1
|
Lim HY, Joo K. Clinical and Genetic Characteristics of Patients with Peripheral Retinal Flecks in Koreans. KOREAN JOURNAL OF OPHTHALMOLOGY 2024; 38:461-470. [PMID: 39434579 DOI: 10.3341/kjo.2024.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
PURPOSE To describe the clinical and genetic features of Korean patients with peripheral retinal flecks unrelated to aging. METHODS A retrospective analysis was conducted on the clinical characteristics of patients with symmetric peripheral retinal flecks. Age-related deposits such as reticular pseudodrusen were excluded, as well as secondary deposits related to intraocular inflammation, tumor, and drug toxicity. Multimodal imaging, electrophysiological examinations, and genetic testing were analyzed. RESULTS A total of 10 patients (two men and eight women) with bilateral peripheral flecks were enrolled in this study. A mean age at diagnosis was 30.5 ± 19.6 years (range, 4-59 years). Within the 10 patients, six were genetically confirmed with monogenic retinal disorders. Biallelic pathogenic variants in RDH5 were found in five patients, and one patient was diagnosed with retinopathy related to Alport syndrome due to a pathogenic variant in COL4A5. Although not genetically confirmed, one case associated with nanophthalmos and another case showing chorioretinal mottling in a carrier of ocular albinism have been identified. In one patient, genetic testing also revealed unknown causes. The mean logarithm of the minimum angle of resolution initial visual acuity was 0.12 ± 0.18 and 0.07 ± 0.18 in right and left eyes, respectively. Night blindness was reported by four patients (40%), with three showing decreased or delayed rod response in electroretinogram, particularly those with RDH5 mutations. Differences in the deposit layers and the patterns of flecks were observed on multimodal imaging. CONCLUSIONS In the study population, we observed various causes and clinical differences in the retinal fleck patterns among Koreans, including RDH5-related fundus albipunctatus and Alport syndrome. Despite reports of night blindness symptoms in some cases, all patients demonstrated satisfactory corrected visual acuity.
Collapse
Affiliation(s)
- Hun Young Lim
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kwangsic Joo
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
2
|
Montaser AB, Gao F, Peters D, Vainionpää K, Zhibin N, Skowronska-Krawczyk D, Figeys D, Palczewski K, Leinonen H. Retinal Proteome Profiling of Inherited Retinal Degeneration Across Three Different Mouse Models Suggests Common Drug Targets in Retinitis Pigmentosa. Mol Cell Proteomics 2024; 23:100855. [PMID: 39389360 PMCID: PMC11602984 DOI: 10.1016/j.mcpro.2024.100855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/14/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024] Open
Abstract
Inherited retinal degenerations (IRDs) are a leading cause of blindness among the population of young people in the developed world. Approximately half of IRDs initially manifest as gradual loss of night vision and visual fields, characteristic of retinitis pigmentosa (RP). Due to challenges in genetic testing, and the large heterogeneity of mutations underlying RP, targeted gene therapies are an impractical largescale solution in the foreseeable future. For this reason, identifying key pathophysiological pathways in IRDs that could be targets for mutation-agnostic and disease-modifying therapies (DMTs) is warranted. In this study, we investigated the retinal proteome of three distinct IRD mouse models, in comparison to sex- and age-matched wild-type mice. Specifically, we used the Pde6βRd10 (rd10) and RhoP23H/WT (P23H) mouse models of autosomal recessive and autosomal dominant RP, respectively, as well as the Rpe65-/- mouse model of Leber's congenital amaurosis type 2 (LCA2). The mice were housed at two distinct institutions and analyzed using LC-MS in three separate facilities/instruments following data-dependent and data-independent acquisition modes. This cross-institutional and multi-methodological approach signifies the reliability and reproducibility of the results. The large-scale profiling of the retinal proteome, coupled with in vivo electroretinography recordings, provided us with a reliable basis for comparing the disease phenotypes and severity. Despite evident inflammation, cellular stress, and downscaled phototransduction observed consistently across all three models, the underlying pathologies of RP and LCA2 displayed many differences, sharing only four general KEGG pathways. The opposite is true for the two RP models in which we identify remarkable convergence in proteomic phenotype even though the mechanism of primary rod death in rd10 and P23H mice is different. Our data highlights the cAMP and cGMP second-messenger signaling pathways as potential targets for therapeutic intervention. The proteomic data is curated and made publicly available, facilitating the discovery of universal therapeutic targets for RP.
Collapse
Affiliation(s)
- Ahmed B Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Fangyuan Gao
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Danielle Peters
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Katri Vainionpää
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ning Zhibin
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dorota Skowronska-Krawczyk
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Daniel Figeys
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Krzysztof Palczewski
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA; Department of Chemistry, University of California, Irvine, California, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
3
|
Mashhadi Z, Yin L, Dosoky NS, Chen W, Davies SS. Plaat1l1 controls feeding induced NAPE biosynthesis and contributes to energy balance regulation in zebrafish. Prostaglandins Other Lipid Mediat 2024; 174:106869. [PMID: 38977258 DOI: 10.1016/j.prostaglandins.2024.106869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
Dysregulation of energy balance leading to obesity is a significant risk factor for cardiometabolic diseases such as diabetes, non-alcoholic fatty liver disease and atherosclerosis. In rodents and several other vertebrates, feeding has been shown to induce a rapid rise in the intestinal levels of N-acyl-ethanolamines (NAEs) and the chronic consumption of a high fat diet abolishes this rise. Administering NAEs to rodents consuming a high fat diet reduces their adiposity, in part by reducing food intake and enhancing fat oxidation, so that feeding-induced intestinal NAE biosynthesis appears to be critical to appropriate regulation of energy balance. However, the contribution of feeding-induced intestinal NAE biosynthesis to appropriate energy balance remains poorly understood in part because there are multiple enzymes that can contribute to NAE biosynthesis and the specific enzyme(s) that are responsible for feeding-induced intestinal NAE biosynthesis have not been identified. The rate-limiting step in the intestinal biosynthesis of NAEs is formation of their immediate precursors, the N-acyl-phosphatidylethanolamines (NAPEs), by phosphatidylethanolamine N-acyltransferases (NATs). At least six NATs are found in humans and multiple homologs of these NATs are found in most vertebrate species. In recent years, the fecundity and small size of zebrafish (Danio rerio), as well as their similarities in feeding behavior and energy balance regulation with mammals, have led to their use to model key features of cardiometabolic disease. We therefore searched the Danio rerio genome to identify all NAT homologs and found two additional NAT homologs besides the previously reported plaat1, rarres3, and rarres3l, and used CRISPR/cas9 to delete these two NAT homologs (plaat1l1 and plaat1l2). While wild-type fish markedly increased their intestinal NAPE levels in response to a meal after fasting, this response was completely ablated in plaat1l1-/-fish. Furthermore, plaat1l1-/- fish fed a standard flake diet had increased weight gain and glucose intolerance compared to wild-type fish. The results support a critical role for feeding-induced NAPE and NAE biosynthesis in regulating energy balance and suggest that restoring this response in obese animals could potentially be used to treat obesity and cardiometabolic disease.
Collapse
Affiliation(s)
- Zahra Mashhadi
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States; Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Linlin Yin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Noura S Dosoky
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Sean S Davies
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States; Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
4
|
Leinonen H, Zhang J, Occelli LM, Seemab U, Choi EH, L P Marinho LF, Querubin J, Kolesnikov AV, Galinska A, Kordecka K, Hoang T, Lewandowski D, Lee TT, Einstein EE, Einstein DE, Dong Z, Kiser PD, Blackshaw S, Kefalov VJ, Tabaka M, Foik A, Petersen-Jones SM, Palczewski K. A combination treatment based on drug repurposing demonstrates mutation-agnostic efficacy in pre-clinical retinopathy models. Nat Commun 2024; 15:5943. [PMID: 39009597 PMCID: PMC11251169 DOI: 10.1038/s41467-024-50033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 06/21/2024] [Indexed: 07/17/2024] Open
Abstract
Inherited retinopathies are devastating diseases that in most cases lack treatment options. Disease-modifying therapies that mitigate pathophysiology regardless of the underlying genetic lesion are desirable due to the diversity of mutations found in such diseases. We tested a systems pharmacology-based strategy that suppresses intracellular cAMP and Ca2+ activity via G protein-coupled receptor (GPCR) modulation using tamsulosin, metoprolol, and bromocriptine coadministration. The treatment improves cone photoreceptor function and slows degeneration in Pde6βrd10 and RhoP23H/WT retinitis pigmentosa mice. Cone degeneration is modestly mitigated after a 7-month-long drug infusion in PDE6A-/- dogs. The treatment also improves rod pathway function in an Rpe65-/- mouse model of Leber congenital amaurosis but does not protect from cone degeneration. RNA-sequencing analyses indicate improved metabolic function in drug-treated Rpe65-/- and rd10 mice. Our data show that catecholaminergic GPCR drug combinations that modify second messenger levels via multiple receptor actions provide a potential disease-modifying therapy against retinal degeneration.
Collapse
Affiliation(s)
- Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211, Kuopio, Finland.
| | - Jianye Zhang
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Laurence M Occelli
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Umair Seemab
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211, Kuopio, Finland
| | - Elliot H Choi
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | | | - Janice Querubin
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Alexander V Kolesnikov
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Anna Galinska
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kordecka
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Thanh Hoang
- Department of Ophthalmology, Department of Cell & Developmental Biology, Ann Arbor, MI, 48105, USA
| | - Dominik Lewandowski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Timothy T Lee
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Elliott E Einstein
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - David E Einstein
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Zhiqian Dong
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Philip D Kiser
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, CA, 92697, USA
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, CA, 92697, USA
- Research Service, VA Long Beach Healthcare System, Long Beach, California, 90822, USA
| | - Seth Blackshaw
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vladimir J Kefalov
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, CA, 92697, USA
| | - Marcin Tabaka
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Andrzej Foik
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | | | - Krzysztof Palczewski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA.
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, CA, 92697, USA.
- Department of Chemistry, University of California-Irvine, Irvine, CA, 92697, USA.
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, 92697, USA.
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
5
|
Nguyen TD, Chen YI, Nguyen AT, Yonas S, Sripati MP, Kuo YA, Hong S, Litvinov M, He Y, Yeh HC, Grady Rylander H. Two-photon autofluorescence lifetime assay of rabbit photoreceptors and retinal pigment epithelium during light-dark visual cycles in rabbit retina. BIOMEDICAL OPTICS EXPRESS 2024; 15:3094-3111. [PMID: 38855698 PMCID: PMC11161359 DOI: 10.1364/boe.511806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 06/11/2024]
Abstract
Two-photon excited fluorescence (TPEF) is a powerful technique that enables the examination of intrinsic retinal fluorophores involved in cellular metabolism and the visual cycle. Although previous intensity-based TPEF studies in non-human primates have successfully imaged several classes of retinal cells and elucidated aspects of both rod and cone photoreceptor function, fluorescence lifetime imaging (FLIM) of the retinal cells under light-dark visual cycle has yet to be fully exploited. Here we demonstrate a FLIM assay of photoreceptors and retinal pigment epithelium (RPE) that reveals key insights into retinal physiology and adaptation. We found that photoreceptor fluorescence lifetimes increase and decrease in sync with light and dark exposure, respectively. This is likely due to changes in all-trans-retinol and all-trans-retinal levels in the outer segments, mediated by phototransduction and visual cycle activity. During light exposure, RPE fluorescence lifetime was observed to increase steadily over time, as a result of all-trans-retinol accumulation during the visual cycle and decreasing metabolism caused by the lack of normal perfusion of the sample. Our system can measure the fluorescence lifetime of intrinsic retinal fluorophores on a cellular scale, revealing differences in lifetime between retinal cell classes under different conditions of light and dark exposure.
