1
|
Zaluski J, Bassetto M, Kiser PD, Tochtrop GP. Advances and therapeutic opportunities in visual cycle modulation. Prog Retin Eye Res 2025; 106:101360. [PMID: 40280538 DOI: 10.1016/j.preteyeres.2025.101360] [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: 02/14/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The visual cycle is a metabolic pathway that enables continuous vision by regenerating the 11-cis-retinal chromophore for photoreceptors opsins. Although integral to normal visual function, the flux of retinoids through this cycle can contribute to a range of retinal pathologies, including Stargardt disease, age-related macular degeneration, and diabetic retinopathy. In such conditions, intermediates and byproducts of the visual cycle, such as bisretinoid components of lipofuscin, can accumulate, concomitant with cellular damage and eventual photoreceptor loss. This has inspired efforts to modulate the visual cycle, aiming to slow or prevent the formation of these toxic intermediates and thus preserve retinal structure and function. Over the past two decades, multiple strategies to modulate the visual cycle have emerged. These include both intrinsic approaches, targeting key enzymes, retinoid-binding proteins, or receptors within the pigment epithelium or photoreceptors (e.g., RPE65, CRBP1, and rhodopsin inhibitors/antagonists) and extrinsic strategies that indirectly alter retinoid availability within the retina (e.g., RBP4 antagonists). Many of these agents have shown promise in animal models of visual cycle-associated retinal diseases, reducing pathological changes, and improving retinal survival. Several have advanced into clinical studies, although none are currently FDA-approved. Challenges remain in optimizing drug specificity and duration of action while minimizing side effects such as nyctalopia. In this review, we comprehensively examine current and emerging visual cycle modulators, discuss their medicinal chemistry, mechanisms of action, efficacy in preclinical and clinical studies, and highlight future opportunities for drug discovery aimed at safely and effectively preserving vision through modulation of this biochemical pathway.
Collapse
Affiliation(s)
- Jordan Zaluski
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marco Bassetto
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA
| | - Philip D Kiser
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA; Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, CA, 92697, USA.
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
2
|
Sassone F, Estay-Ahumada C, Roux MJ, Ciocca D, Rossolillo P, Birling MC, Sparrow JR, Montenegro D, Hicks D. Interruption of the visual cycle in a novel animal model induces progressive vision loss resembling Stargardts Disease. Sci Rep 2024; 14:30880. [PMID: 39730605 DOI: 10.1038/s41598-024-81869-y] [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: 04/19/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Mutations in the gene ABCA4 coding for photoreceptor-specific ATP-binding cassette subfamily A member 4, are responsible for Stargardts Disease type 1 (STGD1), the most common form of inherited macular degeneration. STGD1 typically declares early in life and leads to severe visual handicap. Abca4 gene-deletion mouse models of STGD1 accumulate lipofuscin, a hallmark of the disease, but unlike the human disease show no or only moderate structural changes and no functional decline. The human macula is highly enriched in cones, and reasoning that the low cone percentage in mice retinas (< 3%) might compromise faithful modelling of human maculopathies, we performed sub-retinal injections of CRISPR/Cas9-abca4 Adeno-Associated Virus constructs into young Sand Rats (Psammomys obesus), a diurnal rodent containing > 30% cones. Compared to control injections of AAV-abca4-GFP, treated eyes exhibited extensive retinal degeneration by two months. Sanger sequencing of the CRISPR targeted sequence show a clear edition of Abca4 gene. Non-invasive fundus imaging showed widespread photoreceptor loss, confirmed by ocular coherence tomography. Functional recording by single flash and flicker electroretinography showed significant decline in photopic (cone) light responses. Post-mortem real-time PCR, immunohistochemistry and western blotting showed significant decrease of cone-specific (MW cone opsin) but not rod-specific (rhodopsin) markers. Transmission electron microscopy showed large numbers of lipid inclusions in treated but not control retinal pigmented epithelium. Finally, ultra-high performance liquid chromatography analysis of whole P. obesus eyes showed the presence of all-trans retinal-dimer, not detected in rod-rich rat eyes. In conclusion, Abca4 knockout in P. obesus results in a predominantly cone degeneration phenotype, more accurately reflecting the etiology of human STGD1, and should be valuable for characterizing pathogenic pathways and exploring treatment options.
Collapse
Affiliation(s)
- Fabiana Sassone
- INCI-UPR3212-CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | | | - Michel J Roux
- IGBMC/Institut Clinique de La Souris - CNRS UMR 7104 Inserm U 1258, 1 Rue Laurent Fries, BP 10142, 67404, Illkirch CEDEX, France
| | - Dominique Ciocca
- Chronobiotron UAR3415-CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Paola Rossolillo
- IGBMC/Institut Clinique de La Souris - CNRS UMR 7104 Inserm U 1258, 1 Rue Laurent Fries, BP 10142, 67404, Illkirch CEDEX, France
| | - Marie-Christine Birling
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de La Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch Graffenstaden, France
| | - Janet R Sparrow
- Departments of Ophthalmology, and Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Diego Montenegro
- Departments of Ophthalmology, and Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - David Hicks
- INCI-UPR3212-CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France.
