd10 mice treated with (+)-PTZ beginning at P14, which was significantly greater than mice in the later-onset (+)-PTZ injection groups (~25 μm) and non-treated rd10 mice (~25 μm). Following the visual function studies performed in the living mice, eyes were harvested at P42 for histologic analysis. While the inner retina was largely intact in all (+)-PTZ-injection groups, there was a marked reduction in the outer retina of non-treated rd10 mice (e.g. in the outer nuclear layer there were ~10 PRCs/100 μm retinal length). The rd10 mice treated with (+)-PTZ beginning at P14 had ~20 PRCs/100 μm retinal length, whereas the mice in groups beginning P18, P21 and P24 had ~16 PRCs/100 μm retinal length. In conclusion, the data indicate that delaying (+)-PTZ injection past the onset of PRC death in rd10 mice - even by a few days - can negatively impact the long-term preservation of retinal function. Our findings suggest that optimizing the administration of Sig1R ligands is critical for retinal neuroprotection.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Shannon Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States; Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, United States.
| |
Collapse
|
111
|
Kutsyr O, Sánchez-Sáez X, Martínez-Gil N, de Juan E, Lax P, Maneu V, Cuenca N. Gradual Increase in Environmental Light Intensity Induces Oxidative Stress and Inflammation and Accelerates Retinal Neurodegeneration. Invest Ophthalmol Vis Sci 2021; 61:1. [PMID: 32744596 PMCID: PMC7441298 DOI: 10.1167/iovs.61.10.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) is a blinding neurodegenerative disease of the retina that can be affected by many factors. The present study aimed to analyze the effect of different environmental light intensities in rd10 mice retina. Methods C57BL/6J and rd10 mice were bred and housed under three different environmental light intensities: scotopic (5 lux), mesopic (50 lux), and photopic (300 lux). Visual function was studied using electroretinography and optomotor testing. The structural and morphological integrity of the retinas was evaluated by optical coherence tomography imaging and immunohistochemistry. Additionally, inflammatory processes and oxidative stress markers were analyzed by flow cytometry and western blotting. Results When the environmental light intensity was higher, retinal function decreased in rd10 mice and was accompanied by light-dependent photoreceptor loss, followed by morphological alterations, and synaptic connectivity loss. Moreover, light-dependent retinal degeneration was accompanied by an increased number of inflammatory cells, which became more activated and phagocytic, and by an exacerbated reactive gliosis. Furthermore, light-dependent increment in oxidative stress markers in rd10 mice retina pointed to a possible mechanism for light-induced photoreceptor degeneration. Conclusions An increase in rd10 mice housing light intensity accelerates retinal degeneration, activating cell death, oxidative stress pathways, and inflammatory cells. Lighting intensity is a key factor in the progression of retinal degeneration, and standardized lighting conditions are advisable for proper analysis and interpretation of experimental results from RP animal models, and specifically from rd10 mice. Also, it can be hypothesized that light protection could be an option to slow down retinal degeneration in some cases of RP.
Collapse
|
112
|
Locus-Level Changes in Macular Sensitivity in Patients with Retinitis Pigmentosa Treated with Oral N-acetylcysteine. Am J Ophthalmol 2021; 221:105-114. [PMID: 32795434 DOI: 10.1016/j.ajo.2020.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/27/2020] [Accepted: 08/03/2020] [Indexed: 01/10/2023]
Abstract
PURPOSE To identify characteristics of loci associated with locus-level sensitivity loss or improvement during treatment with N-acetylcysteine (NAC) in retinitis pigmentosa (RP). DESIGN Retrospective analysis of prospectively collected data in the FIGHT RP clinical trial. METHODS Patients (n = 30) were treated with 600, 1,200, or 1,800 mg of NAC twice daily for 3 months and then 3 times/day for 3 months. Microperimetry locus-level changes between baseline and month 6 were correlated with baseline characteristics of loci using regression models. The main outcome measurement was locus-level sensitivity change ≥6 dB. RESULTS Baseline mean sensitivity (3,468 loci; 51 evaluable eyes) was 7.7 dB and for foveal, parafoveal, and perifoveal loci were 20.2, 11.8, and 5.8 dB. During treatment, 287 loci (8.28%) increased ≥6 dB, and 119 of 1,613 loci with baseline sensitivity ≥6 dB decreased ≥6 dB (7.38%). A higher dose of NAC was associated with lower likelihood of sensitivity loss ≥6 dB (P = .033). Loci with low baseline sensitivity were more likely to decrease ≥6 dB (P = .034) but also more likely to increase ≥6 dB (P < .001). Foveal versus perifoveal loci (P < .001) and superior versus inferior loci (P = .005) were more likely to increase ≥6 dB. CONCLUSIONS Higher doses of NAC reduced risk of macular loci sensitivity loss in RP. Greater sensitivity depression reversibility in the fovea during treatment suggests that high foveal cone density protects cones from irreversible loss of function in RP making them more likely to show improved function during NAC treatment.
Collapse
|
113
|
Raghu G, Berk M, Campochiaro PA, Jaeschke H, Marenzi G, Richeldi L, Wen FQ, Nicoletti F, Calverley PMA. The Multifaceted Therapeutic Role of N-Acetylcysteine (NAC) in Disorders Characterized by Oxidative Stress. Curr Neuropharmacol 2021; 19:1202-1224. [PMID: 33380301 PMCID: PMC8719286 DOI: 10.2174/1570159x19666201230144109] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/27/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress, which results in the damage of diverse biological molecules, is a ubiquitous cellular process implicated in the etiology of many illnesses. The sulfhydryl-containing tripeptide glutathione (GSH), which is synthesized and maintained at high concentrations in all cells, is one of the mechanisms by which cells protect themselves from oxidative stress. N-acetylcysteine (NAC), a synthetic derivative of the endogenous amino acid L-cysteine and a precursor of GSH, has been used for several decades as a mucolytic and as an antidote to acetaminophen (paracetamol) poisoning. As a mucolytic, NAC breaks the disulfide bonds of heavily cross-linked mucins, thereby reducing mucus viscosity. In vitro, NAC has antifibrotic effects on lung fibroblasts. As an antidote to acetaminophen poisoning, NAC restores the hepatic GSH pool depleted in the drug detoxification process. More recently, improved knowledge of the mechanisms by which NAC acts has expanded its clinical applications. In particular, the discovery that NAC can modulate the homeostasis of glutamate has prompted studies of NAC in neuropsychiatric diseases characterized by impaired glutamate homeostasis. This narrative review provides an overview of the most relevant and recent evidence on the clinical application of NAC, with a focus on respiratory diseases, acetaminophen poisoning, disorders of the central nervous system (chronic neuropathic pain, depression, schizophrenia, bipolar disorder, and addiction), cardiovascular disease, contrast-induced nephropathy, and ophthalmology (retinitis pigmentosa).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peter M. A. Calverley
- Address correspondence to this author at Clinical Science Centre, University Hospital Aintree, Longmoor Lane, Liverpool UK L9 7AL; Tel: +44 151 529 5886, Fax: +44 151 529 5888; E-mail:
| |
Collapse
|
114
|
Koyanagi Y, Ueno S, Ito Y, Kominami T, Komori S, Akiyama M, Murakami Y, Ikeda Y, Sonoda KH, Terasaki H. Relationship Between Macular Curvature and Common Causative Genes of Retinitis Pigmentosa in Japanese Patients. Invest Ophthalmol Vis Sci 2021; 61:6. [PMID: 32749464 PMCID: PMC7441377 DOI: 10.1167/iovs.61.10.6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To determine the relationship between the macular curvature and the causative genes of retinitis pigmentosa (RP). Methods We examined the medical records of the right eyes of 65 cases with RP (31 men and 34 women; average age, 47.6 years). There were 31 cases with the EYS variants, 11 cases with the USH2A variants, six cases with the RPGR variants, 13 cases with the RP1 variants, and four cases with the RP1L1 variants. The mean curvature of Bruch's membrane was calculated within 6 mm of the fovea as the mean macular curvature index (MMCI, 1/µm). We used multiple linear regression analysis to determine the independence of the causative genes contributing to the MMCIs after adjustments for age, sex, axial length, and width of the ellipsoid zone. Results The median MMCI was −31.2 × 10−5/µm for the RPGR eyes, −16.5 × 10−5/µm for the RP1L1 eyes, −13.0 × 10−5/µm for the RP1 eyes, −9.8 × 10−5/µm for the EYS eyes, and −9.0 × 10−5/µm for the USH2A eyes. Compared with the EYS gene as the reference gene, the RPGR gene was significantly related to the MMCI values after adjusting for the other parameters (P = 5.30 × 10−6). In contrast, the effects of the other genes, USH2A, RP1, and RP1L1, were not significantly different from that of the EYS gene (P = 0.26, P = 0.49, and P = 0.92, respectively). Conclusions The RPGR gene had a stronger effect on the steep macular curvature than the other ciliopathy-related genes.