Collapse
Affiliation(s)
- Trung Duc Nguyen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Yuan-I Chen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Anh-Thu Nguyen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Siem Yonas
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Manasa P Sripati
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Yu-An Kuo
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Soonwoo Hong
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Mitchell Litvinov
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Yujie He
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Hsin-Chih Yeh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
- Texas Materials Institute, University of Texas at Austin, Austin, TX, USA
| | - H Grady Rylander
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Leung M, Steinman J, Li D, Lor A, Gruesen A, Sadah A, van Kuijk FJ, Montezuma SR, Kondkar AA, Radhakrishnan R, Lobo GP. The Logistical Backbone of Photoreceptor Cell Function: Complementary Mechanisms of Dietary Vitamin A Receptors and Rhodopsin Transporters. Int J Mol Sci 2024; 25:4278. [PMID: 38673863 PMCID: PMC11050646 DOI: 10.3390/ijms25084278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
In this review, we outline our current understanding of the mechanisms involved in the absorption, storage, and transport of dietary vitamin A to the eye, and the trafficking of rhodopsin protein to the photoreceptor outer segments, which encompasses the logistical backbone required for photoreceptor cell function. Two key mechanisms of this process are emphasized in this manuscript: ocular and systemic vitamin A membrane transporters, and rhodopsin transporters. Understanding the complementary mechanisms responsible for the generation and proper transport of the retinylidene protein to the photoreceptor outer segment will eventually shed light on the importance of genes encoded by these proteins, and their relationship on normal visual function and in the pathophysiology of retinal degenerative diseases.
Collapse
Affiliation(s)
- Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Jeremy Steinman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Dorothy Li
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Anjelynt Lor
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Ahmed Sadah
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Frederik J. van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Sandra R. Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 12271, Saudi Arabia;
| | - Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Glenn P. Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| |
Collapse
|
7
|
Plau J, Morgan CE, Fedorov Y, Banerjee S, Adams DJ, Blaner WS, Yu EW, Golczak M. Discovery of Nonretinoid Inhibitors of CRBP1: Structural and Dynamic Insights for Ligand-Binding Mechanisms. ACS Chem Biol 2023; 18:2309-2323. [PMID: 37713257 PMCID: PMC10591915 DOI: 10.1021/acschembio.3c00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023]
Abstract
The dysregulation of retinoid metabolism has been linked to prevalent ocular diseases including age-related macular degeneration and Stargardt disease. Modulating retinoid metabolism through pharmacological approaches holds promise for the treatment of these eye diseases. Cellular retinol-binding protein 1 (CRBP1) is the primary transporter of all-trans-retinol (atROL) in the eye, and its inhibition has recently been shown to protect mouse retinas from light-induced retinal damage. In this report, we employed high-throughput screening to identify new chemical scaffolds for competitive, nonretinoid inhibitors of CRBP1. To understand the mechanisms of interaction between CRBP1 and these inhibitors, we solved high-resolution X-ray crystal structures of the protein in complex with six selected compounds. By combining protein crystallography with hydrogen/deuterium exchange mass spectrometry, we quantified the conformational changes in CRBP1 caused by different inhibitors and correlated their magnitude with apparent binding affinities. Furthermore, using molecular dynamic simulations, we provided evidence for the functional significance of the "closed" conformation of CRBP1 in retaining ligands within the binding pocket. Collectively, our study outlines the molecular foundations for understanding the mechanism of high-affinity interactions between small molecules and CRBPs, offering a framework for the rational design of improved inhibitors for this class of lipid-binding proteins.
Collapse
Affiliation(s)
- Jacqueline Plau
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Christopher E. Morgan
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department
of Chemistry, Thiel College, Greenville, Pennsylvania 16125, United States
| | - Yuriy Fedorov
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Surajit Banerjee
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14850, United States
- Northeastern
Collaborative Access Team, Argonne National
Laboratory, Argonne, Illinois 60439, United States
| | - Drew J. Adams
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - William S. Blaner
- Department
of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
| | - Edward W. Yu
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Marcin Golczak
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
8
|
Hong JD, Palczewski K. A short story on how chromophore is hydrolyzed from rhodopsin for recycling. Bioessays 2023; 45:e2300068. [PMID: 37454357 PMCID: PMC10614701 DOI: 10.1002/bies.202300068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023]
Abstract
The photocycle of visual opsins is essential to maintain the light sensitivity of the retina. The early physical observations of the rhodopsin photocycle by Böll and Kühne in the 1870s inspired over a century's worth of investigations on rhodopsin biochemistry. A single photon isomerizes the Schiff-base linked 11-cis-retinylidene chromophore of rhodopsin, converting it to the all-trans agonist to elicit phototransduction through photoactivated rhodopsin (Rho*). Schiff base hydrolysis of the agonist is a key step in the photocycle, not only diminishing ongoing phototransduction but also allowing for entry and binding of fresh 11-cis chromophore to regenerate the rhodopsin pigment and maintain light sensitivity. Many challenges have been encountered in measuring the rate of this hydrolysis, but recent advancements have facilitated studies of the hydrolysis within the native membrane environment of rhodopsin. These techniques can now be applied to study hydrolysis of agonist in other opsin proteins that mediate phototransduction or chromophore turnover. In this review, we discuss the progress that has been made in characterizing the rhodopsin photocycle and the journey to characterize the hydrolysis of its all-trans-retinylidene agonist.
Collapse
Affiliation(s)
- John D. Hong
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California Irvine, Irvine, CA 92697, USA
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California Irvine, Irvine, CA 92697, USA
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
9
|
Zampatti S, Peconi C, Calvino G, Ferese R, Gambardella S, Cascella R, Sebastiani J, Falsini B, Cusumano A, Giardina E. A Splicing Variant in RDH8 Is Associated with Autosomal Recessive Stargardt Macular Dystrophy. Genes (Basel) 2023; 14:1659. [PMID: 37628710 PMCID: PMC10454646 DOI: 10.3390/genes14081659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Stargardt macular dystrophy is a genetic disorder, but in many cases, the causative gene remains unrevealed. Through a combined approach (whole-exome sequencing and phenotype/family-driven filtering algorithm) and a multilevel validation (international database searching, prediction scores calculation, splicing analysis assay, segregation analyses), a biallelic mutation in the RDH8 gene was identified to be responsible for Stargardt macular dystrophy in a consanguineous Italian family. This paper is a report on the first family in which a biallelic deleterious mutation in RDH8 is detected. The disease phenotype is consistent with the expected phenotype hypothesized in previous studies on murine models. The application of the combined approach to genetic data and the multilevel validation allowed the identification of a splicing mutation in a gene that has never been reported before in human disorders.
Collapse
Affiliation(s)
- Stefania Zampatti
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (S.Z.)
| | - Cristina Peconi
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (S.Z.)
| | - Giulia Calvino
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (S.Z.)
| | | | - Stefano Gambardella
- Neuromed IRCSS, 86077 Pozzilli, Italy
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy
| | - Raffaella Cascella
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (S.Z.)
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, 1000 Tirana, Albania
| | | | - Benedetto Falsini
- Macula & Genoma Foundation, 00133 Rome, Italy; (J.S.)
- Department of Ophthalmology, Policlinico A. Gemelli, IRCCS/Catholic University, 00133 Rome, Italy
- Macula & Genoma Foundation USA, New York, NY 10017, USA
| | - Andrea Cusumano
- Macula & Genoma Foundation, 00133 Rome, Italy; (J.S.)
- Macula & Genoma Foundation USA, New York, NY 10017, USA
- Department of Ophthalmology, Tor Vergata University, 00133 Rome, Italy
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (S.Z.)
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
10
|
Kim HJ, Zhao J, Walewski JL, Sparrow JR. A High Fat Diet Fosters Elevated Bisretinoids. J Biol Chem 2023; 299:104784. [PMID: 37146972 DOI: 10.1016/j.jbc.2023.104784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/07/2023] Open
Abstract
High dietary fat intake is associated with metabolic dysregulation, but little is known regarding the effects of a high fat diet (HFD) on photoreceptor cell functioning. We explored the intersection of a high fat diet (HFD) and the visual cycle adducts that form in photoreceptor cells by non-enzymatic reactions. In black C57BL/6J mice and albino C57BL/6Jc2j mice raised on a high fat diet until age 3, 6 or 12 months, chromatographically quantified bisretinoids were increased relative to mice on a standard diet. In vivo measurement of fundus autofluorescence, the source of which is bisretinoid, also revealed a significant increase in the HFD-mice. Additionally, mice provided with a diet high in fat presented with elevated retinol-binding protein 4 (RBP4) the protein responsible for transporting retinol in plasma. Vitamin A was elevated in plasma although not in ocular tissue. Bisretinoids form in photoreceptor cell outer segments by random reactions of retinaldehyde with phosphatidylethanolamine. We found that the latter phospholipid was significantly increased in mice fed a HFD versus mice on a control diet. In leptin-deficient ob/ob mice, a genetic model of obesity, plasma levels of Rbp4 protein were higher but bisretinoids in retina were not elevated. Photoreceptor cell viability measured as outer nuclear layer thickness was reduced in the ob/ob mice relative to wild-type. The accelerated formation of bisretinoid we observed in diet induced obese mice is related to the high fat intake and to increased delivery of vitamin A to the visual cycle.
Collapse
Affiliation(s)
- Hye Jin Kim
- Departments of Ophthalmology, Columbia University Medical Center, 635 W. 165(th) Street, New York NY, 10032
| | - Jin Zhao
- Departments of Ophthalmology, Columbia University Medical Center, 635 W. 165(th) Street, New York NY, 10032
| | - Jose L Walewski
- Departments of Medicine, Columbia University Medical Center, 635 W. 165(th) Street, New York NY, 10032
| | - Janet R Sparrow
- Departments of Ophthalmology, Columbia University Medical Center, 635 W. 165(th) Street, New York NY, 10032; Departments of Pathology and Cell Biology, Columbia University Medical Center, 635 W. 165(th) Street, New York NY, 10032.
| |
Collapse
|
11
|
Uppal S, Poliakov E, Gentleman S, Redmond TM. The Amphipathic Helix in Visual Cycle Proteins: A Review. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:533-537. [PMID: 37440083 PMCID: PMC11299856 DOI: 10.1007/978-3-031-27681-1_78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The visual cycle is a complex biological process that involves the sequential action of proteins in the retinal pigment epithelial (RPE) cells and photoreceptors to modify and shuttle visual retinoids. A majority of the visual cycle proteins are membrane proteins, either integral or peripheral membrane proteins. Despite significant progress in understanding their physiological function, very limited structural information is available for the visual cycle proteins. Moreover, the mechanism of membrane interaction is not yet clear in all cases. Here, we demonstrate the presence of an amphipathic helix in selected RPE visual cycle proteins, using in silico tools, and highlight their role in membrane association and function.