| |
Collapse
|
3
|
Lee M, Leskova W, Eshaq RS, Amezquita Z, Harris NR. Mechanisms of retinal photoreceptor loss in spontaneously hypertensive rats. Exp Eye Res 2024; 247:110065. [PMID: 39222765 PMCID: PMC11412233 DOI: 10.1016/j.exer.2024.110065] [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: 07/08/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Retinal neurodegenerative diseases, including hypertensive retinopathy, involve progressive damage to retinal neurons, leading to visual impairment. In this study, we investigated the pathological mechanisms underlying retinal neurodegeneration in spontaneously hypertensive rats (SHR), using Wistar Kyoto (WKY) rats as normotensive controls. We observed that SHR exhibited significantly higher blood pressure and decreased retinal thickness, indicating retinal neurodegeneration. Molecular tests including quantitative real-time polymerase chain reaction, immunoblot, and immunofluorescent staining showed elevated levels of the pro-inflammatory cytokine tumor necrosis factor-α, apoptotic markers (Fas, FasL, caspase-8, active caspase-3, and cleaved poly (ADP-ribose) polymerase), and necroptotic markers (receptor-interacting protein kinase-1 and -3) in SHR retinas. Additionally, we found elevated transforming growth factor-β (TGF-β) levels in the retinal pigment epithelium (RPE) of SHR, with a decrease in lecithin retinol acyltransferase (LRAT), which regulates retinoid metabolism and photoreceptor health. In human RPE cells (ARPE-19), TGF-β administration suppressed mRNA and protein levels of LRAT; and vactosertib, a selective inhibitor of TGF-β receptor kinase type 1, reversed the effect of TGF-β. These findings suggest that hypertension-induced retinal neurodegeneration involves inflammation, apoptosis, necroptosis, and disrupted retinoid metabolism, providing potential therapeutic targets for hypertensive retinopathy.
Collapse
Affiliation(s)
- Minsup Lee
- Department of Molecular & Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Wendy Leskova
- Department of Molecular & Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Randa S Eshaq
- Department of Molecular & Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Zithlaly Amezquita
- Department of Molecular & Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Norman R Harris
- Department of Molecular & Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA.
| |
Collapse
|
4
|
Abstract
The continuous function of vertebrate photoreceptors requires regeneration of their visual pigment following its destruction upon activation by light (photobleaching). For rods, the chromophore required for the regeneration of rhodopsin is derived from the adjacent retinal pigmented epithelium (RPE) cells through a series of reactions collectively known as the RPE visual cycle. Mounting biochemical and functional evidence demonstrates that, for cones, pigment regeneration is supported by the parallel supply with chromophore by two pathways-the canonical RPE visual cycle and a second, cone-specific retina visual cycle that involves the Müller glial cells in the neural retina. In this article, we review historical information that led to the discovery of the retina visual cycle and discuss what is currently known about the reactions and molecular components of this pathway and its functional role in supporting cone-mediated vision.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Ophthalmology, Gavin Herbert Eye Institute-Center for Translational Vision Research, University of California, Irvine, California, USA; ,
| | - Vladimir J Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute-Center for Translational Vision Research, University of California, Irvine, California, USA; ,
| |
Collapse
|
5
|
Li S, Jin M. Ablation of Fatty Acid Transport Protein-4 Enhances Cone Survival, M-cone Vision, and Synthesis of Cone-Tropic 9- cis-Retinal in rd12 Mouse Model of Leber Congenital Amaurosis. J Neurosci 2024; 44:e1994232024. [PMID: 38811164 PMCID: PMC11223470 DOI: 10.1523/jneurosci.1994-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/03/2023] [Accepted: 05/22/2024] [Indexed: 05/31/2024] Open
Abstract
The canonical visual cycle employing RPE65 as the retinoid isomerase regenerates 11-cis-retinal to support both rod- and cone-mediated vision. Mutations of RPE65 are associated with Leber congenital amaurosis that results in rod and cone photoreceptor degeneration and vision loss of affected patients at an early age. Dark-reared Rpe65-/- mouse has been known to form isorhodopsin that employs 9-cis-retinal as the photosensitive chromophore. The mechanism regulating 9-cis-retinal synthesis and the role of the endogenous 9-cis-retinal in cone survival and function remain largely unknown. In this study, we found that ablation of fatty acid transport protein-4 (FATP4), a negative regulator of 11-cis-retinol synthesis catalyzed by RPE65, increased the formation of 9-cis-retinal, but not 11-cis-retinal, in a light-independent mechanism in both sexes of RPE65-null rd12 mice. Both rd12 and rd12;Fatp4-/- mice contained a massive amount of all-trans-retinyl esters in the eyes, exhibiting comparable scotopic vision and rod degeneration. However, expression levels of M- and S-opsins as well as numbers of M- and S-cones surviving in the superior retinas of rd12;Fatp4-/ - mice were at least twofold greater than those in age-matched rd12 mice. Moreover, FATP4 deficiency significantly shortened photopic b-wave implicit time, improved M-cone visual function, and substantially deaccelerated the progression of cone degeneration in rd12 mice, whereas FATP4 deficiency in mice with wild-type Rpe65 alleles neither induced 9-cis-retinal formation nor influenced cone survival and function. These results identify FATP4 as a new regulator of synthesis of 9-cis-retinal, which is a "cone-tropic" chromophore supporting cone survival and function in the retinas with defective RPE65.