Collapse
|
115
|
Gopalakrishnan S, Mehrvar S, Maleki S, Schmitt H, Summerfelt P, Dubis AM, Abroe B, Connor TB, Carroll J, Huddleston W, Ranji M, Eells JT. Photobiomodulation preserves mitochondrial redox state and is retinoprotective in a rodent model of retinitis pigmentosa. Sci Rep 2020; 10:20382. [PMID: 33230161 PMCID: PMC7684292 DOI: 10.1038/s41598-020-77290-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/26/2020] [Indexed: 01/06/2023] Open
Abstract
Photobiomodulation (PBM) by far-red (FR) to near-infrared (NIR) light has been demonstrated to restore the function of damaged mitochondria, increase the production of cytoprotective factors and prevent cell death. Our laboratory has shown that FR PBM improves functional and structural outcomes in animal models of retinal injury and retinal degenerative disease. The current study tested the hypothesis that a brief course of NIR (830 nm) PBM would preserve mitochondrial metabolic state and attenuate photoreceptor loss in a model of retinitis pigmentosa, the P23H transgenic rat. P23H rat pups were treated with 830 nm light (180 s; 25 mW/cm2; 4.5 J/cm2) using a light-emitting diode array (Quantum Devices, Barneveld, WI) from postnatal day (p) 10 to p25. Sham-treated rats were restrained, but not treated with 830 nm light. Retinal metabolic state, function and morphology were assessed at p30 by measurement of mitochondrial redox (NADH/FAD) state by 3D optical cryo-imaging, electroretinography (ERG), spectral-domain optical coherence tomography (SD-OCT), and histomorphometry. PBM preserved retinal metabolic state, retinal function, and retinal morphology in PBM-treated animals compared to the sham-treated group. PBM protected against the disruption of the oxidation state of the mitochondrial respiratory chain observed in sham-treated animals. Scotopic ERG responses over a range of flash intensities were significantly greater in PBM-treated rats compared to sham controls. SD-OCT studies and histological assessment showed that PBM preserved the structural integrity of the retina. These findings demonstrate for the first time a direct effect of NIR PBM on retinal mitochondrial redox status in a well-established model of retinal disease. They show that chronic proteotoxic stress disrupts retinal bioenergetics resulting in mitochondrial dysfunction, and retinal degeneration and that therapies normalizing mitochondrial metabolism have considerable potential for the treatment of retinal degenerative disease.
Collapse
Affiliation(s)
| | - Shima Mehrvar
- Biophotonics Laboratory, Department of Computer and Electrical Engineering and Computer Science, Florida Atlantic University, Boca Ratan, FL, USA
| | - Sepideh Maleki
- Biophotonics Laboratory, Department of Computer and Electrical Engineering and Computer Science, Florida Atlantic University, Boca Ratan, FL, USA
| | - Heather Schmitt
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | - Phyllis Summerfelt
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Adam M Dubis
- Department of Ophthalmology, University College London, London, UK
| | - Betsy Abroe
- College of Nursing, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Thomas B Connor
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Joseph Carroll
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wendy Huddleston
- Department of Kinesiology, College of Health Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Mahsa Ranji
- Biophotonics Laboratory, Department of Computer and Electrical Engineering and Computer Science, Florida Atlantic University, Boca Ratan, FL, USA.
| | - Janis T Eells
- Department of Biomedical Sciences, Photobiomodulation Laboratory, College of Health Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
116
|
Expression of Pro-Angiogenic Markers Is Enhanced by Blue Light in Human RPE Cells. Antioxidants (Basel) 2020; 9:antiox9111154. [PMID: 33233546 PMCID: PMC7699675 DOI: 10.3390/antiox9111154] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
Inherited retinal dystrophies are characterized by photoreceptor death. Oxidative stress usually occurs, increasing vision loss, and oxidative damage is often reported in retinitis pigmentosa (RP). More than 300 genes have been reported as RP causing. In contrast, choroidal neovascularization (CNV) only occasionally develops in the late stages of RP. We herein study the regulation of RP causative genes that are likely linked to CNV onset under oxidative conditions. We studied how the endogenous adduct N-retinylidene-N-retinylethanolamine (A2E) affects the expression of angiogenic markers in human retinal pigment epithelium (H-RPE) cells and a possible correlation with RP-causing genes. H-RPE cells were exposed to A2E and blue light for 3 and 6h. By transcriptome analysis, genes differentially expressed between A2E-treated cells and untreated ones were detected. The quantification of differential gene expression was performed by the Limma R package. Enrichment pathway analysis by the FunRich tool and gene prioritization by ToppGene allowed us to identify dysregulated genes involved in angiogenesis and linked to RP development. Two RP causative genes, AHR and ROM1, can be associated with an increased risk of CNV development. Genetic analysis of RP patients affected by CNV will confirm this hypothesis.
Collapse
|
117
|
Alambiaga-Caravaca AM, Domenech-Monsell IM, Sebastián-Morelló M, Miranda M, Balaguer-Fernández C, Calatayud-Pascual A, Rodilla V, López-Castellano A. HPLC-UV analytical validation of a method for quantification of progesterone in ex vivo trans-corneal and trans-scleral diffusion studies. J Pharm Biomed Anal 2020; 193:113749. [PMID: 33217709 DOI: 10.1016/j.jpba.2020.113749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022]
Abstract
Progesterone (PG) diminishes free radical damage and thus can afford protection against oxidative stress affecting the retina. The therapeutic use of PG is limited because it is a highly hydrophobic steroid hormone with very low solubility in water. This is the main drawback for the therapeutic application of PG at ocular level. The aims of this study were: (i) to analyze if PG causes ocular irritation (ii) to validate a HPLC method to determine PG in ex vivo studies and (iii) to evaluate PG permeation through cornea and sclera. A high performance liquid chromatographic method was developed and validated to detect PG incorporated to β-cyclodextrin using a Waters Sunfire C18 (150 × 4.6 mm) reverse-phase column packed with 5 μm silica particles using a mobile phase consisted of a mixture of acetonitrile (ACN) and pure water 80:20 (v/v), pH 7.4. The limit of detection and the limit of quantification for 50 μL injection of PG were found to be 0.42 and 1.26 μg/mL, respectively. The calibration curve showed excellent linearity over the concentration range (0.5 μg/mL to 100 μg/mL). As proof of concept, ex-vivo experiments to investigate PG permeation through cornea and sclera with vertical diffusion cells were carried out to quantify PG diffusion. Ex vivo experiments demonstrate its applicability to investigate permeation levels of PG from 6.57 ± 0.37 μg/cm2 at cornea and 8.13 ± 0.85 μg/cm2 sclera. In addition, at the end of diffusion studies the amount of PG retained in each tissue was also quantified, and it was 40.87 ± 9.84 μg/cm2 (mean ± SD; n = 6) in cornea and 56.11 ± 16.67 μg/cm2 (mean ± SD; n = 6) in sclera.
Collapse
Affiliation(s)
- Adrián M Alambiaga-Caravaca
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain
| | - Iris M Domenech-Monsell
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain
| | - María Sebastián-Morelló
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain
| | - María Miranda
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain; Departamentos de Farmacia y #Ciencias Biomédicas, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain
| | - Cristina Balaguer-Fernández
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain
| | - Aracely Calatayud-Pascual
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain
| | - Vicent Rodilla
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain.
| | - Alicia López-Castellano
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115, Valencia, Spain.
| |
Collapse
|
118
|
Campochiaro PA, Iftikhar M, Hafiz G, Akhlaq A, Tsai G, Wehling D, Lu L, Wall GM, Singh MS, Kong X. Oral N-acetylcysteine improves cone function in retinitis pigmentosa patients in phase I trial. J Clin Invest 2020; 130:1527-1541. [PMID: 31805012 DOI: 10.1172/jci132990] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/03/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUNDIn retinitis pigmentosa (RP), rod photoreceptors degenerate from 1 of many mutations, after which cones are compromised by oxidative stress. N-acetylcysteine (NAC) reduces oxidative damage and increases cone function/survival in RP models. We tested the safety, tolerability, and visual function effects of oral NAC in RP patients.METHODSSubjects (n = 10 per cohort) received 600 mg (cohort 1), 1200 mg (cohort 2), or 1800 mg (cohort 3) NAC bid for 12 weeks and then tid for 12 weeks. Best-corrected visual acuity (BCVA), macular sensitivity, ellipsoid zone (EZ) width, and aqueous NAC were measured. Linear mixed-effects models were used to estimate the rates of changes during the treatment period.RESULTSThere were 9 drug-related gastrointestinal adverse events that resolved spontaneously or with dose reduction (maximum tolerated dose 1800 mg bid). During the 24-week treatment period, mean BCVA significantly improved at 0.4 (95% CI: 0.2-0.6, P < 0.001), 0.5 (95% CI: 0.3-0.7, P < 0.001), and 0.2 (95% CI: 0.02-0.4, P = 0.03) letters/month in cohorts 1, 2, and 3, respectively. There was no significant improvement in mean sensitivity over time in cohorts 1 and 2, but there was in cohort 3 (0.15 dB/month, 95% CI: 0.04-0.26). There was no significant change in mean EZ width in any cohort.CONCLUSIONOral NAC is safe and well tolerated in patients with moderately advanced RP and may improve suboptimally functioning macular cones. A randomized, placebo-controlled trial is needed to determine if oral NAC can provide long-term stabilization and/or improvement in visual function in patients with RP.TRIAL REGISTRATIONNCT03063021.FUNDINGMr. and Mrs. Robert Wallace, Mr. and Mrs. Jonathan Wallace, Rami and Eitan Armon, Marc Sumerlin, Cassandra Hanley, and Nacuity Pharmaceuticals, Inc.
Collapse
Affiliation(s)
- Peter A Campochiaro
- Wilmer Eye Institute and.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | - Xiangrong Kong
- Wilmer Eye Institute and.,Department of Biostatistics.,Department of Epidemiology, and.,Department of Health, Behavior and Society, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
119
|
Antioxidant and Biological Properties of Mesenchymal Cells Used for Therapy in Retinitis Pigmentosa. Antioxidants (Basel) 2020; 9:antiox9100983. [PMID: 33066211 PMCID: PMC7602011 DOI: 10.3390/antiox9100983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Both tissue repair and regeneration are a priority in regenerative medicine. Retinitis pigmentosa (RP), a complex retinal disease characterized by the progressive loss of impaired photoreceptors, is currently lacking effective therapies: this represents one of the greatest challenges in the field of ophthalmological research. Although this inherited retinal dystrophy is still an incurable genetic disease, the oxidative damage is an important pathogenetic element that may represent a viable target of therapy. In this review, we summarize the current neuroscientific evidence regarding the effectiveness of cell therapies in RP, especially those based on mesenchymal cells, and we focus on their therapeutic action: limitation of both oxidative stress and apoptotic processes triggered by the disease and promotion of cell survival. Cell therapy could therefore represent a feasible therapeutic option in RP.