Collapse
Affiliation(s)
- Sheetal Uppal
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eugenia Poliakov
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan Gentleman
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - T Michael Redmond
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
12
|
Pasquaré SJ, Chamorro-Aguirre E, Gaveglio VL. The endocannabinoid system in the visual process. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2022. [DOI: 10.1016/j.jpap.2022.100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
13
|
Zhuang X, Ma J, Xu G, Sun Z. SHP-1 knockdown suppresses mitochondrial biogenesis and aggravates mitochondria-dependent apoptosis induced by all trans retinal through the STING/AMPK pathways. Mol Med 2022; 28:125. [PMID: 36273174 PMCID: PMC9588232 DOI: 10.1186/s10020-022-00554-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Oxidative stress-caused damage to the retinal pigment epithelium (RPE) underlies the onset and progression of age-related macular degeneration (AMD). Impaired mitochondrial biogenesis sensitizes RPE cells to mitochondrial dysfunction, energy insufficiency and death. Src-homology 2 domain-containing phosphatase (SHP)-1 is important in regulating immune responses and cell survival. However, its roles in cell survival are not always consistent. Until now, the effects of SHP-1 on RPE dysfunction, especially mitochondrial homeostasis, remain to be elucidated. We sought to clarify the effects of SHP-1 in RPE cells in response to atRAL-induced oxidative stress and determine the regulatory mechanisms involved. METHODS In the all trans retinal (atRAL)-induced oxidative stress model, we used the vector of lentivirus to knockdown the expression of SHP-1 in ARPE-19 cells. CCK-8 assay, Annexin V/PI staining and JC-1 staining were utilized to determine the cell viability, cell apoptosis and mitochondrial membrane potential. We also used immunoprecipitation to examine the ubiquitination modification of stimulator of interferon genes (STING) and its interaction with SHP-1. The expression levels of mitochondrial marker, proteins related to mitochondrial biogenesis, and signaling molecules involved were examined by western blotting analysis. RESULTS We found that SHP-1 knockdown predisposed RPE cells to apoptosis, aggravated mitochondrial damage, and repressed mitochondrial biogenesis after treatment with atRAL. Immunofluoresent staining and immunoprecipitation analysis confirmed that SHP-1 interacted with the endoplasmic reticulum-resident STING and suppressed K63-linked ubiquitination and activation of STING. Inhibition of STING with the specific antagonist H151 attenuated the effects of SHP-1 knockdown on mitochondrial biogenesis and oxidative damage. The adenosine monophosphate-activated protein kinase (AMPK) pathway acted as the crucial downstream target of STING and was involved in the regulatory processes. CONCLUSIONS These findings suggest that SHP-1 knockdown potentiates STING overactivation and represses mitochondrial biogenesis and cell survival, at least in part by blocking the AMPK pathway in RPE cells. Therefore, restoring mitochondrial health by regulating SHP-1 in RPE cells may be a potential therapeutic strategy for degenerative retinal diseases including AMD.
Collapse
Affiliation(s)
- Xiaonan Zhuang
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Jun Ma
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Gezhi Xu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Zhongcui Sun
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Network biology analysis of P23H rhodopsin interactome identifies protein and mRNA quality control mechanisms. Sci Rep 2022; 12:17405. [PMID: 36258031 PMCID: PMC9579138 DOI: 10.1038/s41598-022-22316-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/12/2022] [Indexed: 02/05/2023] Open
Abstract
Rhodopsin is essential for phototransduction, and many rhodopsin mutations cause heritable retinal degenerations. The P23H rhodopsin variant generates a misfolded rhodopsin protein that photoreceptors quickly target for degradation by mechanisms that are incompletely understood. To gain insight into how P23H rhodopsin is removed from rods, we used mass spectrometry to identify protein interaction partners of P23H rhodopsin immunopurified from RhoP23H/P23H mice and compared them with protein interaction partners of wild-type rhodopsin from Rho+/+ mice. We identified 286 proteins associated with P23H rhodopsin and 276 proteins associated with wild-type rhodopsin. 113 proteins were shared between wild-type and mutant rhodopsin protein interactomes. In the P23H rhodopsin protein interactome, we saw loss of phototransduction, retinal cycle, and rhodopsin protein trafficking proteins but gain of ubiquitin-related proteins when compared with the wild-type rhodopsin protein interactome. In the P23H rhodopsin protein interactome, we saw enrichment of gene ontology terms related to ER-associated protein degradation, ER stress, and translation. Protein-protein interaction network analysis revealed that translational and ribosomal quality control proteins were significant regulators in the P23H rhodopsin protein interactome. The protein partners identified in our study may provide new insights into how photoreceptors recognize and clear mutant rhodopsin, offering possible novel targets involved in retinal degeneration pathogenesis.
Collapse
|
15
|
Das A, Imanishi Y. Drug Discovery Strategies for Inherited Retinal Degenerations. BIOLOGY 2022; 11:1338. [PMID: 36138817 PMCID: PMC9495580 DOI: 10.3390/biology11091338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Inherited retinal degeneration is a group of blinding disorders afflicting more than 1 in 4000 worldwide. These disorders frequently cause the death of photoreceptor cells or retinal ganglion cells. In a subset of these disorders, photoreceptor cell death is a secondary consequence of retinal pigment epithelial cell dysfunction or degeneration. This manuscript reviews current efforts in identifying targets and developing small molecule-based therapies for these devastating neuronal degenerations, for which no cures exist. Photoreceptors and retinal ganglion cells are metabolically demanding owing to their unique structures and functional properties. Modulations of metabolic pathways, which are disrupted in most inherited retinal degenerations, serve as promising therapeutic strategies. In monogenic disorders, great insights were previously obtained regarding targets associated with the defective pathways, including phototransduction, visual cycle, and mitophagy. In addition to these target-based drug discoveries, we will discuss how phenotypic screening can be harnessed to discover beneficial molecules without prior knowledge of their mechanisms of action. Because of major anatomical and biological differences, it has frequently been challenging to model human inherited retinal degeneration conditions using small animals such as rodents. Recent advances in stem cell-based techniques are opening new avenues to obtain pure populations of human retinal ganglion cells and retinal organoids with photoreceptor cells. We will discuss concurrent ideas of utilizing stem-cell-based disease models for drug discovery and preclinical development.
Collapse
Affiliation(s)
- Arupratan Das
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yoshikazu Imanishi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Wen Y, Dai B, Zhang X, Zhu H, Xie C, Xia J, Sun Y, Zhu M, Tong J, Shen Y. Retinal Transcriptomics Analysis Reveals the Underlying Mechanism of Disturbed Emmetropization Induced by Wavelength Defocus. Curr Eye Res 2022; 47:908-917. [PMID: 35225751 DOI: 10.1080/02713683.2022.2048395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/24/2022] [Indexed: 12/28/2022]
Abstract
PURPOSE Wavelength signals play a vital role in refractive development. This study aimed to explore the retinal transcriptome signature in these processes. METHODS Guinea pigs were randomly divided into three groups exposed to white, blue, or green environmental light for eight weeks. Refraction and axial length were evaluated every 4 weeks, and the retinal transcriptome was profiled at 8 weeks. RESULTS Compared with the white group, ocular refraction significantly decreased and ocular axial length significantly extended in the green group whereas these parameters showed opposite trends in the blue group. RNA-sequencing showed that, compared with the white group, 184 and 171 differentially expressed genes (DEGs) were found in the blue and green groups, respectively. Among these DEGs, only 31 overlapped. These two sets of DEGs were enriched in distinct biological processes and pathways. There were 268 DEGs between the blue and green groups, which were primarily enriched in the extracellular matrix, and metabolism, receptor activity, and ion binding processes. In addition, nine human genes, including ECEL1, CHRND, SHBG, PRSS56, OVOL1, RDH5, WNT7B, PEBP4, CA12, were identified to be related to myopia development and wavelength response, indicating the potential role of these genes in human wavelength-induced myopia. CONCLUSIONS In this study, we identified retinal targets and pathways involved in the response to wavelength signals in emmetropization.
Collapse
Affiliation(s)
- Yingying Wen
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Binbin Dai
- Department of Ophthalmology, Taizhou Hospital, Taizhou, Zhejiang, China
| | - Xuhong Zhang
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong Zhu
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen Xie
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianhua Xia
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Sun
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Miaomiao Zhu
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianping Tong
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ye Shen
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Piano I, Corsi F, Polini B, Gargini C. Nutraceutical Molecules Slow Down Retinal Degeneration, in Tvrm4 Mice a Model of Retinitis Pigmentosa, by Genetic Modulation of Anti-oxidant Pathway. Front Neurosci 2022; 16:868750. [PMID: 35516813 PMCID: PMC9063314 DOI: 10.3389/fnins.2022.868750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022] Open
Abstract
Rhodopsin (RHO) mutations are responsible for 25–40% of the dominant cases of retinitis pigmentosa (RP) with different severity and progression rates. The Tvrm4 mice, heterozygous for an I307N dominant mutation of RHO, display a normal retinal phenotype when raised in ambient light conditions, but undergo photoreceptor degeneration when briefly exposed to strong white light. Here, The Tvrm4 mice is pre-treated with naringenin 100 mg/kg/die, quercetin 100 mg/kg/die, naringenin 50 + quercercetin 100 mg/kg/die or vehicle dimethyl sulfoxide (DMSO 0.025%) in the drinking water for 35 days. On the 30th day, retinal degeneration was induced by exposure for 1 min to the white light of 12,000 lux intensity, and the treatment was repeated for another 5 days. At the end of the protocol retinal functionality was tested by recording an electroretinogram (ERG). The retinal tissue was collected and was used for further analyses, including immunohistochemically, biochemical, and molecular biology assays. The data obtained show that treatment with nutraceutical molecules is effective in counteracting retinal degeneration by preserving the functionality of photoreceptors and increasing the antioxidant and anti-apoptotic pathways of retinal cells. The present data confirm that nutraceutical molecules are effective in slowing photoreceptor degeneration in a mutation-independent way by modulating the antioxidant response of the retina at the gene expression level.
Collapse
Affiliation(s)
- Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
- *Correspondence: Ilaria Piano,
| | | | - Beatrice Polini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Interdepartmental Center for Nutraceutical Research and Nutrition or Health, University of Pisa, Pisa, Italy
| |
Collapse
|
18
|
O’Connor C, Varshosaz P, Moise AR. Mechanisms of Feedback Regulation of Vitamin A Metabolism. Nutrients 2022; 14:nu14061312. [PMID: 35334970 PMCID: PMC8950952 DOI: 10.3390/nu14061312] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Vitamin A is an essential nutrient required throughout life. Through its various metabolites, vitamin A sustains fetal development, immunity, vision, and the maintenance, regulation, and repair of adult tissues. Abnormal tissue levels of the vitamin A metabolite, retinoic acid, can result in detrimental effects which can include congenital defects, immune deficiencies, proliferative defects, and toxicity. For this reason, intricate feedback mechanisms have evolved to allow tissues to generate appropriate levels of active retinoid metabolites despite variations in the level and format, or in the absorption and conversion efficiency of dietary vitamin A precursors. Here, we review basic mechanisms that govern vitamin A signaling and metabolism, and we focus on retinoic acid-controlled feedback mechanisms that contribute to vitamin A homeostasis. Several approaches to investigate mechanistic details of the vitamin A homeostatic regulation using genomic, gene editing, and chromatin capture technologies are also discussed.