Collapse
Affiliation(s)
- Songhua Li
- Neuroscience Center, Louisiana State University School of Medicine, LSU Health New Orleans, New Orleans, Louisiana 70112
| | - Minghao Jin
- Neuroscience Center, Louisiana State University School of Medicine, LSU Health New Orleans, New Orleans, Louisiana 70112
- Department of Ophthalmology, Louisiana State University School of Medicine, LSU Health New Orleans, New Orleans, Louisiana 70112
| |
Collapse
|
6
|
Getz TE, Chrenek MA, Papania JT, Shelton DA, Markand S, Iuvone PM, Kozmik Z, Boatright JH, Nickerson JM. Conditional Knockouts of Interphotoreceptor Retinoid Binding Protein Suggest Two Independent Mechanisms for Retinal Degeneration and Myopia. Invest Ophthalmol Vis Sci 2024; 65:32. [PMID: 38904640 PMCID: PMC11193143 DOI: 10.1167/iovs.65.6.32] [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: 03/24/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
Purpose Interphotoreceptor retinoid-binding protein's (IRBP) role in eye growth and its involvement in cell homeostasis remain poorly understood. One hypothesis proposes early conditional deletion of the IRBP gene could lead to a myopic response with retinal degeneration, whereas late conditional deletion (after eye size is determined) could cause retinal degeneration without myopia. Here, we sought to understand if prior myopia was required for subsequent retinal degeneration in the absence of IRBP. This study investigates if any cell type or developmental stage is more important in myopia or retinal degeneration. Methods IBRPfl/fl mice were bred with 5 Cre-driver lines: HRGP-Cre, Chx10-Cre, Rho-iCre75, HRGP-Cre Rho-iCre75, and Rx-Cre. Mice were analyzed for IRBP gene expression through digital droplet PCR (ddPCR). Young adult (P30) mice were tested for retinal degeneration and morphology using spectral-domain optical coherence tomography (SD-OCT) and hematoxylin and eosin (H&E) staining. Function was analyzed using electroretinograms (ERGs). Eye sizes and axial lengths were compared through external eye measurements and whole eye biometry. Results Across all outcome measures, when bred to IRBPfl/fl, HRGP-Cre and Chx10-Cre lines showed no differences from IRBPfl/fl alone. With the Rho-iCre75 line, small but significant reductions were seen in retinal thickness with SD-OCT imaging and postmortem H&E staining without increased axial length. Both the HRGP-Cre+Rho-iCre75 and the Rx-Cre lines showed significant decreases in retinal thickness and outer nuclear layer cell counts. Using external eye measurements and SD-OCT imaging, both lines showed an increase in eye size. Finally, function in both lines was roughly halved across scotopic, photopic, and flicker ERGs. Conclusions Our studies support hypotheses that for both eye size determination and retinal homeostasis, there are two critical timing windows when IRBP must be expressed in rods or cones to prevent myopia (P7-P12) and degeneration (P21 and later). The rod-specific IRBP knockout (Rho-iCre75) showed significant retinal functional losses without myopia, indicating that the two phenotypes are independent. IRBP is needed for early development of photoreceptors and eye size, whereas Rho-iCre75 IRBPfl/fl knockout results in retinal degeneration without myopia.
Collapse
Affiliation(s)
- Tatiana E. Getz
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Jack T. Papania
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Debresha A. Shelton
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Shanu Markand
- Kirksville College of Osteopathic Medicine, A.T. Still University, Kirksville, Missouri, United States
| | - P. Michael Iuvone
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Jeffrey H. Boatright
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| |
Collapse
|
7
|
Poli G, Demontis GC, Sodi A, Saba A, Rizzo S, Macchia M, Tuccinardi T. An in silico toolbox for the prediction of the potential pathogenic effects of missense mutations in the dimeric region of hRPE65. J Enzyme Inhib Med Chem 2023; 38:2162047. [PMID: 36629452 PMCID: PMC9848331 DOI: 10.1080/14756366.2022.2162047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
hRPE65 is a fundamental enzyme of the retinoid visual cycle, and many missense mutations affecting its expression or function are associated with a wide range of diseases. Many hRPE65 missense mutations lack a clear pathogenicity classification or are labelled as VUS. In this context, we recently developed a protocol based on µs-long molecular dynamics simulations to study the potential pathogenic effect of hRPE65 missense mutations. In the present work, the structure-based protocol was integrated with a hRPE65-tailored consensus bioinformatics strategy, named ConPath, that showed high performance in predicting known pathogenic/benign hRPE65 missense mutations. The combined strategy was used to perform a multi-level evaluation of the potential pathogenicity of 13 different hRPE65 VUS, which were classified based on their likelihood of pathogenic effect. The obtained results provide information that may support the reclassification of these VUS and help clinicians evaluate the eligibility for gene therapy of patients diagnosed with such variants.