Collapse
|
120
|
Iuliano L, Fogliato G, Corbelli E, Bandello F, Codenotti M. Blind patients in end-stage inherited retinal degeneration: multimodal imaging of candidates for artificial retinal prosthesis. Eye (Lond) 2020; 35:289-298. [PMID: 33037412 DOI: 10.1038/s41433-020-01188-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To characterize the imaging features of blind patients with end-stage inherited retinal degeneration (IRD) and to assess possible morpho-functional correlations. METHODS In this observational cross-sectional study, we reviewed the clinical data and multimodal imaging of 40 eyes of 21 blind (light perception or less) institutional patients affected by end-stage IRD screened for Alpha AMS (Retina Implant AG, Reutlingen, Germany) retinal prosthesis eligibility. Analysis was carried out using spectral-domain optical coherence tomography (SD-OCT), fluorescein angiography and fundus autofluorescence. RESULTS Among patients with IRD-related low vision, the extrapolated prevalence of the blind was roughly 10%, median age 60.4 years with a disease duration of 40.4 years, showing epiretinal membranes (80%), hyperreflective intraretinal nodules (90%) and the absence of the ellipsoid zone (77.5%) on SD-OCT examination. Cystoid macular oedema was present in 52.5% of eyes, the majority of which being of the microcystoid subtype (42.5%), while 37.5% of eyes also lacked outer and inner retinal layer segmentation. Disease duration was found to be predictive of disrupted retinal layers (P = 0.029) and microcystoid macular oedema (P = 0.035), which was also more frequent in eyes without light perception (P = 0.013). CONCLUSIONS Eyes without vision due to end-stage IRD have a typical imaging pattern, predominantly characterized by epiretinal membranes, hyperreflective intraretinal nodules and the absence of the ellipsoid zone. Furthermore, microcystoid macular oedema and retinal layer disruption may be considered as signs of longstanding disease.
Collapse
Affiliation(s)
- Lorenzo Iuliano
- Department of Ophthalmology, Vita-Salute University, San Raffaele Scientific Institute, Milan, Italy.
| | - Giovanni Fogliato
- Department of Ophthalmology, Vita-Salute University, San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Corbelli
- Department of Ophthalmology, Vita-Salute University, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Bandello
- Department of Ophthalmology, Vita-Salute University, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Codenotti
- Department of Ophthalmology, Vita-Salute University, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
121
|
Bone Marrow-Derived Mononuclear Cell Transplants Decrease Retinal Gliosis in Two Animal Models of Inherited Photoreceptor Degeneration. Int J Mol Sci 2020; 21:ijms21197252. [PMID: 33008136 PMCID: PMC7583887 DOI: 10.3390/ijms21197252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Inherited photoreceptor degenerations are not treatable diseases and a frequent cause of blindness in working ages. In this study we investigate the safety, integration and possible rescue effects of intravitreal and subretinal transplantation of adult human bone-marrow-derived mononuclear stem cells (hBM-MSCs) in two animal models of inherited photoreceptor degeneration, the P23H-1 and the Royal College of Surgeons (RCS) rat. Immunosuppression was started one day before the injection and continued through the study. The hBM-MSCs were injected in the left eyes and the animals were processed 7, 15, 30 or 60 days later. The retinas were cross-sectioned, and L- and S- cones, microglia, astrocytes and Müller cells were immunodetected. Transplantations had no local adverse effects and the CD45+ cells remained for up to 15 days forming clusters in the vitreous and/or a 2–3-cells-thick layer in the subretinal space after intravitreal or subretinal injections, respectively. We did not observe increased photoreceptor survival nor decreased microglial cell numbers in the injected left eyes. However, the injected eyes showed decreased GFAP immunoreactivity. We conclude that intravitreal or subretinal injection of hBM-MSCs in dystrophic P23H-1 and RCS rats causes a decrease in retinal gliosis but does not have photoreceptor neuroprotective effects, at least in the short term. However, this treatment may have a potential therapeutic effect that merits further investigation.
Collapse
|
122
|
Wang Q, Banerjee S, So C, Qiu C, Lam HIC, Tse D, Völgyi B, Pan F. Unmasking inhibition prolongs neuronal function in retinal degeneration mouse model. FASEB J 2020; 34:15282-15299. [PMID: 32985731 DOI: 10.1096/fj.202001315rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 11/11/2022]
Abstract
All neurodegenerative diseases involve a relatively long period of timeframe from the onset of the disease to complete loss of functions. Extending this timeframe, even at a reduced level of function, would improve the quality of life of patients with these devastating diseases. The retina, as the part of the central nervous system and a frequent site of many distressing neurodegenerative disease, provides an ideal model to investigate the feasibility of extending the functional timeframe through pharmacologic intervention. Retinitis Pigmentosa (RP) is a group of blinding diseases. Although the rate of progression and degree of visual loss varies, there is usually a prolonged time before patients totally lose their photoreceptors and vision. It is believed that inhibitory mechanisms are still intact and may become relatively strong after the gradual loss of photoreceptors in RP patients. Therefore, it is possible that light-evoked responses of retinal ganglion cells and visual information processes in retinal circuits could be "unmasked" by blocking these inhibitory mechanisms restoring some level of visual function. Our results indicate that if the inhibition in the inner retina was unmasked in the retina of the rd10 mouse (the well-characterized RP mimicking, clinically relevant mouse model), the light-evoked responses of many retinal ganglion cells can be induced and restore their normal light sensitivity. GABA A receptor plays a major role in this masking inhibition. ERG b-wave and behavioral tests of spatial vision partly recovered after the application of PTX. Hence, removing retinal inhibition unmasks signalling mediated by surviving cones, thereby restoring some degree of visual function. These results may offer a novel strategy to restore the visual function with the surviving cones in RP patients and other gradual and progressive neurodegenerative diseases.
Collapse
Affiliation(s)
- Qin Wang
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Seema Banerjee
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chunghim So
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chunting Qiu
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Hang-I Christie Lam
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Dennis Tse
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Béla Völgyi
- Department of Experimental Zoology and Neurobiology, Szentágothai Research Centre, MTA NAP Retinal Electrical Synapses Research Group, University of Pécs, Pécs, Hungary
| | - Feng Pan
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong.,The Centre for Eye and Vision Research, Hong Kong
| |
Collapse
|
123
|
Murakami Y, Nakabeppu Y, Sonoda KH. Oxidative Stress and Microglial Response in Retinitis Pigmentosa. Int J Mol Sci 2020; 21:ijms21197170. [PMID: 32998461 PMCID: PMC7583782 DOI: 10.3390/ijms21197170] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 09/27/2020] [Indexed: 12/28/2022] Open
Abstract
An imbalance between the production of reactive oxygen species (ROS) and anti-oxidant capacity results in oxidative injury to cellular components and molecules, which in turn disturbs the homeostasis of cells and organs. Although retinitis pigmentosa (RP) is a hereditary disease, non-genetic biological factors including oxidative stress also modulate or contribute to the disease progression. In animal models of RP, the degenerating retina exhibits marked oxidative damage in the nucleic acids, proteins, and lipids, and anti-oxidant treatments substantially suppress photoreceptor cell death and microgliosis. Although the mechanisms by which oxidative stress mediates retinal degeneration have not been fully elucidated, our group has shown that oxidative DNA damage and its defense system are key regulators of microglial activation and photoreceptor degeneration in RP. In this review, we summarize the current evidence regarding oxidative stress in animal models and patients with RP. The clinical efficacy of anti-oxidant treatments for RP has not been fully established. Nevertheless, elucidating key biological processes that underlie oxidative damage in RP will be pivotal to understanding the pathology and developing a potent anti-oxidant strategy that targets specific cell types or molecules under oxidative stress.
Collapse
Affiliation(s)
- Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
- Correspondence: ; Tel.: +81-92-642-5648; Fax: +81-92-642-5663
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan;
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| |
Collapse
|
124
|
Tauroursodeoxycholic Acid Protects Retinal Pigment Epithelial Cells from Oxidative Injury and Endoplasmic Reticulum Stress In Vitro. Biomedicines 2020; 8:biomedicines8090367. [PMID: 32967221 PMCID: PMC7555559 DOI: 10.3390/biomedicines8090367] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal degeneration is characterized by the dysfunction of retinal cells. Oxidative and endoplasmic reticulum (ER) stress play an important role in the pathogenesis and progression of retinal degeneration. Tauroursodeoxycholic acid (TUDCA) has been demonstrated to have protective effects in in vitro and in vivo retinal degeneration models. To fully understand the molecular mechanisms of TUDCA’s protection, we first treated human retinal pigment epithelial (RPE) cells, ARPE-19, with H2O2 or H2O2 plus TUDCA for 24 h. RPE cells co-exposed to TUDCA had higher cell viability and lower cell death rate compared to cells exposed to H2O2 alone. TUDCA significantly increased antioxidant capacity in H2O2-treated RPE cells by decreasing the generation of reactive oxygen species (ROS) and Malondialdehyde (MDA), upregulating the expression of antioxidant genes, and increasing the generation of glutathione (GSH). TUDCA also inhibited inflammation in H2O2-challenged RPE cells by decreasing the expression of proinflammatory cytokines. Furthermore, TUDCA suppressed thapsigargin-induced ER stress in RPE cells, as demonstrated by decreased the expression of CCAAT-enhancer-binding protein homologous protein (CHOP) and apoptosis. Our present study suggests that TUDCA can protect RPE cells against oxidative damage, inflammation, and ER stress and may benefit patients with retinal degeneration.
Collapse
|
125
|
Chan TC, Wilkinson Berka JL, Deliyanti D, Hunter D, Fung A, Liew G, White A. The role of reactive oxygen species in the pathogenesis and treatment of retinal diseases. Exp Eye Res 2020; 201:108255. [PMID: 32971094 DOI: 10.1016/j.exer.2020.108255] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) normally play an important physiological role in health regulating cellular processes and signal transduction. The amount of ROS is usually kept in fine balance with the generation of ROS largely being offset by the body's antioxidants. A tipping of this balance has increasingly been recognised as a contributor to human disease. The retina, as a result of its cellular anatomy and physical location, is a potent generator of ROS that has been linked to several major retinal diseases. This review will provide a summary of the role of oxidative stress in the pathogenesis of diabetic retinopathy, age-related macular degeneration, myopia, retinal vein occlusion, retinitis pigmentosa and retinopathy of prematurity. Therapies aimed at controlling oxidative stress in these diseases are also examined.