Collapse
Affiliation(s)
- Catherine O’Connor
- MD Program, Northern Ontario School of Medicine, 317-MSE Bldg., 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada;
| | - Parisa Varshosaz
- Biology and Biomolecular Sciences Ph.D. Program, Northern Ontario School of Medicine, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Alexander R. Moise
- Medical Sciences Division, Northern Ontario School of Medicine, 317-MSE Bldg., 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- Department of Chemistry and Biochemistry, Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: ; Tel.: +1-705-662-7253
| |
Collapse
|
19
|
Sajovic J, Meglič A, Glavač D, Markelj Š, Hawlina M, Fakin A. The Role of Vitamin A in Retinal Diseases. Int J Mol Sci 2022; 23:1014. [PMID: 35162940 PMCID: PMC8835581 DOI: 10.3390/ijms23031014] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/24/2022] Open
Abstract
Vitamin A is an essential fat-soluble vitamin that occurs in various chemical forms. It is essential for several physiological processes. Either hyper- or hypovitaminosis can be harmful. One of the most important vitamin A functions is its involvement in visual phototransduction, where it serves as the crucial part of photopigment, the first molecule in the process of transforming photons of light into electrical signals. In this process, large quantities of vitamin A in the form of 11-cis-retinal are being isomerized to all-trans-retinal and then quickly recycled back to 11-cis-retinal. Complex machinery of transporters and enzymes is involved in this process (i.e., the visual cycle). Any fault in the machinery may not only reduce the efficiency of visual detection but also cause the accumulation of toxic chemicals in the retina. This review provides a comprehensive overview of diseases that are directly or indirectly connected with vitamin A pathways in the retina. It includes the pathophysiological background and clinical presentation of each disease and summarizes the already existing therapeutic and prospective interventions.
Collapse
Affiliation(s)
- Jana Sajovic
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Andrej Meglič
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Damjan Glavač
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Špela Markelj
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Marko Hawlina
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Ana Fakin
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| |
Collapse
|
20
|
Kawamura S, Tachibanaki S. Molecular basis of rod and cone differences. Prog Retin Eye Res 2021; 90:101040. [PMID: 34974196 DOI: 10.1016/j.preteyeres.2021.101040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022]
Abstract
In the vertebrate retina, rods and cones both detect light, but they are different in functional aspects such as light sensitivity and time resolution, for example, and in some of cell biological aspects. For functional aspects, both photoreceptors are known to share a common mechanism, phototransduction cascade, consisting of a series of enzyme reactions to convert a photon-capture signal to an electrical signal. To understand the mechanisms of the functional differences between rods and cones at the molecular level, we compared biochemically each of the reactions in the phototransduction cascade between rods and cones using the cells isolated and purified from carp retina. Although proteins in the cascade are functionally similar between rods and cones, their activities together with their expression levels are mostly different between these photoreceptors. In general, reactions to generate a response are slightly less effective, as a total, in cones than in rods, but each of the reactions for termination and recovery of a response are much more effective in cones. These findings explain lower light sensitivity and briefer light responses in cones than in rods. In addition, our considerations suggest that a Ca2+-binding protein, S-modulin or recoverin, has a currently unnoticed role in shaping light responses. With comparison of the expression levels of proteins and/or mRNAs using purified cells, several proteins were found to be specifically or predominantly expressed in cones. These proteins would be of interest for future studies on the difference between rods and cones.
Collapse
Affiliation(s)
- Satoru Kawamura
- Graduate School of Frontier Biosciences, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan; Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan.
| | - Shuji Tachibanaki
- Graduate School of Frontier Biosciences, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan; Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
21
|
Ortega JT, Jastrzebska B. Rhodopsin as a Molecular Target to Mitigate Retinitis Pigmentosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1371:61-77. [PMID: 34962636 DOI: 10.1007/5584_2021_682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Retinitis pigmentosa (RP) is a group of hereditary degenerative diseases affecting 1 of 4000 people worldwide and being the most prevalent cause of visual handicap among working populations in developed countries. These disorders are mainly related to the abnormalities in the rod G protein-coupled receptor (GPCR), rhodopsin reflected in the dysregulated membrane trafficking, stability and phototransduction processes that lead to progressive loss of retina function and eventually blindness. Currently, there is no cure for RP, and the therapeutic options are limited. Targeting rhodopsin with small molecule chaperones to improve the folding and stability of the mutant receptor is one of the most promising pharmacological approaches to alleviate the pathology of RP. This review provides an update on the current knowledge regarding small molecule compounds that have been evaluated as rhodopsin modulators to be considered as leads for the development of novel therapies for RP.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology, School of Medicine, Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
22
|
Martin Ask N, Leung M, Radhakrishnan R, Lobo GP. Vitamin A Transporters in Visual Function: A Mini Review on Membrane Receptors for Dietary Vitamin A Uptake, Storage, and Transport to the Eye. Nutrients 2021; 13:nu13113987. [PMID: 34836244 PMCID: PMC8620617 DOI: 10.3390/nu13113987] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 11/23/2022] Open
Abstract
Vitamins are essential compounds obtained through diet that are necessary for normal development and function in an organism. One of the most important vitamins for human physiology is vitamin A, a group of retinoid compounds and carotenoids, which generally function as a mediator for cell growth, differentiation, immunity, and embryonic development, as well as serving as a key component in the phototransduction cycle in the vertebrate retina. For humans, vitamin A is obtained through the diet, where provitamin A carotenoids such as β-carotene from plants or preformed vitamin A such as retinyl esters from animal sources are absorbed into the body via the small intestine and converted into all-trans retinol within the intestinal enterocytes. Specifically, once absorbed, carotenoids are cleaved by carotenoid cleavage oxygenases (CCOs), such as Beta-carotene 15,15’-monooxygenase (BCO1), to produce all-trans retinal that subsequently gets converted into all-trans retinol. CRBP2 bound retinol is then converted into retinyl esters (REs) by the enzyme lecithin retinol acyltransferase (LRAT) in the endoplasmic reticulum, which is then packaged into chylomicrons and sent into the bloodstream for storage in hepatic stellate cells in the liver or for functional use in peripheral tissues such as the retina. All-trans retinol also travels through the bloodstream bound to retinol binding protein 4 (RBP4), where it enters cells with the assistance of the transmembrane transporters, stimulated by retinoic acid 6 (STRA6) in peripheral tissues or retinol binding protein 4 receptor 2 (RBPR2) in systemic tissues (e.g., in the retina and the liver, respectively). Much is known about the intake, metabolism, storage, and function of vitamin A compounds, especially with regard to its impact on eye development and visual function in the retinoid cycle. However, there is much to learn about the role of vitamin A as a transcription factor in development and cell growth, as well as how peripheral cells signal hepatocytes to secrete all-trans retinol into the blood for peripheral cell use. This article aims to review literature regarding the major known pathways of vitamin A intake from dietary sources into hepatocytes, vitamin A excretion by hepatocytes, as well as vitamin A usage within the retinoid cycle in the RPE and retina to provide insight on future directions of novel membrane transporters for vitamin A in retinal cell physiology and visual function.
Collapse
|
23
|
Lešták J. NEUROTRANSMISSION IN VISUAL ANALYZER AND BIONIC EYE. A REVIEW. CESKA A SLOVENSKA OFTALMOLOGIE : CASOPIS CESKE OFTALMOLOGICKE SPOLECNOSTI A SLOVENSKE OFTALMOLOGICKE SPOLECNOSTI 2021; 77:55-59. [PMID: 33985334 DOI: 10.31348/2020/28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIMS The aim of the work is to point out the transmission of electrical voltage changes in the visual analyser and thus the efficiency of the bionic eye. MATERIAL AND METHODS The review deals with the question of the transmission of electrical changes in visual path voltage under physiological and pathological conditions. In particular, it points to feedback autoregulatory damage not only of primarily altered cellular structures, but of all other, both horizontally and vertically localized. Based on the results of functional magnetic resonance imaging and electrophysiological methods, it shows the pathology of the entire visual pathway in three eye diseases: retinitis pigmentosa, age-related macular degeneration and glaucoma. RESULTS The thesis also provides an overview of possible systems that are used to replace lost vision, from epiretinal, subretinal, suprachoroidal implants, through stimulation of the optic nerve, corpus geniculatum laterale to the visual cortex. CONCLUSION Due to the pathology of neurotransmission, bionic eye systems cannot be expected to be restored after stabilization of binocular functions.
Collapse
|
24
|
Kim JY, Park S, Park HJ, Kim SH, Lew H, Kim GJ. PEDF-Mediated Mitophagy Triggers the Visual Cycle by Enhancing Mitochondrial Functions in a H 2O 2-Injured Rat Model. Cells 2021; 10:cells10051117. [PMID: 34066394 PMCID: PMC8148157 DOI: 10.3390/cells10051117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
Retinal degenerative diseases result from oxidative stress and mitochondrial dysfunction, leading to the loss of visual acuity. Damaged retinal pigment epithelial (RPE) and photoreceptor cells undergo mitophagy. Pigment epithelium-derived factor (PEDF) protects from oxidative stress in RPE and improves mitochondrial functions. Overexpression of PEDF in placenta-derived mesenchymal stem cells (PD-MSCs; PD-MSCsPEDF) provides therapeutic effects in retinal degenerative diseases. Here, we investigated whether PD-MSCsPEDF restored the visual cycle through a mitophagic mechanism in RPE cells in hydrogen peroxide (H2O2)-injured rat retinas. Compared with naïve PD-MSCs, PD-MSCsPEDF augmented mitochondrial biogenesis and translation markers as well as mitochondrial respiratory states. In the H2O2-injured rat model, intravitreal administration of PD-MSCsPEDF restored total retinal layer thickness compared to that of naïve PD-MSCs. In particular, PTEN-induced kinase 1 (PINK1), which is the major mitophagy marker, exhibited increased expression in retinal layers and RPE cells after PD-MSCPEDF transplantation. Similarly, expression of the visual cycle enzyme retinol dehydrogenase 11 (RDH11) showed the same patterns as PINK1 levels, resulting in improved visual activity. Taken together, these findings suggest that PD-MSCsPEDF facilitate mitophagy and restore the loss of visual cycles in H2O2-injured rat retinas and RPE cells. These data indicate a new strategy for next-generation MSC-based treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Jae Yeon Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
- Research Institute of Placental Science, CHA University, Seongnam 13488, Korea
| | - Sohae Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
| | - Hee Jung Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
| | - Se Ho Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
| | - Helen Lew
- CHA Bundang Medical Center, Department of Ophthalmology, CHA University, Seongnam 13496, Korea;
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
- Research Institute of Placental Science, CHA University, Seongnam 13488, Korea
- Correspondence: ; Tel.: +82-31-881-7145
| |
Collapse
|
25
|
Chatterjee S, Zhou J, Dasgupta R, Cramer-Blok A, Timmer M, van der Stelt M, Ubbink M. Protein Dynamics Influence the Enzymatic Activity of Phospholipase A/Acyltransferases 3 and 4. Biochemistry 2021; 60:1178-1190. [PMID: 33749246 PMCID: PMC8154263 DOI: 10.1021/acs.biochem.0c00974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/04/2021] [Indexed: 11/29/2022]
Abstract
Phospholipase A/acyltransferase 3 (PLAAT3) and PLAAT4 are enzymes involved in the synthesis of bioactive lipids. Despite sequential and structural similarities, the two enzymes differ in activity and specificity. The relation between the activity and dynamics of the N-terminal domains of PLAAT3 and PLAAT4 was studied. PLAAT3 has a much higher melting temperature and exhibits less nanosecond and millisecond dynamics in the active site, in particular in loop L2(B6), as shown by NMR spectroscopy and molecular dynamics calculations. Swapping the L2(B6) loops between the two PLAAT enzymes results in strongly increased phospholipase activity in PLAAT3 but no reduction in PLAAT4 activity, indicating that this loop contributes to the low activity of PLAAT3. The results show that, despite structural similarity, protein dynamics differ substantially between the PLAAT variants, which can help to explain the activity and specificity differences.