Collapse
Affiliation(s)
- Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Andrea Sodi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, Florence, Italy
| | - Alessandro Saba
- Department of Surgical Pathology, Molecular Medicine and of the Critical Area, University of Pisa, Pisa, Italy
| | - Stanislao Rizzo
- Ophthalmology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy,Catholic University Sacro Cuore, Rome, Italy,Consiglio Nazionale delle Ricerche, Istituto di Neuroscienze, Pisa, Italy
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Pisa, Italy,CONTACT Tiziano Tuccinardi Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, 56126, Italy
| |
Collapse
|
8
|
DeRamus ML, Jasien JV, Eppstein JM, Koala P, Kraft TW. Retinal Responses to Visual Stimuli in Interphotoreceptor Retinoid Binding-Protein Knock-Out Mice. Int J Mol Sci 2023; 24:10655. [PMID: 37445836 PMCID: PMC10341985 DOI: 10.3390/ijms241310655] [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: 04/07/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) is an abundant glycoprotein in the subretinal space bound by the photoreceptor (PR) outer segments and the processes of the retinal pigmented epithelium (RPE). IRBP binds retinoids, including 11-cis-retinal and all-trans-retinol. In this study, visual function for demanding visual tasks was assessed in IRBP knock-out (KO) mice. Surprisingly, IRBP KO mice showed no differences in scotopic critical flicker frequency (CFF) compared to wildtype (WT). However, they did have lower photopic CFF than WT. IRBP KO mice had reduced scotopic and photopic acuity and contrast sensitivity compared to WT. IRBP KO mice had a significant reduction in outer nuclear layer (ONL) thickness, PR outer and inner segment, and full retinal thickness (FRT) compared to WT. There were fewer cones in IRBP KO mice. Overall, these results confirm substantial loss of rods and significant loss of cones within 30 days. Absence of IRBP resulted in cone circuit damage, reducing photopic flicker, contrast sensitivity, and spatial frequency sensitivity. The c-wave was reduced and accelerated in response to bright steps of light. This result also suggests altered retinal pigment epithelium activity. There appears to be a compensatory mechanism such as higher synaptic gain between PRs and bipolar cells since the loss of the b-wave did not linearly follow the loss of rods, or the a-wave. Scotopic CFF is normal despite thinning of ONL and reduced scotopic electroretinogram (ERG) in IRBP KO mice, suggesting either a redundancy or plasticity in circuits detecting (encoding) scotopic flicker at threshold even with substantial rod loss.
Collapse
Affiliation(s)
| | | | | | | | - Timothy W. Kraft
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.V.J.); (J.M.E.); (P.K.)
| |
Collapse
|
9
|
Rivera-Colón AG, Rayamajhi N, Minhas BF, Madrigal G, Bilyk KT, Yoon V, Hüne M, Gregory S, Cheng CHC, Catchen JM. Genomics of Secondarily Temperate Adaptation in the Only Non-Antarctic Icefish. Mol Biol Evol 2023; 40:msad029. [PMID: 36806940 PMCID: PMC9985337 DOI: 10.1093/molbev/msad029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
White-blooded Antarctic icefishes, a family within the adaptive radiation of Antarctic notothenioid fishes, are an example of extreme biological specialization to both the chronic cold of the Southern Ocean and life without hemoglobin. As a result, icefishes display derived physiology that limits them to the cold and highly oxygenated Antarctic waters. Against these constraints, remarkably one species, the pike icefish Champsocephalus esox, successfully colonized temperate South American waters. To study the genetic mechanisms underlying secondarily temperate adaptation in icefishes, we generated chromosome-level genome assemblies of both C. esox and its Antarctic sister species, Champsocephalus gunnari. The C. esox genome is similar in structure and organization to that of its Antarctic congener; however, we observe evidence of chromosomal rearrangements coinciding with regions of elevated genetic divergence in pike icefish populations. We also find several key biological pathways under selection, including genes related to mitochondria and vision, highlighting candidates behind temperate adaptation in C. esox. Substantial antifreeze glycoprotein (AFGP) pseudogenization has occurred in the pike icefish, likely due to relaxed selection following ancestral escape from Antarctica. The canonical AFGP locus organization is conserved in C. esox and C. gunnari, but both show a translocation of two AFGP copies to a separate locus, previously unobserved in cryonotothenioids. Altogether, the study of this secondarily temperate species provides an insight into the mechanisms underlying adaptation to ecologically disparate environments in this otherwise highly specialized group.
Collapse
Affiliation(s)
- Angel G Rivera-Colón
- Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Niraj Rayamajhi
- Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL
| | | | - Giovanni Madrigal
- Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Kevin T Bilyk
- Department of Biology, Montclair State University, Montclair, NJ
| | - Veronica Yoon
- Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Mathias Hüne
- Centro de Investigación para la Conservación de los Ecosistemas Australes, Punta Arenas, Chile
| | - Susan Gregory
- British Antarctic Survey, Cambridge, United Kingdom
- Government of South Georgia and the South Sandwich Islands, Stanley, Falklands
| | - C H Christina Cheng
- Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Julian M Catchen
- Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
10
|
Towards a New Biomarker for Diabetic Retinopathy: Exploring RBP3 Structure and Retinoids Binding for Functional Imaging of Eyes In Vivo. Int J Mol Sci 2023; 24:ijms24054408. [PMID: 36901838 PMCID: PMC10002987 DOI: 10.3390/ijms24054408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Diabetic retinopathy (DR) is a severe disease with a growing number of afflicted patients, which places a heavy burden on society, both socially and financially. While there are treatments available, they are not always effective and are usually administered when the disease is already at a developed stage with visible clinical manifestation. However, homeostasis at a molecular level is disrupted before visible signs of the disease are evident. Thus, there has been a constant search for effective biomarkers that could signal the onset of DR. There is evidence that early detection and prompt disease control are effective in preventing or slowing DR progression. Here, we review some of the molecular changes that occur before clinical manifestations are observable. As a possible new biomarker, we focus on retinol binding protein 3 (RBP3). We argue that it displays unique features that make it a very good biomarker for non-invasive, early-stage DR detection. Linking chemistry to biological function and focusing on new developments in eye imaging and two-photon technology, we describe a new potential diagnostic tool that would allow rapid and effective quantification of RBP3 in the retina. Moreover, this tool would also be useful in the future to monitor therapeutic effectiveness if levels of RBP3 are elevated by DR treatments.