Collapse
Affiliation(s)
- Thomas Cw Chan
- Discipline of Ophthalmology and Eye Health, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Jennifer L Wilkinson Berka
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Medical Building 181, Grattan Street, Parkville, Victoria, 3010, Australia; Department of Diabetes, The Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Devy Deliyanti
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Medical Building 181, Grattan Street, Parkville, Victoria, 3010, Australia; Department of Diabetes, The Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Damien Hunter
- Discipline of Ophthalmology and Eye Health, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; Centre for Vision Research, Westmead Institute of Medical Research, New South Wales, Australia
| | - Adrian Fung
- Westmead and Central Clinical Schools, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; Faculty of Medicine Health and Human Sciences, Macquarie University, New South Wales, Australia; Save Sight Institute, 8 Macquarie St, Sydney, Australia
| | - Gerald Liew
- Discipline of Ophthalmology and Eye Health, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; Centre for Vision Research, Westmead Institute of Medical Research, New South Wales, Australia
| | - Andrew White
- Discipline of Ophthalmology and Eye Health, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; Centre for Vision Research, Westmead Institute of Medical Research, New South Wales, Australia; Save Sight Institute, 8 Macquarie St, Sydney, Australia; Personal Eyes, Level 6, 34 Charles St, Parramatta, 2150, Australia.
| |
Collapse
|
126
|
Novel Insights into Beta 2 Adrenergic Receptor Function in the rd10 Model of Retinitis Pigmentosa. Cells 2020; 9:cells9092060. [PMID: 32917020 PMCID: PMC7563182 DOI: 10.3390/cells9092060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
Background: In retinitis pigmentosa (RP), inherited rod death is followed by cone loss and blindness. Why cones die is still a matter of consideration. Here, we investigate the pathogenic role of the sympathetic transmission in the rd10 mouse model of RP. Methods: Retinal levels of beta adrenergic receptor (BAR) 2 and norepinephrine (NE) were measured. After administration of the BAR1/2 blocker propranolol or the hypoxia-inducible factor (HIF)-1 activator dimethyloxalylglycine (DMOG), retinal levels of HIF-1α, BAR2 or proteins involved in BAR2 desensitization were also measured. In DMOG treated mice, expression and localization of BAR2, inflammatory markers and cone arrestin were determined. Finally, rd10 mice were subjected to electroretinogram (ERG) analysis to assess rod and cone function. Results: In the rd10 retina, BAR2 overexpression and NE accumulation were found, with BAR2 immunoreactivity localized to Müller cells. BAR2 overexpression was likely due to desensitization defects. Upregulated levels of BAR2 were drastically reduced by propranolol that also restored desensitization defects. Due to the low level of HIF-1 consequent to the hyperoxic environment in the rd10 retina, we hypothesized a link between HIF-1 and BAR2. HIF-1α stabilization with DMOG resulted in i. increased HIF-1α accumulation, ii. decreased BAR2 levels, iii. restored desensitization processes, iv. reduced expression of inflammatory markers and v. increased cone survival without improved retinal function. Conclusions: Our results support a pathogenic role of the sympathetic system in RP that might help to understand why rd10 mice show a positive response to BAR blockers.
Collapse
|
127
|
Olivares-González L, Velasco S, Millán JM, Rodrigo R. Intravitreal administration of adalimumab delays retinal degeneration in rd10 mice. FASEB J 2020; 34:13839-13861. [PMID: 32816354 DOI: 10.1096/fj.202000044rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
Retinitis pigmentosa (RP) is a group of inherited retinal dystrophies characterized by the progressive and irreversible loss of vision. We previously found that intraperitoneal administration of Adalimumab, a monoclonal anti-TNFα antibody, slowed down retinal degeneration in the murine model of RP, the rd10 mice. The aims of this study were to improve its neuroprotective effect and to deepen understanding of the molecular mechanisms involved in this effect. We analyzed (i) the in vitro effect of Adalimumab on the TNFα-mediated cell death in retinal cells; (ii) the effect of a single intravitreal injection of Adalimumab on retinal degeneration in rd10 mice at postnatal day (P) 23. In vitro studies showed that TNFα induced caspase and poly ADP ribose polymerase (PARP) activation, downregulation of (kinase receptor-interacting protein 1) RIPK1 and upregulation of RIPK3 in retinal cells. Adalimumab reduced cell death probably through the inhibition of caspase 3 activation. In vivo studies suggested that PARP and NLRP3 inflammasome are mainly activated and to a lesser extent caspase-dependent mechanisms in rd10 retinas at P23. Necroptosis seems to be inhibited by the downregulation of RIPK1. Adalimumab prevented from retinal degeneration without affecting caspase -dependent mechanisms but decreasing PARP activation, microglia activation as well as NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lorena Olivares-González
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center, Valencia, Spain
| | - Sheyla Velasco
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center, Valencia, Spain
| | - José María Millán
- Rare Diseases Networking Biomedical Research Centre (CIBERER), Madrid, Spain.,Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain.,Molecular, Cellular and Genomic Biomedicine, Health Research Institute La Fe, Valencia, Spain
| | - Regina Rodrigo
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center, Valencia, Spain.,Rare Diseases Networking Biomedical Research Centre (CIBERER), Madrid, Spain.,Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| |
Collapse
|
128
|
Limoli PG, Limoli CSS, Morales MU, Vingolo EM. Mesenchymal stem cell surgery, rescue and regeneration in retinitis pigmentosa: clinical and rehabilitative prognostic aspects. Restor Neurol Neurosci 2020; 38:223-237. [PMID: 32310198 PMCID: PMC7504992 DOI: 10.3233/rnn-190970] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose: To assess whether treatment with the Limoli Retinal Restoration Technique (LRRT) can be performed in patients with retinitis pigmentosa (RP), grafting the autologous cells in a deep scleral pocket above the choroid of each eye to exert their beneficial effect on the residual retinal cells. Methods: The patients were subjected to a complete ophthalmological examination, including best corrected visual acuity (BCVA), close-up visus measurements, spectral domain-optical coherence tomography (SD-OCT), microperimetry (MY), and electroretinography (ERG). Furthermore, the complete ophthalmological examination was carried out at baseline (T0) and at 6 months (T180) after surgery. The Shapiro–Wilk test was used to assess the normality of distribution of the investigated parameters. A mixed linear regression model was used to analyse the difference in all the studied parameters at T0 and T180, and to compare the mean change between the two groups. All statistical analyses were performed with STATA 14.0 (Collage Station, Texas, USA). Results: LRRT treatment was performed in 34 eyes of 25 RP patients recruited for the study. The eyes were classified in two groups on the basis of foveal thickness (FT) assessed by SD-OCT: 14 eyes in Group A (FT≤190μm) and the remaining 20 ones in Group B (FT > 190μm). Although it had not reached the statistical significance, Group B showed a better improvement in BCVA, residual close-up visus and sensitivity than Group A. Conclusions: Previous studies have described the role of LRRT in slowing down retinal degenerative diseases. Consequently, this surgical procedure could improve the clinical and rehabilitative prognostic parameters in RP patients. On the other hand, further clinical research and studies with longer follow-up will be needed to evaluate its efficacy.
Collapse
Affiliation(s)
| | | | - Marco Ulises Morales
- Division of Clinical Neurosciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Enzo Maria Vingolo
- Department of Sense Organs, Faculty of Medicine and Odontology, Sapienza University of Rome, p.le A. Moro, Rome, Italy
| |
Collapse
|
129
|
Yao A, Wijngaarden P. Metabolic pathways in context:
mTOR
signalling in the retina and optic nerve ‐ A review. Clin Exp Ophthalmol 2020; 48:1072-1084. [DOI: 10.1111/ceo.13819] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/21/2020] [Accepted: 07/05/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Anthony Yao
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital East Melbourne, Victoria Australia
| | - Peter Wijngaarden
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital East Melbourne, Victoria Australia
- Ophthalmology, Department of Surgery University of Melbourne Melbourne, Victoria Australia
| |
Collapse
|
130
|
Perdices L, Fuentes-Broto L, Segura F, Cuenca N, Orduna-Hospital E, Pinilla I. Epigallocatechin Gallate Slows Retinal Degeneration, Reduces Oxidative Damage, and Modifies Circadian Rhythms in P23H Rats. Antioxidants (Basel) 2020; 9:antiox9080718. [PMID: 32784376 PMCID: PMC7465727 DOI: 10.3390/antiox9080718] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/31/2022] Open
Abstract
Retinitis pigmentosa (RP) includes a group of genetic disorders that involve the loss of visual function due to mutations mainly in photoreceptors but also in other retinal cells. Apoptosis, retinal disorganization, and inflammation are common in the progression of the disease. Epigallocatechin gallate (EGCG) has been proved as beneficial in different eye diseases. Pigmented heterozygous P23H rat was used as an animal model of RP. Visual function was assessed by optomotor and electroretinogram (ERG) and circadian rhythms were evaluated by telemetry. Hepatic oxidative damage and antioxidant defenses were assessed using biochemical tests. The visual function of the EGCG P23H group was preserved, with a deterioration in the activity period and lower values in the interdaily stability parameter. Control rats treated with EGCG were less active than the sham group. EGCG increased antioxidant levels in P23H rats but reduced total hepatic antioxidant capacity by almost 42% in control rats compared to the sham group. We conclude that treatment with EGCG improves visual function and antioxidant status in P23H rats but diminishes antioxidant defenses in wild-type control animals, and slightly worsens activity circadian rhythms. Further studies are necessary to clarify the beneficial effects in disease conditions and in healthy organisms.