Collapse
Affiliation(s)
- Soumya
Deep Chatterjee
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Juan Zhou
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Rubin Dasgupta
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Anneloes Cramer-Blok
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Monika Timmer
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Mario van der Stelt
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Marcellus Ubbink
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| |
Collapse
|
26
|
Membrane binding properties of the C-terminal segment of retinol dehydrogenase 8. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183605. [PMID: 33766534 DOI: 10.1016/j.bbamem.2021.183605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/01/2021] [Accepted: 03/12/2021] [Indexed: 12/18/2022]
Abstract
Light absorption by rhodopsin leads to the release of all-trans retinal (ATRal) in the lipid phase of photoreceptor disc membranes. Retinol dehydrogenase 8 (RDH8) then reduces ATRal into all-trans retinol, which is the first step of the visual cycle. The membrane binding of RDH8 has been postulated to be mediated by one or more palmitoylated cysteines located in its C-terminus. Different peptide variants of the C-terminus of RDH8 were thus used to obtain information on the mechanism of membrane binding of this enzyme. Steady-state and time-resolved fluorescence measurements were performed using short and long C-terminal segments of bovine RDH8, comprising one or two tryptophan residues. The data demonstrate that the amphipathic alpha helical structure of the first portion of the C-terminus of RDH8 strongly contributes to its membrane binding, which is also favored by palmitoylation of at least one of the cysteines located in the last portion of the C-terminus.
Collapse
|
27
|
Uyama H, Mandai M, Takahashi M. Stem-cell-based therapies for retinal degenerative diseases: Current challenges in the establishment of new treatment strategies. Dev Growth Differ 2021; 63:59-71. [PMID: 33315237 PMCID: PMC7986097 DOI: 10.1111/dgd.12704] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/13/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022]
Abstract
Various advances have been made in the treatment of retinal diseases, including new treatment strategies and innovations in surgical devices. However, the treatment of degenerative retinal diseases, such as retinitis pigmentosa (RP) and age-related macular degeneration (AMD), continues to pose a significant challenge. In this review, we focus on the use of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) to treat retinal diseases by harnessing the ability of stem cells to differentiate into different body tissues. The retina is a tissue specialized for light sensing, and its degradation leads to vision loss. As part of the central nervous system, the retina has very low regenerative capability, and therefore, treatment options are limited once it degenerates. Nevertheless, innovations in methods to induce the generation of retinal cells and tissues from ESCs/iPSCs enable the development of novel approaches for these irreversible diseases. Here we review some historical background and current clinical trials involving the use of stem-cell-derived retinal pigment epithelial cells for AMD treatment and stem cell-derived retinal cells/tissues for RP therapy. Finally, we discuss our future vision of regenerative treatment for retinal diseases with a partial focus on our studies and introduce other interesting approaches for restoring vision.
Collapse
Affiliation(s)
- Hirofumi Uyama
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Ophthalmology, Kobe City Eye Hospital, Kobe, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Ophthalmology, Kobe City Eye Hospital, Kobe, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Ophthalmology, Kobe City Eye Hospital, Kobe, Japan
| |
Collapse
|
28
|
Chen C, Chen J, Wang Y, Liu Z, Wu Y. Ferroptosis drives photoreceptor degeneration in mice with defects in all-trans-retinal clearance. J Biol Chem 2020; 296:100187. [PMID: 33334878 PMCID: PMC7948481 DOI: 10.1074/jbc.ra120.015779] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
The death of photoreceptor cells in dry age-related macular degeneration (AMD) and autosomal recessive Stargardt disease (STGD1) is closely associated with disruption in all-trans-retinal (atRAL) clearance in neural retina. In this study, we reveal that the overload of atRAL leads to photoreceptor degeneration through activating ferroptosis, a nonapoptotic form of cell death. Ferroptosis of photoreceptor cells induced by atRAL resulted from increased ferrous ion (Fe2+), elevated ACSL4 expression, system Xc- inhibition, and mitochondrial destruction. Fe2+ overload, tripeptide glutathione (GSH) depletion, and damaged mitochondria in photoreceptor cells exposed to atRAL provoked reactive oxygen species (ROS) production, which, together with ACSL4 activation, promoted lipid peroxidation and thereby evoked ferroptotic cell death. Moreover, exposure of photoreceptor cells to atRAL activated COX2, a well-accepted biomarker for ferroptosis onset. In addition to GSH supplement, inhibiting either Fe2+ by deferoxamine mesylate salt (DFO) or lipid peroxidation with ferrostatin-1 (Fer-1) protected photoreceptor cells from ferroptosis caused by atRAL. Abca4-/-Rdh8-/- mice exhibiting defects in atRAL clearance is an animal model for dry AMD and STGD1. We observed that ferroptosis was indeed present in neural retina of Abca4-/-Rdh8-/- mice after light exposure. More importantly, photoreceptor atrophy and ferroptosis in light-exposed Abca4-/-Rdh8-/- mice were effectively alleviated by intraperitoneally injected Fer-1, a selective inhibitor of ferroptosis. Our study suggests that ferroptosis is one of the important pathways of photoreceptor cell death in retinopathies arising from excess atRAL accumulation and should be pursued as a novel target for protection against dry AMD and STGD1.
Collapse
Affiliation(s)
- Chao Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, Fujian, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen City, Fujian, China
| | - Yan Wang
- Department of Ophthalmology, Shenzhen Hospital, Southern Medical University, Shenzhen City, Guangdong, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, Fujian, China
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, Fujian, China; Xiamen Eye Center of Xiamen University, Xiamen City, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen City, Guangdong, China.
| |
Collapse
|
29
|
Ortega JT, Parmar T, Golczak M, Jastrzebska B. Protective Effects of Flavonoids in Acute Models of Light-Induced Retinal Degeneration. Mol Pharmacol 2020; 99:60-77. [PMID: 33154094 DOI: 10.1124/molpharm.120.000072] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
Degeneration of photoreceptors caused by excessive illumination, inherited mutations, or aging is the principal pathology of blinding diseases. Pharmacological compounds that stabilize the visual receptor rhodopsin and modulate the cellular pathways triggering death of photoreceptors could avert this pathology. Interestingly, flavonoids can modulate the cellular processes, such as oxidative stress, inflammatory responses, and apoptosis, that are activated during retinal degeneration. As we found previously, flavonoids also bind directly to unliganded rod opsin, enhancing its folding, stability, and regeneration. In addition, flavonoids stimulate rhodopsin gene expression. Thus, we evaluated the effect of two main dietary flavonoids, quercetin and myricetin, in ATP-binding cassette subfamily A member 4 -/- /retinol dehydrogenase 8 -/- and wild-type BALB/c mice susceptible to light-induced photoreceptor degeneration. Using in vivo imaging, such as optical coherence tomography, scanning laser ophthalmoscopy, and histologic assessment of retinal morphology, we found that treatment with these flavonoids prior to light insult remarkably protected retina from deterioration and preserved its function. Using high-performance liquid chromatography-mass spectrometry analysis, we detected these flavonoids in the eye upon their intraperitoneal administration. The molecular events associated with the protective effect of quercetin and myricetin were related to the elevated expression of photoreceptor-specific proteins, rhodopsin and cone opsins, decreased expression of the specific inflammatory markers, and the shift of the equilibrium between cell death regulators BCL2-associated X protein (BAX) and B-cell lymphoma 2 toward an antiapoptotic profile. These results were confirmed in photoreceptor-derived 661W cells treated with either H2O2 or all-trans-retinal stressors implicated in the mechanism of retinal degeneration. Altogether, flavonoids could have significant prophylactic value for retinal degenerative diseases. SIGNIFICANCE STATEMENT: Flavonoids commonly present in food exhibit advantageous effects in blinding diseases. They bind to and stabilize unliganded rod opsin, which in excess accelerates degenerative processes in the retina. Additionally, flavonoids enhance the expression of the visual receptors, rod and cone opsins; inhibit the inflammatory reactions; and induce the expression of antiapoptotic markers in the retina, preventing the degeneration in vivo. Thus, flavonoids could have a prophylactic value for retinal degenerative diseases.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Tanu Parmar
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Marcin Golczak
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Beata Jastrzebska
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
30
|
Widjaja-Adhi MAK, Golczak M. The molecular aspects of absorption and metabolism of carotenoids and retinoids in vertebrates. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158571. [PMID: 31770587 PMCID: PMC7244374 DOI: 10.1016/j.bbalip.2019.158571] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Vitamin A is an essential nutrient necessary for numerous basic physiological functions, including reproduction and development, immune cell differentiation and communication, as well as the perception of light. To evade the dire consequences of vitamin A deficiency, vertebrates have evolved specialized metabolic pathways that enable the absorption, transport, and storage of vitamin A acquired from dietary sources as preformed retinoids or provitamin A carotenoids. This evolutionary advantage requires a complex interplay between numerous specialized retinoid-transport proteins, receptors, and enzymes. Recent advances in molecular and structural biology resulted in a rapid expansion of our understanding of these processes at the molecular level. This progress opened new avenues for the therapeutic manipulation of retinoid homeostasis. In this review, we summarize current research related to the biochemistry of carotenoid and retinoid-processing proteins with special emphasis on the structural aspects of their physiological actions. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
Affiliation(s)
- Made Airanthi K Widjaja-Adhi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Marcin Golczak
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America; Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America.
| |
Collapse
|
31
|
Feldman T, Yakovleva M, Viljanen M, Lindström M, Donner K, Ostrovsky M. Dark-adaptation in the eyes of a lake and a sea population of opossum shrimp (Mysis relicta): retinoid isomer dynamics, rhodopsin regeneration, and recovery of light sensitivity. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2020; 206:871-889. [PMID: 32880702 PMCID: PMC7603447 DOI: 10.1007/s00359-020-01444-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 11/30/2022]
Abstract
We have studied dark-adaptation at three levels in the eyes of the crustacean Mysis relicta over 2-3 weeks after exposing initially dark-adapted animals to strong white light: regeneration of 11-cis retinal through the retinoid cycle (by HPLC), restoration of native rhodopsin in photoreceptor membranes (by MSP), and recovery of eye photosensitivity (by ERG). We compare two model populations ("Sea", Sp, and "Lake", Lp) inhabiting, respectively, a low light and an extremely dark environment. 11-cis retinal reached 60-70% of the pre-exposure levels after 2 weeks in darkness in both populations. The only significant Lp/Sp difference in the retinoid cycle was that Lp had much higher levels of retinol, both basal and light-released. In Sp, rhodopsin restoration and eye photoresponse recovery parallelled 11-cis retinal regeneration. In Lp, however, even after 3 weeks only ca. 25% of the rhabdoms studied had incorporated new rhodopsin, and eye photosensitivity showed only incipient recovery from severe depression. The absorbance spectra of the majority of the Lp rhabdoms stayed constant around 490-500 nm, consistent with metarhodopsin II dominance. We conclude that sensitivity recovery of Sp eyes was rate-limited by the regeneration of 11-cis retinal, whilst that of Lp eyes was limited by inertia in photoreceptor membrane turnover.