Collapse
|
11
|
Ciliary neurotrophic factor-mediated neuroprotection involves enhanced glycolysis and anabolism in degenerating mouse retinas. Nat Commun 2022; 13:7037. [PMID: 36396639 PMCID: PMC9672129 DOI: 10.1038/s41467-022-34443-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective cytokine in multiple models of retinal degeneration. To understand mechanisms underlying its broad neuroprotective effects, we have investigated the influence of CNTF on metabolism in a mouse model of photoreceptor degeneration. CNTF treatment improves the morphology of photoreceptor mitochondria, but also leads to reduced oxygen consumption and suppressed respiratory chain activities. Molecular analyses show elevated glycolytic pathway gene transcripts and active enzymes. Metabolomics analyses detect significantly higher levels of ATP and the energy currency phosphocreatine, elevated glycolytic pathway metabolites, increased TCA cycle metabolites, lipid biosynthetic pathway intermediates, nucleotides, and amino acids. Moreover, CNTF treatment restores the key antioxidant glutathione to the wild type level. Therefore, CNTF significantly impacts the metabolic status of degenerating retinas by promoting aerobic glycolysis and augmenting anabolic activities. These findings reveal cellular mechanisms underlying enhanced neuronal viability and suggest potential therapies for treating retinal degeneration.
Collapse
|
12
|
Gabrielle PH. Lipid metabolism and retinal diseases. Acta Ophthalmol 2022; 100 Suppl 269:3-43. [PMID: 36117363 DOI: 10.1111/aos.15226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/24/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE The retina has enormous lipids demands and must meet those needs. Retinal lipid homeostasis appears to be based on the symbiosis between neurons, Müller glial cells (MGC), and retinal pigment epithelium (RPE) cells, which can be impacted in several retinal diseases. The current research challenge is to better understand lipid-related mechanisms involved in retinal diseases, such as age-related macular degeneration (AMD) and glaucoma. RESULTS In a first axis, in vitro and focus on Müller glial cell, we aimed to characterize whether the 24S-hydroxycholesterol (24S-OHC), an overexpressed end-product of cholesterol elimination pathway in neural tissue and likely produced by suffering retinal ganglion cells in glaucoma, may modulate MGC membrane organization, such as lipid rafts, to trigger cellular signalling pathways related to retinal gliosis. We have found that lipid composition appears to be a key factor of membrane architecture, especially for lipid raft microdomain formation, in MGC. However, 24S-OHC did not appear to trigger retinal gliosis via the modulation of lipid or protein composition within lipid rafts microdomains. This study provided a better understanding of the complex mechanisms involved in the pathophysiology of glaucoma. On a second clinical ax, we focused on the lipid-related mechanisms involved in the dysfunction of aging RPE and the appearance of drusenoid deposits in AMD. Using the Montrachet population-based study, we intend to report the frequency of reticular pseudodrusen (RPD) and its ocular and systemic risk factors, particularly related to lipid metabolisms, such as plasma lipoprotein levels, carotenoids levels, and lipid-lowering drug intake. Our study showed that RPD was less common in subjects taking lipid-lowering drugs. Lipid-lowering drugs, such as statins, may reduce the risk of RPD through their effect on the production and function of lipoproteins. This observation highlights the potential role of retinal lipid trafficking via lipoproteins between photoreceptors and retinal pigment epithelium cells in RPD formation. Those findings have been complemented with preliminary results on the analysis of plasma fatty acid (FA) profile, a surrogate marker of short-term dietary lipid intake, according to the type of predominant drusenoid deposit, soft drusen or RPD, in age-related maculopathy. CONCLUSION Further research on lipid metabolism in retinal diseases is warranted to better understand the pathophysiology of retinal diseases and develop new promising diagnostic, prognostic, and therapeutic tools for our patients.
Collapse
Affiliation(s)
- Pierre-Henry Gabrielle
- Eye and Nutrition Research Group, Center for Taste and Feeding Behaviour, AgroSup Dijon, CNRS, INRAe, The University Bourgogne Franche-Comté, Dijon, France.,Department of Ophthalmology, Dijon University Hospital, Dijon, France.,The Save Sight Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Widjaja-Adhi MAK, Kolesnikov AV, Vasudevan S, Park PSH, Kefalov VJ, Golczak M. Acyl-CoA:wax alcohol acyltransferase 2 modulates the cone visual cycle in mouse retina. FASEB J 2022; 36:e22390. [PMID: 35665537 DOI: 10.1096/fj.202101855rrr] [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: 12/01/2021] [Revised: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 11/11/2022]
Abstract
The daylight and color vision of diurnal vertebrates depends on cone photoreceptors. The capability of cones to operate and respond to changes in light brightness even under high illumination is attributed to their fast rate of recovery to the ground photosensitive state. This process requires the rapid replenishing of photoisomerized visual chromophore (11-cis-retinal) to regenerate cone visual pigments. Recently, several gene candidates have been proposed to contribute to the cone-specific retinoid metabolism, including acyl-CoA wax alcohol acyltransferase 2 (AWAT2, aka MFAT). Here, we evaluated the role of AWAT2 in the regeneration of visual chromophore by the phenotypic characterization of Awat2-/- mice. The global absence of AWAT2 enzymatic activity did not affect gross retinal morphology or the rate of visual chromophore regeneration by the canonical RPE65-dependent visual cycle. Analysis of Awat2 expression indicated the presence of the enzyme throughout the murine retina, including the retinal pigment epithelium (RPE) and Müller cells. Electrophysiological recordings revealed reduced maximal rod and cone dark-adapted responses in AWAT2-deficient mice compared to control mice. While rod dark adaptation was not affected by the lack of AWAT2, M-cone dark adaptation both in isolated retina and in vivo was significantly suppressed. Altogether, these results indicate that while AWAT2 is not required for the normal operation of the canonical visual cycle, it is a functional component of the cone-specific visual chromophore regenerative pathway.