Collapse
Affiliation(s)
- Lorena Perdices
- Aragon Institute for Health Research (IIS Aragón), 50009 Zaragoza, Spain; (L.P.); (I.P.)
| | - Lorena Fuentes-Broto
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Universidad de Zaragoza, 50009 Zaragoza, Spain
- Correspondence: ; Tel.: +34-976-761-706
| | - Francisco Segura
- Department of Applied Physics, Universidad de Zaragoza, 50009 Zaragoza, Spain;
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 San Vicente del Raspeig, Alicante, Spain;
| | | | - Isabel Pinilla
- Aragon Institute for Health Research (IIS Aragón), 50009 Zaragoza, Spain; (L.P.); (I.P.)
- Department of Ophthalmology, Lozano Blesa University Hospital, 50009 Zaragoza, Spain
| |
Collapse
|
131
|
Santhi N, Ball DM. Applications in sleep: How light affects sleep. PROGRESS IN BRAIN RESEARCH 2020; 253:17-24. [PMID: 32771123 DOI: 10.1016/bs.pbr.2020.05.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Sleep is an active physiological state that plays a critical role in our physical and mental health and well-being. It is generated by a complex interplay between two oscillators, namely, the circadian oscillator and the sleep-wake homeostat. Sleep propensity is a function of wakefulness, that is, the longer one is awake the greater the homeostatic sleep pressure. Sleep onset occurs as a wake promoting circadian signal subsides, coinciding with an evening rise in melatonin and drop in core temperature. Light is one of the strongest time signals for the circadian oscillator. Poor sleep is a prevalent complaint today, attributable, in part, to our easy access to artificial light, especially after dusk. This non-visual effect of light is mediated by a multi-component photoreceptive system, consisting of rods, cones and melanopsin-expressing intrinsically-photosensitive retinal ganglion cells (ipRGC). Perhaps, with this available biological knowledge we can engineer artificial light to minimize its disruptive effect on sleep. We will highlight this by discussing circadian photoreception and its effect on sleep, in the blind population.
Collapse
Affiliation(s)
- Nayantara Santhi
- Department of Psychology, Northumbria University, Newcastle upon Tyne, England.
| | - Danny M Ball
- Institute of Cognitive Neuroscience, Department of Psychology, University College London, London, England
| |
Collapse
|
132
|
Oishi A, Noda K, Birtel J, Miyake M, Sato A, Hasegawa T, Miyata M, Numa S, Charbel Issa P, Tsujikawa A. Effect of smoking on macular function and retinal structure in retinitis pigmentosa. Brain Commun 2020; 2:fcaa117. [PMID: 33134916 PMCID: PMC7585699 DOI: 10.1093/braincomms/fcaa117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/16/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022] Open
Abstract
Retinitis pigmentosa is an inherited neurodegenerative disease of the retina. We investigated smoking as a modifiable environmental factor for the progression of this currently untreatable disease. Clinical data, smoking history, macular function and morphology including visual acuity, visual field sensitivity, ellipsoid zone width and central retinal thickness were investigated. Association between pack × years and these parameters were evaluated using generalized estimating equation models to adjust confounding factors such as age and sex. A total of 410 patients with retinitis pigmentosa (≥20 years old; 209 female) were included, 164 had a smoking history. Patients without smoking history revealed a better visual acuity than smokers (0.39 versus 0.57, P = 0.001). The pack × years index was associated with worse visual acuity and thinner central retinal thickness after adjusting for age and sex (P = 0.0047 and 0.0099, respectively). Visual field and ellipsoid zone width showed a non-significant decline with increasing pack × years. This study indicates an association of smoking with worse macular function and structural integrity in retinitis pigmentosa patients, and hence a potential detrimental effect of smoking on the disease course.
Collapse
Affiliation(s)
- Akio Oishi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
- Correspondence to: Akio Oishi, MD, PhD Department of Ophthalmology and Visual Sciences Kyoto University Graduate School of Medicine, Shogoin Kawahara-cho 54, Sakyo-ku, Kyoto 606-8507, Japan E-mail:
| | - Kazunori Noda
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Johannes Birtel
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Atsuyasu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Tomoko Hasegawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Manabu Miyata
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Shogo Numa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Peter Charbel Issa
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| |
Collapse
|
133
|
Zhao T, Liang Q, Meng X, Duan P, Wang F, Li S, Liu Y, Yin ZQ. Intravenous Infusion of Umbilical Cord Mesenchymal Stem Cells Maintains and Partially Improves Visual Function in Patients with Advanced Retinitis Pigmentosa. Stem Cells Dev 2020; 29:1029-1037. [PMID: 32679004 DOI: 10.1089/scd.2020.0037] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary retinal degeneration disease with no effective therapeutic approaches. Inflammatory and immune disorders are thought to play an important role in the pathogenesis of RP. Human umbilical cord mesenchymal stem cells (UCMSCs), with multiple biological functions such as anti-inflammation and immunoregulation, have been applied in different systemic diseases. We conducted a phase I/II clinical trial aiming to evaluate the safety and efficacy of intravenous administration of UCMSCs in advanced RP patients. All 32 subjects were intravenously infused with one dose of 108 UCMSCs and were followed up for 12 months. No serious local or systemic adverse effects occurred in the whole follow-up. Most patients improved their best corrected visual acuity (BCVA) in the first 3 months. The proportions of patients with improved or maintained BCVA were 96.9%, 95.3%, 93.8%, 95.4%, 90.6%, and 90.6% at the 1st, 2nd, 3rd, 6th, 9th, and 12th month follow-up, respectively. Most of the patients (81.3%) maintained or improved their visual acuities for 12 months. The average NEI VFQ-25 questionnaire scores were significantly improved at the third month (P < 0.05). The average visual field sensitivity and flash visual evoked potential showed no significant difference (P = 0.185, P = 0.711). Our results indicated that the intravenous infusion of UCMSCs was safe for advanced RP patients. Most of the patients improved or maintained their visual functions in a long term. The life qualities were improved significantly in the first 3 months, suggesting that the intravenous infusion of UCMSCs may be a promising therapeutic approach for advanced RP patients.
Collapse
Affiliation(s)
- Tongtao Zhao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qingling Liang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaohong Meng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ping Duan
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fang Wang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shiying Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
134
|
Greenwald SH, Brown EE, Scandura MJ, Hennessey E, Farmer R, Pawlyk BS, Xiao R, Vandenberghe LH, Pierce EA. Gene Therapy Preserves Retinal Structure and Function in a Mouse Model of NMNAT1-Associated Retinal Degeneration. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:582-594. [PMID: 32775493 PMCID: PMC7397406 DOI: 10.1016/j.omtm.2020.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022]
Abstract
No treatment is available for nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1)-associated retinal degeneration, an inherited disease that leads to severe vision loss early in life. Although the causative gene, NMNAT1, plays an essential role in nuclear nicotinamide adenine dinucleotide (NAD)+ metabolism in tissues throughout the body, NMNAT1-associated disease is isolated to the retina. Since this condition is recessive, supplementing the retina with a normal copy of NMNAT1 should protect vulnerable cells from disease progression. We tested this hypothesis in a mouse model that harbors the p.Val9Met mutation in Nmnat1 and consequently develops a retinal degenerative phenotype that recapitulates key features of the human disease. Gene augmentation therapy, delivered by subretinal injection of adeno-associated virus (AAV) carrying a normal human copy of NMNAT1, rescued retinal structure and function. Due to the early-onset profile of the phenotype, a rapidly activating self-complementary AAV was required to initiate transgene expression during the narrow therapeutic window. These data represent the first proof of concept for a therapy to treat patients with NMNAT1-associated disease.
Collapse
Affiliation(s)
- Scott H Greenwald
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Emily E Brown
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Michael J Scandura
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Erin Hennessey
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Raymond Farmer
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Basil S Pawlyk
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Ru Xiao
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.,Ocular Genomics Institute, Grousebeck Gene Therapy Center, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Luk H Vandenberghe
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.,Ocular Genomics Institute, Grousebeck Gene Therapy Center, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
135
|
Takeda A, Yanai R, Murakami Y, Arima M, Sonoda KH. New Insights Into Immunological Therapy for Retinal Disorders. Front Immunol 2020; 11:1431. [PMID: 32719682 PMCID: PMC7348236 DOI: 10.3389/fimmu.2020.01431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/03/2020] [Indexed: 12/24/2022] Open
Abstract
In the twentieth century, a conspicuous lack of effective treatment strategies existed for managing several retinal disorders, including age-related macular degeneration; diabetic retinopathy (DR); retinopathy of prematurity (ROP); retinitis pigmentosa (RP); uveitis, including Behçet's disease; and vitreoretinal lymphoma (VRL). However, in the first decade of this century, advances in biomedicine have provided new treatment strategies in the field of ophthalmology, particularly biologics that target vascular endothelial growth factor or tumor necrosis factor (TNF)-α. Furthermore, clinical trials on gene therapy specifically for patients with autosomal recessive or X-linked RP have commenced. The overall survival rates of patients with VRL have improved, owing to earlier diagnoses and better treatment strategies. However, some unresolved problems remain such as primary or secondary non-response to biologics or chemotherapy, and the lack of adequate strategies for treating most RP patients. In this review, we provide an overview of the immunological mechanisms of the eye under normal conditions and in several retinal disorders, including uveitis, DR, ROP, RP, and VRL. In addition, we discuss recent studies that describe the inflammatory responses that occur during the course of these retinal disorders to provide new insights into their diagnosis and treatment.