Collapse
Affiliation(s)
- Tatiana Feldman
- Department of Molecular Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, Russia, 119991.,Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygin st. 4, Moscow, Russia, 119334
| | - Marina Yakovleva
- Department of Molecular Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, Russia, 119991
| | - Martta Viljanen
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Magnus Lindström
- Tvärminne Zoological Station, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Kristian Donner
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| | - Mikhail Ostrovsky
- Department of Molecular Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, Russia, 119991.,Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygin st. 4, Moscow, Russia, 119334
| |
Collapse
|
32
|
Sears AE, Albiez S, Gulati S, Wang B, Kiser P, Kovacik L, Engel A, Stahlberg H, Palczewski K. Single particle cryo-EM of the complex between interphotoreceptor retinoid-binding protein and a monoclonal antibody. FASEB J 2020; 34:13918-13934. [PMID: 32860273 PMCID: PMC7589273 DOI: 10.1096/fj.202000796rr] [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: 04/02/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 01/08/2023]
Abstract
Interphotoreceptor retinoid‐binding protein (IRBP) is a highly expressed protein secreted by rod and cone photoreceptors that has major roles in photoreceptor homeostasis as well as retinoid and polyunsaturated fatty acid transport between the neural retina and retinal pigment epithelium. Despite two crystal structures reported on fragments of IRBP and decades of research, the overall structure of IRBP and function within the visual cycle remain unsolved. Here, we studied the structure of native bovine IRBP in complex with a monoclonal antibody (mAb5) by cryo‐electron microscopy, revealing the tertiary and quaternary structure at sufficient resolution to clearly identify the complex components. Complementary mass spectrometry experiments revealed the structure and locations of N‐linked carbohydrate post‐translational modifications. This work provides insight into the structure of IRBP, displaying an elongated, flexible three‐dimensional architecture not seen among other retinoid‐binding proteins. This work is the first step in elucidation of the function of this enigmatic protein.
Collapse
Affiliation(s)
- Avery E Sears
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, USA.,Department of Ophthalmology, Gavin Herbert Eye Institute, University of California-Irvine, Irvine, CA, USA
| | - Stefan Albiez
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Basel, Switzerland
| | | | - Benlian Wang
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH, USA
| | - Philip Kiser
- Department of Physiology & Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, USA.,Research Service, VA Long Beach Healthcare System, Long Beach, CA, USA
| | - Lubomir Kovacik
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Basel, Switzerland
| | - Andreas Engel
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Basel, Switzerland
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Basel, Switzerland
| | - Krzysztof Palczewski
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California-Irvine, Irvine, CA, USA.,Department of Physiology & Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, USA.,Department of Chemistry, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
33
|
Sun D, Schur RM, Sears AE, Gao SQ, Sun W, Naderi A, Kern T, Palczewski K, Lu ZR. Stable Retinoid Analogue Targeted Dual pH-Sensitive Smart Lipid ECO/pDNA Nanoparticles for Specific Gene Delivery in the Retinal Pigment Epithelium. ACS APPLIED BIO MATERIALS 2020; 3:3078-3086. [PMID: 34327311 DOI: 10.1021/acsabm.0c00130] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Da Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Rebecca M Schur
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Avery E Sears
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106
| | - Song-Qi Gao
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Wenyu Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Amirreza Naderi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Timothy Kern
- Department of Ophthalmology, University of California Irvine, Irvine, CA 92697-7600
| | - Krzysztof Palczewski
- Department of Ophthalmology, University of California Irvine, Irvine, CA 92697-7600
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
34
|
Liao C, Cai B, Feng Y, Chen J, Wu Y, Zhuang J, Liu Z, Wu Y. Activation of JNK signaling promotes all- trans-retinal-induced photoreceptor apoptosis in mice. J Biol Chem 2020; 295:6958-6971. [PMID: 32265302 DOI: 10.1074/jbc.ra120.013189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/02/2020] [Indexed: 11/06/2022] Open
Abstract
Disrupted clearance of all-trans-retinal (atRAL), a component of the visual (retinoid) cycle in the retina, may cause photoreceptor atrophy in autosomal recessive Stargardt disease (STGD1) and dry age-related macular degeneration (AMD). However, the mechanisms underlying atRAL-induced photoreceptor loss remain elusive. Here, we report that atRAL activates c-Jun N-terminal kinase (JNK) signaling at least partially through reactive oxygen species production, which promoted mitochondria-mediated caspase- and DNA damage-dependent apoptosis in photoreceptor cells. Damage to mitochondria in atRAL-exposed photoreceptor cells resulted from JNK activation, leading to decreased expression of Bcl2 apoptosis regulator (Bcl2), increased Bcl2 antagonist/killer (Bak) levels, and cytochrome c (Cyt c) release into the cytosol. Cytosolic Cyt c specifically provoked caspase-9 and caspase-3 activation and thereby initiated apoptosis. Phosphorylation of JNK in atRAL-loaded photoreceptor cells induced the appearance of γH2AX, a sensitive marker for DNA damage, and was also associated with apoptosis onset. Suppression of JNK signaling protected photoreceptor cells against atRAL-induced apoptosis. Moreover, photoreceptor cells lacking Jnk1 and Jnk2 genes were more resistant to atRAL-associated cytotoxicity. The Abca4 -/- Rdh8 -/- mouse model displays defects in atRAL clearance that are characteristic of STGD1 and dry AMD. We found that JNK signaling was activated in the neural retina of light-exposed Abca4 -/- Rdh8 -/- mice. Of note, intraperitoneal administration of JNK-IN-8, which inhibits JNK signaling, effectively ameliorated photoreceptor degeneration and apoptosis in light-exposed Abca4 -/- Rdh8 -/- mice. We propose that pharmacological inhibition of JNK signaling may represent a therapeutic strategy for preventing photoreceptor loss in retinopathies arising from atRAL overload.
Collapse
Affiliation(s)
- Chunyan Liao
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, FJ 361102, China
| | - Binxiang Cai
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, FJ 361102, China
| | - Yufeng Feng
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen City, FJ 361003, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen City, FJ 361102, China
| | - Yiping Wu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, FJ 361102, China
| | - Jingbin Zhuang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, FJ 361102, China
| | - Zuguo Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, FJ 361102, China
| | - Yalin Wu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen City, FJ 361102, China .,Xiamen Eye Center of Xiamen University, Xiamen City, FJ 361001, China.,Shenzhen Research Institute of Xiamen University, Shenzhen City, GD 518063, China
| |
Collapse
|
35
|
Lešták J, Fůs M. Neuroprotection in glaucoma-electrophysiology. Exp Ther Med 2020; 19:2401-2405. [PMID: 32256717 PMCID: PMC7086198 DOI: 10.3892/etm.2020.8509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/06/2019] [Indexed: 11/15/2022] Open
Abstract
Hypertensive glaucoma is defined as a group of diseases with progressive loss of the neuroretinal margin of the optic disc that causes characteristic degenerative optic neuropathy. The present study provided an updated summary of the physiology and pathology of neurotransmission in the visual path, with the focus on glaucoma. The results of positron emission tomography, functional magnetic resonance imaging and mainly electrophysiological methods demonstrated pathogenesis of nerve cell damage in the visual pathway. Based on these conclusions, neuroprotection in glaucoma was proposed. This consists mainly of the reduction of the intraocular pressure. It is followed by a decrease of glutamate in the synaptic cleft and blockade of its binding to the NMDA receptors. The supply of energy substrates to altered nerve cells is also indispensable. Therapy should be systemic due to impairment of the complete visual path.
Collapse
Affiliation(s)
- Ján Lešták
- Eye Clinic, JL Faculty of Biomedical Engineering CTU in Prague, 158 00 Prague 5, Czech Republic
- CTU in Prague, Faculty of Biomedical Engineering, 272 01 Kladno 2, Czech Republic
| | - Martin Fůs
- Eye Clinic, JL Faculty of Biomedical Engineering CTU in Prague, 158 00 Prague 5, Czech Republic
- CTU in Prague, Faculty of Biomedical Engineering, 272 01 Kladno 2, Czech Republic
| |
Collapse
|
36
|
Reay WR, Cairns MJ. The role of the retinoids in schizophrenia: genomic and clinical perspectives. Mol Psychiatry 2020; 25:706-718. [PMID: 31666680 PMCID: PMC7156347 DOI: 10.1038/s41380-019-0566-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/23/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022]
Abstract
Signalling by retinoid compounds is vital for embryonic development, with particular importance for neurogenesis in the human brain. Retinoids, metabolites of vitamin A, exert influence over the expression of thousands of transcripts genome wide, and thus, act as master regulators of many important biological processes. A significant body of evidence in the literature now supports dysregulation of the retinoid system as being involved in the aetiology of schizophrenia. This includes mechanistic insights from large-scale genomic, transcriptomic and, proteomic studies, which implicate disruption of disparate aspects of retinoid biology such as transport, metabolism, and signalling. As a result, retinoids may present a valuable clinical opportunity in schizophrenia via novel pharmacotherapies and dietary intervention. Further work, however, is required to expand on the largely observational data collected thus far and confirm causality. This review will highlight the fundamentals of retinoid biology and examine the evidence for retinoid dysregulation in schizophrenia.
Collapse
Affiliation(s)
- William R. Reay
- 0000 0000 8831 109Xgrid.266842.cSchool of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW Australia ,grid.413648.cCentre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW Australia
| | - Murray J. Cairns
- 0000 0000 8831 109Xgrid.266842.cSchool of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW Australia ,grid.413648.cCentre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW Australia
| |
Collapse
|
37
|
Comparison between the enzymatic activity, structure and substrate binding of mouse and human lecithin retinol acyltransferase. Biochem Biophys Res Commun 2019; 519:832-837. [PMID: 31561851 DOI: 10.1016/j.bbrc.2019.09.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 11/22/2022]
Abstract
Lecithin retinol acyltransferase (LRAT) is involved in the visual cycle where it catalyzes the formation of all-trans retinyl ester. The mouse animal model has been widely used to study LRAT. Primary sequence alignment shows 80% identity and 90% similarity between human and mouse LRAT. However, human LRAT has a proline at position 173 (hLRAT (P173)) while an arginine can be found at this position for the mouse protein (mLRAT (R173)). Moreover, residue 173 is important for the human protein since a substitution mutation of this residue to a leucine (P173L-hLRAT) caused night blindness in a patient. The present study was thus undertaken to determine whether mouse and human LRAT have a similar enzymatic activity, structure and substrate binding affinity using a truncated form of LRAT (tLRAT). The enzymatic activity and binding affinity to the substrate, all-trans retinol, of mtLRAT (R173) were found to be 2.7- and 3.9-fold lower, respectively, than that of htLRAT (P173). Moreover, the enzymatic activity of P173L-htLRAT is 6.3-fold lower compared to that of htLRAT (P173). Furthermore, a significant difference was observed between the intrinsic fluorescence emission as well as between the circular dichroism spectra of mtLRAT (R173) and htLRAT (P173). In addition, mtLRAT proteins are less thermostable than htLRAT proteins, which suggests that structural differences exist between the mouse and human proteins. Altogether, these data strongly suggest that the much lower catalytic activity of mtLRAT (R173) compared to that of htLRAT (P173) mostly results from differences between their structure, predominantly revealed by their dissimilar thermal stability, as well as their efficiency to bind all-trans retinol. Therefore, conclusions regarding the behavior of human LRAT based on measurements performed with mouse LRAT must be made with caution. Also, the much lower enzymatic activity of P173L-htLRAT compared to that of htLRAT (P173) might explain the night blindness of a patient carrying this mutation.