Collapse
Affiliation(s)
| | - Alexander V Kolesnikov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California, USA
| | - Sreelakshmi Vasudevan
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Vladimir J Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California, USA.,Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA.,Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
14
|
Wang J, Tao Z, Deng H, Cui Y, Xu Z, Lyu Q, Zhao J. Therapeutic implications of nanodrug and tissue engineering for retinal pigment epithelium-related diseases. NANOSCALE 2022; 14:5657-5677. [PMID: 35352082 DOI: 10.1039/d1nr08337f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The retinal pigment epithelium (RPE), as a single layer of cells that performs multiple functions posteriorly in the eye, is a promising target site for the prevention and treatment of several clinical diseases, including proliferative diabetic retinopathy, age-related macular degeneration, chorionic neovascularization, and retinitis pigmentosa. In recent decades, several nanodrug delivery platforms and tissue-engineered RPE have been widely developed to treat RPE-related diseases. This work summarizes the recent advances in nanoplatforms and tissue engineering scaffolds developed in these fields. The diseases associated with pathological RPE and their common therapy strategies are first introduced. Then, the recent progress made with a variety of drug delivery systems is presented, with an emphasis on the modification strategies of nanomaterials for targeted delivery. Tissue engineering-mediated RPE transplantation for treating these diseases is subsequently described. Finally, the clinical translation challenges in these fields are discussed in depth. This article will offer readers a better understanding of emerging nanotechnology and tissue engineering related to the treatment of RPE-related diseases and could facilitate their widespread use in experiments in vivo and in clinical applications.
Collapse
Affiliation(s)
- Jiao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
| | - Zhengyang Tao
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
| | - Hongwei Deng
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
| | - Yubo Cui
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| | - Zhirong Xu
- Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Qinghua Lyu
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen Eye Hospital affiliated to Jinan University, School of Optometry, Shenzhen University, Shenzhen 518000, China.
- Institute of Microscale Optoelectronics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Zhao
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
| |
Collapse
|
15
|
Mano F, Sugioka K, Kuniyoshi K, Kondo H, Kusaka S. Identification of Interphotoreceptor retinoid-binding protein in the Schisis cavity fluid of a patient with congenital X-linked Retinoschisis. BMC Ophthalmol 2022; 22:14. [PMID: 34991515 PMCID: PMC8740355 DOI: 10.1186/s12886-021-02234-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022] Open
Abstract
Background This case report describes the surgical outcome in a patient with congenital X-linked retinoschisis (CXLRS) and the results of proteomic analysis of surgically extracted samples from both vitreous and intraschisis cavities by mass spectrometry. Case presentation A 3-month-old boy presented with extensive retinoschisis involving macula and retinal periphery in both eyes. Genetic analysis confirmed retinoschisin 1 mutation (c.554C > T), and an electroretinogram showed significant reduction of b-wave and decreased cone and rod responses, which led to a diagnosis of CXLRS. By performing pars plana vitrectomy, including inner wall retinectomy, clear visual axes with stable retinal conditions and functional vision in both eyes were obtained during the 4 years of follow-up. Proteomic analysis of surgically retrieved fluid from the intraschisis cavity revealed a higher expression of interphotoreceptor retinoid-binding protein (IRBP) than that from the vitreous humor. However, both samples showed equal levels of albumin, transferrin, and pigment epithelium-derived factor. Conclusions Cellular adhesive imperfection in CXLRS may cause IRBP diffusion from the interphotoreceptor matrix, resulting in the strong expression of IRBP in the intraschisis cavity. An impaired retinoid cycle caused by an absence of IRBP in the retina may potentially underlie the pathology of CXLRS. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-021-02234-5.
Collapse
Affiliation(s)
- Fukutaro Mano
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Koji Sugioka
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan. .,Department of Ophthalmology, Kindai University Nara Hospital, 1248-1 Otodacho, Ikoma City, Nara, 630-0293, Japan.
| | - Kazuki Kuniyoshi
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Hiroyuki Kondo
- Department of Ophthalmology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shunji Kusaka
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
16
|
Shome A, Mugisho OO, Niederer RL, Rupenthal ID. Blocking the inflammasome: A novel approach to treat uveitis. Drug Discov Today 2021; 26:2839-2857. [PMID: 34229084 DOI: 10.1016/j.drudis.2021.06.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022]
Abstract
Uveitis is a complex ocular inflammatory disease often accompanied by bacterial or viral infections (infectious uveitis) or underlying autoimmune diseases (non-infectious uveitis). Treatment of the underlying infection along with corticosteroid-mediated suppression of acute inflammation usually resolves infectious uveitis. However, to develop more effective therapies for non-infectious uveitis and to better address acute inflammation in infectious disease, an improved understanding of the underlying inflammatory pathways is needed. In this review, we discuss the disease aetiology, preclinical in vitro and in vivo uveitis models, the role of inflammatory pathways, as well as current and future therapies. In particular, we highlight the involvement of the inflammasome in the development of non-infectious uveitis and how it could be a future target for effective treatment of the disease.