Collapse
Affiliation(s)
- Atsunobu Takeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Ophthalmology, Clinical Research Institute, Kyushu Medical Center, National Hospital Organization, Fukuoka, Japan
| | - Ryoji Yanai
- Department of Ophthalmology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
136
|
Stanković D, Claudius AK, Schertel T, Bresser T, Uhlirova M. A Drosophila model to study retinitis pigmentosa pathology associated with mutations in the core splicing factor Prp8. Dis Model Mech 2020; 13:dmm043174. [PMID: 32424050 PMCID: PMC7328144 DOI: 10.1242/dmm.043174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/04/2020] [Indexed: 12/30/2022] Open
Abstract
Retinitis pigmentosa (RP) represents genetically heterogeneous and clinically variable disease characterized by progressive degeneration of photoreceptors resulting in a gradual loss of vision. The autosomal dominant RP type 13 (RP13) has been linked to the malfunction of PRPF8, an essential component of the spliceosome. Over 20 different RP-associated PRPF8 mutations have been identified in human patients. However, the cellular and molecular consequences of their expression in vivo in specific tissue contexts remain largely unknown. Here, we establish a Drosophila melanogaster model for RP13 by introducing the nine distinct RP mutations into the fly PRPF8 ortholog prp8 and express the mutant proteins in precise spatiotemporal patterns using the Gal4/UAS system. We show that all nine RP-Prp8 mutant proteins negatively impact developmental timing, albeit to a different extent, when expressed in the endocrine cells producing the primary insect moulting hormone. In the developing eye primordium, uncommitted epithelial precursors rather than differentiated photoreceptors appeared sensitive to Prp8 malfunction. Expression of the two most pathogenic variants, Prp8S>F and Prp8H>R, induced apoptosis causing alterations to the adult eye morphology. The affected tissue mounted stress and cytoprotective responses, while genetic programs underlying neuronal function were attenuated. Importantly, the penetrance and expressivity increased under prp8 heterozygosity. In contrast, blocking apoptosis alleviated cell loss but not the redox imbalance. Remarkably, the pathogenicity of the RP-Prp8 mutations in Drosophila correlates with the severity of clinical phenotypes in patients carrying the equivalent mutations, highlighting the suitability of the Drosophila model for in-depth functional studies of the mechanisms underlying RP13 etiology.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Dimitrije Stanković
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Ann-Katrin Claudius
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Thomas Schertel
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Tina Bresser
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Mirka Uhlirova
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne 50931, Germany
| |
Collapse
|
137
|
Thompson DA, Iannaccone A, Ali RR, Arshavsky VY, Audo I, Bainbridge JWB, Besirli CG, Birch DG, Branham KE, Cideciyan AV, Daiger SP, Dalkara D, Duncan JL, Fahim AT, Flannery JG, Gattegna R, Heckenlively JR, Heon E, Jayasundera KT, Khan NW, Klassen H, Leroy BP, Molday RS, Musch DC, Pennesi ME, Petersen-Jones SM, Pierce EA, Rao RC, Reh TA, Sahel JA, Sharon D, Sieving PA, Strettoi E, Yang P, Zacks DN. Advancing Clinical Trials for Inherited Retinal Diseases: Recommendations from the Second Monaciano Symposium. Transl Vis Sci Technol 2020; 9:2. [PMID: 32832209 PMCID: PMC7414644 DOI: 10.1167/tvst.9.7.2] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/12/2020] [Indexed: 12/18/2022] Open
Abstract
Major advances in the study of inherited retinal diseases (IRDs) have placed efforts to develop treatments for these blinding conditions at the forefront of the emerging field of precision medicine. As a result, the growth of clinical trials for IRDs has increased rapidly over the past decade and is expected to further accelerate as more therapeutic possibilities emerge and qualified participants are identified. Although guided by established principles, these specialized trials, requiring analysis of novel outcome measures and endpoints in small patient populations, present multiple challenges relative to study design and ethical considerations. This position paper reviews recent accomplishments and existing challenges in clinical trials for IRDs and presents a set of recommendations aimed at rapidly advancing future progress. The goal is to stimulate discussions among researchers, funding agencies, industry, and policy makers that will further the design, conduct, and analysis of clinical trials needed to accelerate the approval of effective treatments for IRDs, while promoting advocacy and ensuring patient safety.
Collapse
Affiliation(s)
- Debra A Thompson
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alessandro Iannaccone
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC, USA
| | - Robin R Ali
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA.,Institute of Ophthalmology, University College London, London, UK
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC, USA
| | - Isabelle Audo
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, Paris, France.,CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | | | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Kari E Branham
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Artur V Cideciyan
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven P Daiger
- Human Genetics Center, School of Public Health, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Deniz Dalkara
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, Paris, France
| | - Jacque L Duncan
- Department of Ophthalmology, University of California-San Francisco, San Francisco, CA, USA
| | - Abigail T Fahim
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John G Flannery
- Helen Wills Neuroscience Institute, University of California-Berkeley, Berkeley, CA, USA
| | | | - John R Heckenlively
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elise Heon
- Department of Ophthalmology and Vision Sciences, Hospital for Sick Children, Toronto, Ontario, Canada
| | - K Thiran Jayasundera
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Naheed W Khan
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Henry Klassen
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California-Irvine, Irvine, CA, USA
| | - Bart P Leroy
- Department of Ophthalmology and Center Medical Genetics, Ghent University Hospital and University, Ghent, Belgium.,Division of Ophthalmology and Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert S Molday
- Department of Biochemistry/Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David C Musch
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science Center, Portland, OR, USA
| | - Simon M Petersen-Jones
- Small Animal Clinical Sciences, Michigan State University, College of Veterinary Medicine, East Lansing, MI, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Rajesh C Rao
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Jose A Sahel
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, Paris, France.,CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France.,Fondation Ophtalmologique Rothschild, Paris, France.,Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dror Sharon
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Paul A Sieving
- Department of Ophthalmology and Center for Ocular Regenerative Therapy, University of California-Davis School of Medicine, Sacramento, CA, USA.,National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Enrica Strettoi
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Paul Yang
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science Center, Portland, OR, USA
| | - David N Zacks
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | | |
Collapse
|
138
|
Zhang E, Ryu J, Levi SR, Oh JK, Hsu CW, Cui X, Lee TT, Wang NK, Lima de Carvalho JR, Tsang SH. PKM2 ablation enhanced retinal function and survival in a preclinical model of retinitis pigmentosa. Mamm Genome 2020; 31:77-85. [PMID: 32342224 DOI: 10.1007/s00335-020-09837-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/08/2020] [Indexed: 12/13/2022]
Abstract
Retinitis pigmentosa (RP) is a neurodegenerative disorder that causes irreversible vision loss in over 1.5 million individuals world-wide. The genetic heterogeneity of RP necessitates a broad therapy that is able to provide treatment in a gene- and mutation- non-specific manner. In this study, we identify the therapeutic benefits of metabolic reprogramming by targeting pyruvate kinase M2 (PKM2) in a Pde6β preclinical model of RP. The genetic contributions of PKM2 inhibition in retinal degeneration were evaluated through histology and electroretinogram (ERG) followed by a statistical analysis using a linear regression model. Notably, PKM2 ablation resulted in thicker retinal layers in Pde6β-mutated mice as compared to the controls, suggesting greater photoreceptor survival. Consistent with these anatomical findings, ERG analyses revealed that the maximum b-wave is on average greater in Pkm2 knockout mice than in mice with intact Pkm2, indicating enhanced photoreceptor function. These rescue phenotypes from Pkm2 ablation in a preclinical model of RP indicate that a metabolome reprogramming may be useful in treating RP.
Collapse
Affiliation(s)
- Ethan Zhang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Joseph Ryu
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Sarah R Levi
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Jin Kyun Oh
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
- State University of New York At Downstate Medical Center, Brooklyn, NY, USA
| | - Chun Wei Hsu
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Xuan Cui
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
- The College of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, Tianjin, China
| | - Ting-Ting Lee
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Nan-Kai Wang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Jose Ronaldo Lima de Carvalho
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
- Department of Ophthalmology, Empresa Brasileira de Servicos Hospitalares (EBSERH) - Hospital das Clinicas de Pernambuco (HCPE), Federal University of Pernambuco (UFPE), Recife, Brazil
- Department of Ophthalmology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.
- Department of Pathology & Cell Biology, Institute of Human Nutrition, and Columbia Stem Cell Initiative, Columbia University, New York, NY, USA.
| |
Collapse
|
139
|
Xu L, Yu H, Sun H, Yu X, Tao Y. Optimized nonionic emulsifier for the efficient delivery of astaxanthin nanodispersions to retina: in vivo and ex vivo evaluations. Drug Deliv 2020; 26:1222-1234. [PMID: 31747793 PMCID: PMC6882443 DOI: 10.1080/10717544.2019.1682718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Astaxanthin (AST) is a naturally occurring carotenoid with potent anti-oxidative and anti-inflammatory potency against chronic diseases. In this study, we suspended AST in different nonionic emulsifiers to produce nanodispersions. The basic physicochemical properties of the produced AST nanodispersions were verified to select the optimized nonionic emulsifier. Among the tested emulsifiers, Polysorbate 20 produced the AST nanoemulsions with smaller particle diameters, narrower size distributions, and higher AST contents among these emulsifiers. The N-methyl-N-nitrosourea (MNU) administered mouse is a chemically induced retinal degeneration (RD) model with rapid progress rate. AST suspended in Polysorbate 20 was demonstrated to ameliorate the dramatic consequences of MNU on retina architectures and function in several different tests encompassing from electrophysiology to histology and molecular tests. Furthermore, the multi-electrodes array (MEA) was used to detect the firing activities of retinal ganglion cells within the inner retinal circuits. We found that AST nanodispersions could restrain the spontaneous firing response, enhance the light induced firing response, and preserve the basic configurations of visual signal pathway in degenerative retinas. The MEA assay provided an appropriate example to evaluate the potency of pharmacological compounds on retinal plasticity. In summary, emulsifier type affects the basic physicochemical characteristic of AST nanodispersions. Polysorbate 20 acts as an optimized nonionic emulsifier for the efficient delivery of AST nanodispersions to retina. AST nanodispersions can alleviate the photoreceptor loss and rectify the abnormities in visual signal transmission.