Collapse
|
38
|
Xiao T, Ma J, Jiang J, Gan M, Lu B, Luo R, Liu Q, Zhang Q, Liu Z, Zhai J. Rod-Cell-Mimetic Photochromic Layered Ion Channels with Multiple Switchable States for Controllable Ion Transport. Chemistry 2019; 25:12795-12800. [PMID: 31376182 DOI: 10.1002/chem.201902450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/19/2019] [Indexed: 01/10/2023]
Abstract
The controllable ion transport in the photoreceptors of rod cells is essentially important for the light detection and information transduction in visual systems. Herein, inspired by the photochromism-regulated ion transport in rod cells with stacking structure, layered ion channels have been developed with a visual photochromic function induced by the alternate irradiation with visible and UV light. The layered structure is formed by stacking spiropyran-modified montmorillonite 2D nanosheets on the surface of an alumina nanoporous membrane. The visual photochromism resulting from the photoisomerization of spiropyran chromophores reversibly regulates the ion transport through layered ion channels. Furthermore, the cooperation of photochromism and pH value achieves multiple switchable states of layered ion channels for the controllable ion transport mimicking the biological process of the visual cycle. The ion transport properties of these states are explained quantitatively by a theoretical calculation based on the Poisson and Nernst-Plank (PNP) equations.
Collapse
Affiliation(s)
- Tianliang Xiao
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Jing Ma
- School of Space and Environment, Beihang University, Beijing, 100124, P. R. China
| | - Jiaqiao Jiang
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Mengke Gan
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Bingxin Lu
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Rifeng Luo
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Qingqing Liu
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Qianqian Zhang
- The College of Materials Science and Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Zhaoyue Liu
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Jin Zhai
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, P. R. China
| |
Collapse
|
39
|
Araki H, Miura F, Watanabe A, Morinaga C, Kitaoka F, Kitano Y, Sakai N, Shibata Y, Terada M, Goto S, Yamanaka S, Takahashi M, Ito T. Base-Resolution Methylome of Retinal Pigment Epithelial Cells Used in the First Trial of Human Induced Pluripotent Stem Cell-Based Autologous Transplantation. Stem Cell Reports 2019; 13:761-774. [PMID: 31564644 PMCID: PMC6829753 DOI: 10.1016/j.stemcr.2019.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/19/2022] Open
Abstract
The first-in-human trial of induced pluripotent stem cell (iPSC)-based autologous transplantation was successfully performed on a female patient with age-related macular degeneration. Here we delineated the base-resolution methylome of the iPSC-derived retinal pigment epithelium (iRPE) used in this trial. The methylome of iRPE closely resembled that of native RPE (nRPE), although partially methylated domains (PMDs) emerged in iRPE but not nRPE. Most differentially methylated regions between iRPE and nRPE appeared to originate from (de)methylation errors during differentiation, whereas errors at reprogramming resulted in aberrant genomic imprinting and X chromosome reactivation. Moreover, non-CpG methylation was prominent in nRPE but not iRPE. Intriguingly, xenotransplantation to mouse remodeled the iRPE methylome to demethylate a subset of suppressed genes and accumulate non-CpG methylation, but failed to resolve PMDs and hypermethylated CpG islands. Although the impacts of these alterations remain elusive, our findings should provide a useful guide for methylome analyses of other iPSC-derived cells. The methylome of iPSC-derived RPE closely resembled that of native RPE Most methylomic differences originated from errors during differentiation Errors at reprogramming induced loss of imprinting and X chromosome reactivation Some of the differences were mitigated by xenotransplantation to mouse
Collapse
Affiliation(s)
- Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Chikako Morinaga
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe 650-0047, Japan
| | - Fumiyo Kitaoka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yuko Kitano
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Noriko Sakai
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe 650-0047, Japan
| | - Yumiko Shibata
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe 650-0047, Japan
| | - Motoki Terada
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe 650-0047, Japan
| | - So Goto
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe 650-0047, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe 650-0047, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan.
| |
Collapse
|
40
|
Abstract
Retinal function has long been studied with psychophysical methods in humans, whereas detailed functional studies of vision have been conducted mostly in animals owing to the invasive nature of physiological approaches. There are exceptions to this generalization, for example, the electroretinogram. This review examines exciting recent advances using in vivo retinal imaging to understand the function of retinal neurons. In some cases, the methods have existed for years and are still being optimized. In others, new methods such as optophysiology are revealing novel patterns of retinal function in animal models that have the potential to change our understanding of the functional capacity of the retina. Together, the advances in retinal imaging mark an important milestone that shifts attention away from anatomy alone and begins to probe the function of healthy and diseased eyes.
Collapse
Affiliation(s)
- Jennifer J Hunter
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York 14604, USA; , ,
- The Institute of Optics and Department of Biomedical Engineering, University of Rochester, Rochester, New York 14604, USA
| | - William H Merigan
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York 14604, USA; , ,
| | - Jesse B Schallek
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York 14604, USA; , ,
- Department of Neuroscience, University of Rochester, Rochester, New York 14604, USA
| |
Collapse
|
41
|
Kahremany S, Sander CL, Tochtrop GP, Kubas A, Palczewski K. Z-isomerization of retinoids through combination of monochromatic photoisomerization and metal catalysis. Org Biomol Chem 2019; 17:8125-8139. [PMID: 31455964 DOI: 10.1039/c9ob01645g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Catalytic Z-isomerization of retinoids to their thermodynamically less stable Z-isomer remains a challenge. In this report, we present a photochemical approach for the catalytic Z-isomerization of retinoids using monochromatic wavelength UV irradiation treatment. We have developed a straightforward approach for the synthesis of Z-retinoids in high yield, overcoming common obstacles normally associated with their synthesis. Calculations based on density functional theory (DFT) have allowed us to correlate the experimentally observed Z-isomer distribution of retinoids with the energies of chemically important intermediates, which include ground- and excited-state potential energy surfaces. We also demonstrate the application of the current method by synthesizing gram-scale quantities of 9-cis-retinyl acetate 9Z-a. Operational simplicity and gram-scale ability make this chemistry a very practical solution to the problem of Z-isomer retinoid synthesis.
Collapse
Affiliation(s)
- Shirin Kahremany
- Gavin Herbert Eye Institute and the Department of Ophthalmology, University of California, Irvine, California 92697, USA.
| | - Christopher Lane Sander
- Gavin Herbert Eye Institute and the Department of Ophthalmology, University of California, Irvine, California 92697, USA. and Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Adam Kubas
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland.
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute and the Department of Ophthalmology, University of California, Irvine, California 92697, USA.
| |
Collapse
|
42
|
Kahremany S, Kubas A, Tochtrop GP, Palczewski K. Catalytic synthesis of 9-cis-retinoids: mechanistic insights. Dalton Trans 2019; 48:10581-10595. [PMID: 31218312 PMCID: PMC7004310 DOI: 10.1039/c9dt02189b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The regioselective Z-isomerization of thermodynamically stable all-trans retinoids remains challenging, and ultimately limits the availability of much needed therapeutics for the treatment of human diseases. We present here a novel, straightforward approach for the catalytic Z-isomerization of retinoids using conventional heat treatment or microwave irradiation. A screen of 20 transition metal-based catalysts identified an optimal approach for the regioselective production of Z-retinoids. The most effective catalytic system was comprised of a palladium complex with labile ligands. Several mechanistic studies, including isotopic H/D exchange and state-of-the-art quantum chemical calculations using coupled cluster methods indicate that the isomerization is initiated by catalyst dimerization followed by the formation of a cyclic, six-membered chloropalladate catalyst-substrate adduct, which eventually opens to produce the desired Z-isomer. The synthetic development described here, combined with thorough mechanistic analysis of the underlying chemistry, highlights the use of readily available transition metal-based catalysts in straightforward formats for gram-scale drug synthesis.
Collapse
Affiliation(s)
- Shirin Kahremany
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA 92697, USA. and Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Adam Kubas
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland.
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA 92697, USA. and Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
43
|
Hu Y, Wang X, Hu B, Mao Y, Chen Y, Yan L, Yong J, Dong J, Wei Y, Wang W, Wen L, Qiao J, Tang F. Dissecting the transcriptome landscape of the human fetal neural retina and retinal pigment epithelium by single-cell RNA-seq analysis. PLoS Biol 2019; 17:e3000365. [PMID: 31269016 PMCID: PMC6634428 DOI: 10.1371/journal.pbio.3000365] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/16/2019] [Accepted: 06/25/2019] [Indexed: 12/21/2022] Open
Abstract
The developmental pathway of the neural retina (NR) and retinal pigment epithelium (RPE) has been revealed by extensive research in mice. However, the molecular mechanisms underlying the development of the human NR and RPE, as well as the interactions between these two tissues, have not been well defined. Here, we analyzed 2,421 individual cells from human fetal NR and RPE using single-cell RNA sequencing (RNA-seq) technique and revealed the tightly regulated spatiotemporal gene expression network of human retinal cells. We identified major cell classes of human fetal retina and potential crucial transcription factors for each cell class. We dissected the dynamic expression patterns of visual cycle- and ligand-receptor interaction-related genes in the RPE and NR. Moreover, we provided a map of disease-related genes for human fetal retinal cells and highlighted the importance of retinal progenitor cells as potential targets of inherited retinal diseases. Our findings captured the key in vivo features of the development of the human NR and RPE and offered insightful clues for further functional studies.
Collapse
Affiliation(s)
- Yuqiong Hu
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Xiaoye Wang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Boqiang Hu
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Yunuo Mao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Yidong Chen
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Liying Yan
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Jun Yong
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Ji Dong
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Yuan Wei
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Wei Wang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Lu Wen
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Jie Qiao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, Third Hospital, College of Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Biomedical Pioneering Innovation Center, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
44
|
Getter T, Suh S, Hoang T, Handa JT, Dong Z, Ma X, Chen Y, Blackshaw S, Palczewski K. The selective estrogen receptor modulator raloxifene mitigates the effect of all- trans-retinal toxicity in photoreceptor degeneration. J Biol Chem 2019; 294:9461-9475. [PMID: 31073029 DOI: 10.1074/jbc.ra119.008697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Indexed: 12/19/2022] Open
Abstract
The retinoid cycle is a metabolic process in the vertebrate retina that continuously regenerates 11-cis-retinal (11-cisRAL) from the all-trans-retinal (atRAL) isomer. atRAL accumulation can cause photoreceptor degeneration and irreversible visual dysfunction associated with incurable blinding retinal diseases, such as Stargardt disease, retinitis pigmentosa (RP), and atrophic age-related macular degeneration (AMD). The underlying cellular mechanisms leading to retinal degeneration remain uncertain, although previous studies have shown that atRAL promotes calcium influx associated with cell apoptosis. To identify compounds that mitigate the effects of atRAL toxicity, here we developed an unbiased and robust image-based assay that can detect changes in intracellular calcium levels in U2OS cells. Using our assay in a high-throughput screen of 2,400 compounds, we noted that selective estrogen receptor modulators (SERMs) potently stabilize intracellular calcium and thereby counteract atRAL-induced toxicity. In a light-induced retinal degeneration mouse model (Abca4 -/- Rdh8 -/-), raloxifene (a benzothiophene-type scaffold SERM) prevented the onset of photoreceptor apoptosis and thus protected the retina from degeneration. The minor structural differences between raloxifene and one of its derivatives (Y 134) had a major impact on calcium homeostasis after atRAL exposure in vitro, and we verified this differential impact in vivo In summary, the SERM raloxifene has structural and functional neuroprotective effects in the retina. We propose that the highly sensitive image-based assay developed here could be applied for the discovery of additional drug candidates preventing photoreceptor degeneration.