Collapse
Affiliation(s)
- Avik Shome
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Rachael L Niederer
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand; Auckland District Health Board, Auckland, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
17
|
Tonade D, Kern TS. Photoreceptor cells and RPE contribute to the development of diabetic retinopathy. Prog Retin Eye Res 2021; 83:100919. [PMID: 33188897 PMCID: PMC8113320 DOI: 10.1016/j.preteyeres.2020.100919] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/27/2020] [Accepted: 10/31/2020] [Indexed: 12/26/2022]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness. It has long been regarded as vascular disease, but work in the past years has shown abnormalities also in the neural retina. Unfortunately, research on the vascular and neural abnormalities have remained largely separate, instead of being integrated into a comprehensive view of DR that includes both the neural and vascular components. Recent evidence suggests that the most predominant neural cell in the retina (photoreceptors) and the adjacent retinal pigment epithelium (RPE) play an important role in the development of vascular lesions characteristic of DR. This review summarizes evidence that the outer retina is altered in diabetes, and that photoreceptors and RPE contribute to retinal vascular alterations in the early stages of the retinopathy. The possible molecular mechanisms by which cells of the outer retina might contribute to retinal vascular damage in diabetes also are discussed. Diabetes-induced alterations in the outer retina represent a novel therapeutic target to inhibit DR.
Collapse
Affiliation(s)
- Deoye Tonade
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy S Kern
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA; Veterans Administration Medical Center Research Service, Cleveland, OH, USA; Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA; Veterans Administration Medical Center Research Service, Long Beach, CA, USA.
| |
Collapse
|
18
|
Chen J, Shao Y, Sasore T, Moiseyev G, Zhou K, Ma X, Du Y, Ma JX. Interphotoreceptor Retinol-Binding Protein Ameliorates Diabetes-Induced Retinal Dysfunction and Neurodegeneration Through Rhodopsin. Diabetes 2021; 70:788-799. [PMID: 33334874 PMCID: PMC7897347 DOI: 10.2337/db20-0609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
Patients with diabetes often experience visual defects before any retinal pathologies are detected. The molecular mechanism for the visual defects in early diabetes has not been elucidated. Our previous study reported that in early diabetic retinopathy (DR), rhodopsin levels were reduced due to impaired 11-cis-retinal regeneration. Interphotoreceptor retinol-binding protein (IRBP) is a visual cycle protein and important for 11-cis-retinal generation. IRBP levels are decreased in the vitreous and retina of DR patients and animal models. To determine the role of IRBP downregulation in the visual defects in early DR, we induced diabetes in transgenic mice overexpressing IRBP in the retina. IRBP overexpression prevented diabetes-induced decline of retinal function. Furthermore, IRBP overexpression also prevented decreases of rhodopsin levels and 11-cis-retinal generation in diabetic mice. Diabetic IRBP transgenic mice also showed ameliorated retinal oxidative stress, inflammation, apoptosis, and retinal degeneration compared with diabetic wild-type mice. These findings suggest that diabetes-induced IRBP downregulation impairs the regeneration of 11-cis-retinal and rhodopsin, leading to retinal dysfunction in early DR. Furthermore, increased 11-cis-retinal-free opsin constitutively activates the phototransduction pathway, leading to increased oxidative stress and retinal neurodegeneration. Therefore, restored IRBP expression in the diabetic retina may confer a protective effect against retinal degeneration in DR.
Collapse
Affiliation(s)
- Jianglei Chen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Yan Shao
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Temmy Sasore
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Gennadiy Moiseyev
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Kelu Zhou
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Xiang Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Yanhong Du
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
19
|
Kiser PD, Palczewski K. Pathways and disease-causing alterations in visual chromophore production for vertebrate vision. J Biol Chem 2021; 296:100072. [PMID: 33187985 PMCID: PMC7948990 DOI: 10.1074/jbc.rev120.014405] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
All that we view of the world begins with an ultrafast cis to trans photoisomerization of the retinylidene chromophore associated with the visual pigments of rod and cone photoreceptors. The continual responsiveness of these photoreceptors is then sustained by regeneration processes that convert the trans-retinoid back to an 11-cis configuration. Recent biochemical and electrophysiological analyses of the retinal G-protein-coupled receptor (RGR) suggest that it could sustain the responsiveness of photoreceptor cells, particularly cones, even under bright light conditions. Thus, two mechanisms have evolved to accomplish the reisomerization: one involving the well-studied retinoid isomerase (RPE65) and a second photoisomerase reaction mediated by the RGR. Impairments to the pathways that transform all-trans-retinal back to 11-cis-retinal are associated with mild to severe forms of retinal dystrophy. Moreover, with age there also is a decline in the rate of chromophore regeneration. Both pharmacological and genetic approaches are being used to bypass visual cycle defects and consequently mitigate blinding diseases. Rapid progress in the use of genome editing also is paving the way for the treatment of disparate retinal diseases. In this review, we provide an update on visual cycle biochemistry and then discuss visual-cycle-related diseases and emerging therapeutics for these disorders. There is hope that these advances will be helpful in treating more complex diseases of the eye, including age-related macular degeneration (AMD).