Collapse
Affiliation(s)
- Lei Xu
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Haixiang Yu
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongbin Sun
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiang Yu
- Department of Otorhinolaryngology, Jinling Hospital, Clinical Hospital of Medical College, Nanjing University, Nanjing, China
| | - Ye Tao
- Department of Physiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
140
|
Oxidative Stress, a Crossroad Between Rare Diseases and Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9040313. [PMID: 32326494 PMCID: PMC7222183 DOI: 10.3390/antiox9040313] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is an imbalance between production and accumulation of oxygen reactive species and/or reactive nitrogen species in cells and tissues, and the capacity of detoxifying these products, using enzymatic and non-enzymatic components, such as glutathione. Oxidative stress plays roles in several pathological processes in the nervous system, such as neurotoxicity, neuroinflammation, ischemic stroke, and neurodegeneration. The concepts of oxidative stress and rare diseases were formulated in the eighties, and since then, the link between them has not stopped growing. The present review aims to expand knowledge in the pathological processes associated with oxidative stress underlying some groups of rare diseases: Friedreich’s ataxia, diseases with neurodegeneration with brain iron accumulation, Charcot-Marie-Tooth as an example of rare neuromuscular disorders, inherited retinal dystrophies, progressive myoclonus epilepsies, and pediatric drug-resistant epilepsies. Despite the discrimination between cause and effect may not be easy on many occasions, all these conditions are Mendelian rare diseases that share oxidative stress as a common factor, and this may represent a potential target for therapies.
Collapse
|
141
|
Donato L, D’Angelo R, Alibrandi S, Rinaldi C, Sidoti A, Scimone C. Effects of A2E-Induced Oxidative Stress on Retinal Epithelial Cells: New Insights on Differential Gene Response and Retinal Dystrophies. Antioxidants (Basel) 2020; 9:E307. [PMID: 32290199 PMCID: PMC7222197 DOI: 10.3390/antiox9040307] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress represents one of the principal inductors of lifestyle-related and genetic diseases. Among them, inherited retinal dystrophies, such as age-related macular degeneration and retinitis pigmentosa, are well known to be susceptible to oxidative stress. To better understand how high reactive oxygen species levels may be involved in retinal dystrophies onset and progression, we performed a whole RNA-Seq experiment. It consisted of a comparison of transcriptomes' profiles among human retinal pigment epithelium cells exposed to the oxidant agent N-retinylidene-N-retinylethanolamine (A2E), considering two time points (3h and 6h) after the basal one. The treatment with A2E determined relevant differences in gene expression and splicing events, involving several new pathways probably related to retinal degeneration. We found 10 different clusters of pathways involving differentially expressed and differentially alternative spliced genes and highlighted the sub- pathways which could depict a more detailed scenario determined by the oxidative-stress-induced condition. In particular, regulation and/or alterations of angiogenesis, extracellular matrix integrity, isoprenoid-mediated reactions, physiological or pathological autophagy, cell-death induction and retinal cell rescue represented the most dysregulated pathways. Our results could represent an important step towards discovery of unclear molecular mechanisms linking oxidative stress and etiopathogenesis of retinal dystrophies.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (R.D.); (S.A.); (C.R.); (C.S.)
- Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T., 90139 Palermo, Italy
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (R.D.); (S.A.); (C.R.); (C.S.)
- Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T., 90139 Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (R.D.); (S.A.); (C.R.); (C.S.)
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98125 Messina, Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (R.D.); (S.A.); (C.R.); (C.S.)
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (R.D.); (S.A.); (C.R.); (C.S.)
- Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T., 90139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (R.D.); (S.A.); (C.R.); (C.S.)
- Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T., 90139 Palermo, Italy
| |
Collapse
|
142
|
Gorbatyuk MS, Starr CR, Gorbatyuk OS. Endoplasmic reticulum stress: New insights into the pathogenesis and treatment of retinal degenerative diseases. Prog Retin Eye Res 2020; 79:100860. [PMID: 32272207 DOI: 10.1016/j.preteyeres.2020.100860] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
Physiological equilibrium in the retina depends on coordinated work between rod and cone photoreceptors and can be compromised by the expression of mutant proteins leading to inherited retinal degeneration (IRD). IRD is a diverse group of retinal dystrophies with multifaceted molecular mechanisms that are not fully understood. In this review, we focus on the contribution of chronically activated unfolded protein response (UPR) to inherited retinal pathogenesis, placing special emphasis on studies employing genetically modified animal models. As constitutively active UPR in degenerating retinas may activate pro-apoptotic programs associated with oxidative stress, pro-inflammatory signaling, dysfunctional autophagy, free cytosolic Ca2+ overload, and altered protein synthesis rate in the retina, we focus on the regulatory mechanisms of translational attenuation and approaches to overcoming translational attenuation in degenerating retinas. We also discuss current research on the role of the UPR mediator PERK and its downstream targets in degenerating retinas and highlight the therapeutic benefits of reprogramming PERK signaling in preclinical animal models of IRD. Finally, we describe pharmacological approaches targeting UPR in ocular diseases and consider their potential applications to IRD.
Collapse
Affiliation(s)
- Marina S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA.
| | - Christopher R Starr
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| | - Oleg S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| |
Collapse
|
143
|
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: 2.2] [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
|
144
|
Empowering Retinal Gene Therapy with a Specific Promoter for Human Rod and Cone ON-Bipolar Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:505-519. [PMID: 32258214 PMCID: PMC7114634 DOI: 10.1016/j.omtm.2020.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Optogenetic gene therapy holds promise to restore high-quality vision in blind patients and recently reached clinical trials. Although the ON-bipolar cells, the first retinal interneurons, make the most attractive targets for optogenetic vision restoration, they have remained inaccessible to human gene therapy due to the lack of a robust cell-specific promoter. We describe the design and functional evaluation of 770En_454P(hGRM6), a human GRM6 gene-derived, short promoter that drives strong and highly specific expression in both the rod- and cone-type ON-bipolar cells of the human retina. Expression also in cone-type ON-bipolar cells is of importance, since the cone-dominated macula mediates high-acuity vision and is the primary target of gene therapies. 770En_454P(hGRM6)-driven middle-wave opsin expression in ON-bipolar cells achieved lasting restoration of high visual acuity in the rd1 mouse model of late retinal degeneration. The new promoter enables precise manipulation of the inner retinal network and paves the way for clinical application of gene therapies for high-resolution optogenetic vision restoration, raising hopes of significantly improving the life quality of people suffering from blindness.
Collapse
|
145
|
Xiao H, Wang J, Saul A, Smith SB. Comparison of Neuroprotective Effects of Monomethylfumarate to the Sigma 1 Receptor Ligand (+)-Pentazocine in a Murine Model of Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2020; 61:5. [PMID: 32150247 PMCID: PMC7401726 DOI: 10.1167/iovs.61.3.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose Activating the cell survival modulator sigma 1 receptor (Sig1R) delays cone photoreceptor cell loss in Pde6βrd10/J (rd10) mice, a model of retinitis pigmentosa. Beneficial effects are abrogated in rd10 mice lacking NRF2, implicating NRF2 as essential to Sig1R-mediated cone neuroprotection. Here we asked whether activation of NRF2 alone is sufficient to rescue cones in rd10 mice. Methods Expression of antioxidant genes was evaluated in 661W cells and in mouse retinas after treatment with monomethylfumarate (MMF), a potent NRF2 activator. Rd10 mice were administered MMF (50 mg/kg) or the Sig1R ligand (+)-pentazocine (PTZ; 0.5 mg/kg) intraperitoneally (every other day, P14-42). Mice were evaluated for visual acuity (optokinetic tracking response), retinal function (electroretinography) and architecture (SD-OCT); histologic retinal sections were evaluated morphometrically. Results MMF treatment increased Nrf2, Nqo1, Cat, Sod1, and Hmox1 expression in vitro and in vivo. Visual acuity of (+)-PTZ-treated rd10 mice was similar to wild-type mice; however, MMF treatment did not alter acuity compared with nontreated rd10 mice. Cone electroretinography b-wave amplitudes were greater in PTZ-treated than nontreated or MMF-treated rd10 mice. SD-OCT assessment of retinal thickness was greater in (+)-PTZ-treated mice versus nontreated or MMF-treated rd10 mice. Morphometric assessment of the outer nuclear layer revealed approximately 18 cells/100 µm retinal length in (+)-PTZ-treated rd10 mice, but only approximately 10 to 12 cells/100 µm in MMF-treated and nontreated rd10 retinas. Conclusions Activation of NRF2 using MMF, at least at our dosing regimen, is insufficient to attenuate catastrophic photoreceptor damage characteristic of rd10 mice. The data prompt investigation of additional mechanisms involved in Sig1R-mediated retinal neuroprotection.
Collapse
Affiliation(s)
- Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia,United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia,United States
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia,United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia,United States
| | - Alan Saul
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia,United States
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia,United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia,United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia,United States
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia,United States
| |
Collapse
|
146
|
Tao Y, Cai L, Zhou D, Wang C, Ma Z, Dong X, Peng G. CoPP-Induced-Induced HO-1 Overexpression Alleviates Photoreceptor Degeneration With Rapid Dynamics: A Therapeutic Molecular Against Retinopathy. Invest Ophthalmol Vis Sci 2020; 60:5080-5094. [PMID: 31825462 DOI: 10.1167/iovs.19-26876] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) causes progressive photoreceptor degeneration in the retina. The N-methyl-N-nitrosourea (MNU)-administered mouse is used as a chemically induced RP model with rapid progression rate. This study was designed to study heme oxygenase-1 (HO-1) expression in the MNU-administered mice, and to explore the therapeutic effects of cobalt protoporphyrin (CoPP). Methods The HO-1 expression in the retina of MNU-administered mice was analyzed. CoPP was injected intravenously into the MNU-administered mice. Subsequently, the CoPP-treated mice were subjected to functional and morphologic examinations. Results HO-1 was involved in the MNU-induced photoreceptor degeneration. CoPP treatment enhanced retinal HO-1 expression in the MNU-administered mice. Electroretinogram (ERG) examination and behavioral tests showed that CoPP treatment improved the retinal responsiveness of MNU-administered mice. Histologic analysis and optical coherence tomography (OCT) examination showed that retinal architecture of the CoPP-treated mice was more intact than that of the MNU+vehicle group. Cone photoreceptors in the MNU-administered mice were rescued efficiently by CoPP treatment. Furthermore, multielectrode array (MEA) recording showed that CoPP treatment mitigated the spontaneous firing response, enhanced the light-induced firing response, and preserved the basic configurations of visual signal pathway in the MNU-administered mice. Mechanism studies suggested that CoPP afforded these therapeutic effects by modulating the apoptosis cascades and alleviating the oxidative stress in degenerative retinas. Conclusions CoPP alleviated photoreceptor degeneration and rectified the signaling abnormities in MNU-administered mice. CoPP may serve as a potential medication against degenerative retinopathy.