Collapse
Affiliation(s)
- Tamar Getter
- From the Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California 92697, .,the Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Susie Suh
- From the Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California 92697.,the Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Thanh Hoang
- the Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - James T Handa
- the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | | | - Xiuli Ma
- Polgenix Inc., Irvine, California 92617
| | - Yuanyuan Chen
- the Department of Ophthalmology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, and.,the McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Seth Blackshaw
- the Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Krzysztof Palczewski
- From the Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California 92697, .,the Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
45
|
Vancura P, Csicsely E, Leiser A, Iuvone PM, Spessert R. Rhythmic Regulation of Photoreceptor and RPE Genes Important for Vision and Genetically Associated With Severe Retinal Diseases. Invest Ophthalmol Vis Sci 2019; 59:3789-3799. [PMID: 30073352 PMCID: PMC6071477 DOI: 10.1167/iovs.18-24558] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The aim of the present study was to identify candidate genes for mediating daily adjustment of vision. Methods Genes important for vision and genetically associated with severe retinal diseases were tested for 24-hour rhythms in transcript levels in neuronal retina, microdissected photoreceptors, photoreceptor-related pinealocytes, and retinal pigment epithelium-choroid (RPE-choroid) complex by using quantitative PCR. Results Photoreceptors of wildtype mice display circadian clock-dependent regulation of visual arrestins (Arr1, Arr4) and the visual cycle gene Rdh12, whereas cells of the RPE-choroid exhibit light-dependent regulation of the visual cycle key genes Lrat, Rpe65, and Rdh5. Clock-driven rhythmicity of Arr1, Arr4, and Rdh12 was observed also in rat pinealocytes, to persist in a mouse model of diabetic retinopathy (db/db) and, in the case of Arr1, to be abolished in retinae of mice deficient for dopamine D4 receptors. Therefore, the expression rhythms appear to be evolutionary conserved, to be unaffected in diabetic retinopathy, and, for Arr1, to require dopamine signaling via dopamine D4 receptors. Conclusions The data of the present study suggest that daily adjustment of retinal function combines clock-dependent regulation of genes responsible for phototransduction termination (Arr1, Arr4) and detoxification (Rdh12) in photoreceptors with light-dependent regulation of genes responsible for retinoid recycling (Lrat, Rpe65, and Rdh5) in RPE. Furthermore, they indicate circadian and light-dependent regulation of genes genetically associated with severe retinal diseases.
Collapse
Affiliation(s)
- Patrick Vancura
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Erika Csicsely
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Annalisa Leiser
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Rainer Spessert
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
46
|
Gao XR, Huang H, Kim H. Genome-wide association analyses identify 139 loci associated with macular thickness in the UK Biobank cohort. Hum Mol Genet 2019; 28:1162-1172. [PMID: 30535121 PMCID: PMC6423416 DOI: 10.1093/hmg/ddy422] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 11/13/2022] Open
Abstract
The macula, located near the center of the retina in the human eye, is responsible for providing critical functions, such as central, sharp vision. Structural changes in the macula are associated with many ocular diseases, including age-related macular degeneration (AMD) and glaucoma. Although macular thickness is a highly heritable trait, there are no prior reported genome-wide association studies (GWASs) of it. Here we describe the first GWAS of macular thickness, which was measured by spectral-domain optical coherence tomography using 68 423 participants from the UK Biobank cohort. We identified 139 genetic loci associated with macular thickness at genome-wide significance (P < 5 × 10-8). The most significant loci were LINC00461 (P = 5.1 × 10-120), TSPAN10 (P = 1.2 × 10-118), RDH5 (P = 9.2 × 10-105) and SLC6A20 (P = 1.4 × 10-71). Results from gene expression demonstrated that these genes are highly expressed in the retina. Other hits included many previously reported AMD genes, such as NPLOC4 (P = 1.7 × 10-103), RAD51B (P = 9.1 × 10-14) and SLC16A8 (P = 1.7 × 10-8), further providing functional significance of the identified loci. Through cross-phenotype analysis, these genetic loci also exhibited pleiotropic effects with myopia, neurodegenerative diseases (e.g. Parkinson's disease, schizophrenia and Alzheimer's disease), cancer (e.g. breast, ovarian and lung cancers) and metabolic traits (e.g. body mass index, waist circumference and type 2 diabetes). Our findings provide the first insight into the genetic architecture of macular thickness and may further elucidate the pathogenesis of related ocular diseases, such as AMD.
Collapse
Affiliation(s)
- X Raymond Gao
- Departments of Ophthalmology and Visual Science and Biomedical Informatics, Division of Human Genetics, The Ohio State University, Columbus, OH, USA
| | - Hua Huang
- Departments of Ophthalmology and Visual Science and Biomedical Informatics, Division of Human Genetics, The Ohio State University, Columbus, OH, USA
| | - Heejin Kim
- Departments of Ophthalmology and Visual Science and Biomedical Informatics, Division of Human Genetics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
47
|
Harrison EH. Mechanisms of Transport and Delivery of Vitamin A and Carotenoids to the Retinal Pigment Epithelium. Mol Nutr Food Res 2019; 63:e1801046. [PMID: 30698921 DOI: 10.1002/mnfr.201801046] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/15/2019] [Indexed: 11/06/2022]
Abstract
Vision depends on the delivery of vitamin A (retinol) to the retina. Retinol in blood is bound to retinol-binding protein (RBP). Retinal pigment epithelia (RPE) cells express the RBP receptor, STRA6, that facilitates uptake of retinol. The retinol is then converted to retinyl esters by the enzyme lecithin:retinol acyltransferase. The esters are the substrate for RPE65, an enzyme that produces 11-cis retinol, which is converted to 11-cis retinaldehyde for transport to the photoreceptors to form rhodopsin. The dietary xanthophylls, lutein (LUT) and zeaxanthin (ZEA), accumulate in the macula of the eye, providing protection against age-related macular degeneration. To reach the macula, carotenoids cross the RPE. In blood, xanthophylls and β-carotene mostly associate with high-density lipoprotein (HDL) and low-density lipoprotein (LDL), respectively. Studies using a human RPE cell model evaluate the kinetics of cell uptake when carotenoids are delivered in LDL or HDL. For LUT and β-carotene, LDL delivery result in the highest rate of uptake. HDL is more effective in delivering ZEA (and meso-ZEA). This selective HDL-mediated uptake of ZEA, via a scavenger receptor and LDL-mediated uptake of LUT and β-carotene provides a mechanism for the selective accumulation of ZEA > LUT and xanthophylls over β-carotene in the macula.
Collapse
Affiliation(s)
- Earl H Harrison
- Department of Human Sciences, Program in Human Nutrition & Ohio State Biochemistry Program, Ohio State University, Columbus, OH, 43206, USA
| |
Collapse
|
48
|
Abstract
The retinal pigmented epithelium (RPE) is a single layer of polarized epithelial cells which plays many important roles for visual function. One of such roles is production of visual chromophore, 11-cis-retinal through the visual cycle. The visual cycle consists of biochemical processes for regenerating chromophore by a collective action of the RPE and photoreceptor. Photoreceptors harbor the G protein-coupled receptors, opsin which enables to receive light when it bounds to 11-cis-retinal. With absorption of a photon of light, 11-cis-retinal photoisomerizes to all-trans-retinal. All-trans-retinal reduces to all-trans-retinol in the photoreceptor and further recycles back to 11-cis-retinal in the RPE. Acyltransferases and isomerohydrolase(s) along with retinol dehydrogenases sequentially convert all-trans-retinol to 11-cis-retinal in the RPE. Dysfunctions of any retinoid cycle enzymes in the RPE can cause retinal diseases. Phenotyping RPE functions by the use of mutant mouse models will provide great detailed biochemical insights of the visual cycle and further manipulative strategies to protect against retinal degeneration. Here, we describe biochemical analyses of the visual cycle in mouse models using RPE cells.
Collapse
Affiliation(s)
- Bhubanananda Sahu
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
49
|
Ramlogan-Steel CA, Murali A, Andrzejewski S, Dhungel B, Steel JC, Layton CJ. Gene therapy and the adeno-associated virus in the treatment of genetic and acquired ophthalmic diseases in humans: Trials, future directions and safety considerations. Clin Exp Ophthalmol 2019; 47:521-536. [PMID: 30345694 DOI: 10.1111/ceo.13416] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/04/2018] [Accepted: 10/15/2018] [Indexed: 12/27/2022]
Abstract
Voretigene neparvovec-rzyl was recently approved for the treatment of Leber congenital amaurosis, and the use of gene therapy for eye disease is attracting even greater interest. The eye has immune privileged status, is easily accessible, requires a reduced dosage of therapy due to its size and is highly compartmentalized, significantly reducing systemic spread. Adeno-associated virus (AAV), with its low pathogenicity, prolonged expression profile and ability to transduce multiple cell types, has become the leading gene therapy vector. Target diseases have moved beyond currently untreatable inherited dystrophies to common, partially treatable acquired conditions such as exudative age-related macular degeneration and glaucoma, but use of the technology in these conditions imposes added obligations for caution in vector design. This review discusses the current status of AAV gene therapy trials in genetic and acquired ocular diseases, and explores new scientific developments, which could help ensure effective and safe use of the therapy in the future.
Collapse
Affiliation(s)
- Charmaine A Ramlogan-Steel
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia.,Medical and Applied Science, Central Queensland University, School of Health, Rockhampton, Australia
| | - Aparna Murali
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| | - Slawomir Andrzejewski
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| | - Bijay Dhungel
- Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| | - Jason C Steel
- Medical and Applied Science, Central Queensland University, School of Health, Rockhampton, Australia
| | - Christopher J Layton
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| |
Collapse
|
50
|
Baksheeva VE, Tiulina VV, Tikhomirova NK, Gancharova OS, Komarov SV, Philippov PP, Zamyatnin AA, Senin II, Zernii EY. Suppression of Light-Induced Oxidative Stress in the Retina by Mitochondria-Targeted Antioxidant. Antioxidants (Basel) 2018; 8:E3. [PMID: 30577635 PMCID: PMC6356525 DOI: 10.3390/antiox8010003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 01/06/2023] Open
Abstract
Light-induced oxidation of lipids and proteins provokes retinal injuries and results in progression of degenerative retinal diseases, such as, for instance, iatrogenic photic maculopathies. Having accumulated over years retinal injuries contribute to development of age-related macular degeneration (AMD). Antioxidant treatment is regarded as a promising approach to protecting the retina from light damage and AMD. Here, we examine oxidative processes induced in rabbit retina by excessive light illumination with or without premedication using mitochondria-targeted antioxidant SkQ1 (10-(6'-plastoquinonyl)decyltriphenyl-phosphonium). The retinal extracts obtained from animals euthanized within 1⁻7 days post exposure were analyzed for H₂O₂, malondialdehyde (MDA), total antioxidant activity (AOA), and activities of glutathione peroxidase (GPx) and superoxide dismutase (SOD) using colorimetric and luminescence assays. Oxidation of visual arrestin was monitored by immunoblotting. The light exposure induced lipid peroxidation and H₂O₂ accumulation in the retinal cells. Unexpectedly, it prominently upregulated AOA in retinal extracts although SOD and GPx activities were compromised. These alterations were accompanied by accumulation of disulfide dimers of arrestin revealing oxidative stress in the photoreceptors. Premedication of the eyes with SkQ1 accelerated normalization of H₂O₂ levels and redox-status of lipids and proteins, contemporarily enhancing AOA and, likely, sustaining normal activity of GPx. Thus, SkQ1 protects the retina from light-induced oxidative stress and could be employed to suppress oxidative damage of proteins and lipids contributing to AMD.
Collapse
Affiliation(s)
- Viktoriia E Baksheeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Veronika V Tiulina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Natalia K Tikhomirova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Olga S Gancharova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Sergey V Komarov
- Department of Biology and Pathology of Domestic, Laboratory and Exotic Animals, Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow 109472, Russia.
| | - Pavel P Philippov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Andrey A Zamyatnin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Ivan I Senin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Evgeni Yu Zernii
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| |
Collapse
|