Collapse
Affiliation(s)
- Ye Tao
- Lab of Visual Cell Differentiation and Modulation, Department of Physiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Lun Cai
- Department of Neurosurgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dawei Zhou
- Department of Traditional Chinese Medicine, 967(210) Hospital of Chinese People's Liberation Army, Dalian, China
| | - Chunhui Wang
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhao Ma
- Department of Neurosurgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Dong
- Department of Ophthalmology, 967(210) Hospital of Chinese People's Liberation Army, Dalian, China
| | - Guanghua Peng
- Lab of Visual Cell Differentiation and Modulation, Department of Physiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
147
|
Yan W, Long P, Wei D, Yan W, Zheng X, Chen G, Wang J, Zhang Z, Chen T, Chen M. Protection of retinal function and morphology in MNU-induced retinitis pigmentosa rats by ALDH2: an in-vivo study. BMC Ophthalmol 2020; 20:55. [PMID: 32070320 PMCID: PMC7027227 DOI: 10.1186/s12886-020-1330-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 01/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background Retinitis pigmentosa (RP) is a kind of inherited retinal degenerative diseases characterized by the progressive loss of photoreceptors. RP has been a conundrum without satisfactory countermeasures in clinic until now. Acetaldehyde dehydrogenase 2 (ALDH2), a major enzyme involved in aldehyde detoxification, has been demonstrated to be beneficial for a growing number of human diseases, such as cardiovascular dysfunction, diabetes mellitus and neurodegeneration. However, its protective effect against RP remains unknown. Our study explored the impact of ALDH2 on retinal function and structure in N-methyl-N-nitrosourea (MNU)-induced RP rats. Methods Rats were gavaged with 5 mg/kg Alda-1, an ALDH2 agonist, 5 days before and 3 days after MNU administration. Assessments of retinal function and morphology as well as measurement of specific proteins expression level were conducted. Results Electroretinogram recordings showed that Alda-1 administration alleviated the decrease in amplitude caused by MNU, rendering protection of retinal function. Mitigation of photoreceptor degeneration in MNU-treated retinas was observed by optical coherence tomography and retinal histological examination. In addition, Western blotting results revealed that ALDH2 protein expression level was upregulatedwith increased expression of SIRT1 protein after the Alda-1 intervention. Besides, endoplasmic reticulum stress (ERS) was reduced according to the significant downregulation of GRP78 protein, while apoptosis was ameliorated as shown by the decreased expression of PARP1 protein. Conclusions Together, our data demonstrated that ALDH2 could provide preservation of retinal function and morphology against MNU-induced RP, with the underlying mechanism at least partly related to the modulation of SIRT1, ERS and apoptosis.
Collapse
Affiliation(s)
- Weiming Yan
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, 350025, China.,Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Pan Long
- Department of Ophthalmology, The West General Hospital of Chinese PLA, Chendu, 610083, China
| | - Dongyu Wei
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Weihua Yan
- Tong'an No.1 High School of Fujian Province, Xiamen, 361100, China
| | - Xiangrong Zheng
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, 350025, China
| | - Guocang Chen
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, 350025, China
| | - Jiancong Wang
- BeiJing HealthOLight Technology Co. Ltd, Beijing, 10010, China
| | - Zuoming Zhang
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Tao Chen
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Meizhu Chen
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, 350025, China.
| |
Collapse
|
148
|
Li C, Tian Y, Yao A, Zha X, Zhang J, Tao Y. Intravitreal Delivery of Melatonin Is Protective Against the Photoreceptor Loss in Mice: A Potential Therapeutic Strategy for Degenerative Retinopathy. Front Pharmacol 2020; 10:1633. [PMID: 32116667 PMCID: PMC7028754 DOI: 10.3389/fphar.2019.01633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Melatonin is a circadian hormone with potent cytoprotective effects. Retinitis pigmentosa (RP) comprises a heterogeneous group of inherent retinopathies that characterized by the photoreceptor death in bilateral eyes. The N-methyl-N-nitrosourea (MNU) administered mouse is a type of chemically induced RP model with rapid progressive rate. We intend to study the melatonin mediated effects on the MNU administered mice. Melatonin was delivered into the vitreous body of the MNU administered mice. Subsequently, the melatonin treated mice were subjected to histological analysis, optokinetic behavior tests, spectral-domain optical coherence tomography (SD-OCT), and electroretinogram (ERG) examination. Multi-electrodes array (MEA) was used to analyze the status of visual signal transmission within retinal circuits. Biochemical analysis was performed to quantify the expression levels of antioxidative enzymes, oxidative stress markers, and apoptotic factors in the retinas. The intravitreal injection of melatonin ameliorated effectively the MNU induced photoreceptor degeneration. Melatonin therapy mitigated the spontaneous firing response, and preserved the basic configurations of visual signal pathway in MNU administered mice. MEA is effective to evaluate the pharmacological effects on retina. Of note, the cone photoreceptors in degenerative retinas were rescued efficiently by melatonin therapy. Melatonin afforded these protective effects by modulating the apoptotic cascades and alleviating the oxidative stress. These findings suggest that melatonin could act as an alternative treatment for degenerative retinopathy. Melatonin might be used in combination with other therapeutic approaches to alleviate the photoreceptor loss and preserve the visual function of RP patients.
Collapse
Affiliation(s)
- Chong Li
- Department of Neurosurgery, PLA General Hospital, Beijing, China
| | - Yi Tian
- Department of Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Anhui Yao
- Department of Neurosurgery, PLA General Hospital, Beijing, China
| | - Xiaobing Zha
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianbin Zhang
- Department of Occupational and Environmental Health, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Ye Tao
- Department of Ophthalmology, Henan Provincial People’s Hospital, Zhengzhou University, People’s Hospital, Zhengzhou, China
| |
Collapse
|
149
|
Perez-Cervantes C, Smith LA, Nadadur RD, Hughes AEO, Wang S, Corbo JC, Cepko C, Lonfat N, Moskowitz IP. Enhancer transcription identifies cis-regulatory elements for photoreceptor cell types. Development 2020; 147:dev184432. [PMID: 31915147 PMCID: PMC7033740 DOI: 10.1242/dev.184432] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/13/2019] [Indexed: 12/30/2022]
Abstract
Identification of cell type-specific cis-regulatory elements (CREs) is crucial for understanding development and disease, although identification of functional regulatory elements remains challenging. We hypothesized that context-specific CREs could be identified by context-specific non-coding RNA (ncRNA) profiling, based on the observation that active CREs produce ncRNAs. We applied ncRNA profiling to identify rod and cone photoreceptor CREs from wild-type and mutant mouse retinas, defined by presence or absence, respectively, of the rod-specific transcription factor (TF) NrlNrl-dependent ncRNA expression strongly correlated with epigenetic profiles of rod and cone photoreceptors, identified thousands of candidate rod- and cone-specific CREs, and identified motifs for rod- and cone-specific TFs. Colocalization of NRL and the retinal TF CRX correlated with rod-specific ncRNA expression, whereas CRX alone favored cone-specific ncRNA expression, providing quantitative evidence that heterotypic TF interactions distinguish cell type-specific CRE activity. We validated the activity of novel Nrl-dependent ncRNA-defined CREs in developing cones. This work supports differential ncRNA profiling as a platform for the identification of cell type-specific CREs and the discovery of molecular mechanisms underlying TF-dependent CRE activity.
Collapse
Affiliation(s)
- Carlos Perez-Cervantes
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Linsin A Smith
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Rangarajan D Nadadur
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Andrew E O Hughes
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sui Wang
- Departments of Genetics and Ophthalmology, Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph C Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Constance Cepko
- Departments of Genetics and Ophthalmology, Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nicolas Lonfat
- Departments of Genetics and Ophthalmology, Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ivan P Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
150
|
Peng JJ, Song WT, Yao F, Zhang X, Peng J, Luo XJ, Xia XB. Involvement of regulated necrosis in blinding diseases: Focus on necroptosis and ferroptosis. Exp Eye Res 2020; 191:107922. [PMID: 31923413 DOI: 10.1016/j.exer.2020.107922] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/17/2019] [Accepted: 01/05/2020] [Indexed: 12/15/2022]
Abstract
Besides apoptosis, necrosis can also occur in a highly regulated and genetically controlled manner, defined as regulated necrosis, which is characterized by a loss of cell membrane integrity and release of cytoplasmic content. Depending on the involvement of its signal pathway, regulated necrosis can be further classified as necroptosis, ferroptosis, pyroptosis and parthanatos. Numerous studies have demonstrated that regulated necrosis is involved in the pathogenesis of many diseases covering almost all organs including the brain, heart, liver, kidney, intestine, blood vessel, eye and skin, particularly myocardial infarction and stroke. Most recently, growing evidence suggests that multiple types of regulated necrosis contribute to the degeneration of retinal ganglion cells, retinal pigment epithelial cells or photoreceptor cells, which are the main pathologic features for glaucoma, age-related macular degeneration or retinitis pigmentosa, respectively. This review focuses on the involvement of necroptosis and ferroptosis in these blinding diseases.
Collapse
Affiliation(s)
- Jing-Jie Peng
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China; Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Wei-Tao Song
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Fei Yao
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xuan Zhang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Xiao-Bo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|