Collapse
Affiliation(s)
- Philip D Kiser
- The Department of Physiology & Biophysics, University of California, Irvine, California, USA; Research Service, The VA Long Beach Health Care System, Long Beach, California, USA; The Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California, USA.
| | - Krzysztof Palczewski
- The Department of Physiology & Biophysics, University of California, Irvine, California, USA; The Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California, USA; The Department of Chemistry, University of California, Irvine, California, USA.
| |
Collapse
|
20
|
Zeng S, Zhang T, Madigan MC, Fernando N, Aggio-Bruce R, Zhou F, Pierce M, Chen Y, Huang L, Natoli R, Gillies MC, Zhu L. Interphotoreceptor Retinoid-Binding Protein (IRBP) in Retinal Health and Disease. Front Cell Neurosci 2020; 14:577935. [PMID: 33328889 PMCID: PMC7710524 DOI: 10.3389/fncel.2020.577935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/21/2020] [Indexed: 02/05/2023] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP), also known as retinol binding protein 3 (RBP3), is a lipophilic glycoprotein specifically secreted by photoreceptors. Enriched in the interphotoreceptor matrix (IPM) and recycled by the retinal pigment epithelium (RPE), IRBP is essential for the vision of all vertebrates as it facilitates the transfer of retinoids in the visual cycle. It also helps to transport lipids between the RPE and photoreceptors. The thiol-dependent antioxidant activity of IRBP maintains the delicate redox balance in the normal retina. Thus, its dysfunction is suspected to play a role in many retinal diseases. We have reviewed here the latest research on IRBP in both retinal health and disease, including the function and regulation of IRBP under retinal stress in both animal models and the human retina. We have also explored the therapeutic potential of targeting IRBP in retinal diseases. Although some technical barriers remain, it is possible that manipulating the expression of IRBP in the retina will rescue or prevent photoreceptor degeneration in many retinal diseases.
Collapse
Affiliation(s)
- Shaoxue Zeng
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Zhang
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Michele C Madigan
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,School of Optometry and Vision Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia.,The Australian National University Medical School, The Australian National University, Acton, ACT, Australia
| | - Fanfan Zhou
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
| | - Matthew Pierce
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Yingying Chen
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Lianlin Huang
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,School of Optometry and Vision Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia.,The Australian National University Medical School, The Australian National University, Acton, ACT, Australia
| | - Mark C Gillies
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Ling Zhu
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
21
|
Inverse correlation between fatty acid transport protein 4 and vision in Leber congenital amaurosis associated with RPE65 mutation. Proc Natl Acad Sci U S A 2020; 117:32114-32123. [PMID: 33257550 DOI: 10.1073/pnas.2012623117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Fatty acid transport protein 4 (FATP4), a transmembrane protein in the endoplasmic reticulum (ER), is a recently identified negative regulator of the ER-associated retinal pigment epithelium (RPE)65 isomerase necessary for recycling 11-cis-retinal, the light-sensitive chromophore of both rod and cone opsin visual pigments. The role of FATP4 in the disease progression of retinal dystrophies associated with RPE65 mutations is completely unknown. Here we show that FATP4-deficiency in the RPE results in 2.8-fold and 1.7-fold increase of 11-cis- and 9-cis-retinals, respectively, improving dark-adaptation rates as well as survival and function of rods in the Rpe65 R91W knockin (KI) mouse model of Leber congenital amaurosis (LCA). Degradation of S-opsin in the proteasomes, but not in the lysosomes, was remarkably reduced in the KI mouse retinas lacking FATP4. FATP4-deficiency also significantly rescued S-opsin trafficking and M-opsin solubility in the KI retinas. The number of S-cones in the inferior retinas of 4- or 6-mo-old KI;Fatp4 -/- mice was 7.6- or 13.5-fold greater than those in age-matched KI mice. Degeneration rates of S- and M-cones are negatively correlated with expression levels of FATP4 in the RPE of the KI, KI;Fatp4 +/- , and KI;Fatp4 -/- mice. Moreover, the visual function of S- and M-cones is markedly preserved in the KI;Fatp4 -/- mice, displaying an inverse correlation with the FATP4 expression levels in the RPE of the three mutant lines. These findings establish FATP4 as a promising therapeutic target to improve the visual cycle, as well as survival and function of cones and rods in patients with RPE65 mutations.
Collapse
|
22
|
Singh RK, Nasonkin IO. Limitations and Promise of Retinal Tissue From Human Pluripotent Stem Cells for Developing Therapies of Blindness. Front Cell Neurosci 2020; 14:179. [PMID: 33132839 PMCID: PMC7513806 DOI: 10.3389/fncel.2020.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
The self-formation of retinal tissue from pluripotent stem cells generated a tremendous promise for developing new therapies of retinal degenerative diseases, which previously seemed unattainable. Together with use of induced pluripotent stem cells or/and CRISPR-based recombineering the retinal organoid technology provided an avenue for developing models of human retinal degenerative diseases "in a dish" for studying the pathology, delineating the mechanisms and also establishing a platform for large-scale drug screening. At the same time, retinal organoids, highly resembling developing human fetal retinal tissue, are viewed as source of multipotential retinal progenitors, young photoreceptors and just the whole retinal tissue, which may be transplanted into the subretinal space with a goal of replacing patient's degenerated retina with a new retinal "patch." Both approaches (transplantation and modeling/drug screening) were projected when Yoshiki Sasai demonstrated the feasibility of deriving mammalian retinal tissue from pluripotent stem cells, and generated a lot of excitement. With further work and testing of both approaches in vitro and in vivo, a major implicit limitation has become apparent pretty quickly: the absence of the uniform layer of Retinal Pigment Epithelium (RPE) cells, which is normally present in mammalian retina, surrounds photoreceptor layer and develops and matures first. The RPE layer polarize into apical and basal sides during development and establish microvilli on the apical side, interacting with photoreceptors, nurturing photoreceptor outer segments and participating in the visual cycle by recycling 11-trans retinal (bleached pigment) back to 11-cis retinal. Retinal organoids, however, either do not have RPE layer or carry patches of RPE mostly on one side, thus directly exposing most photoreceptors in the developing organoids to neural medium. Recreation of the critical retinal niche between the apical RPE and photoreceptors, where many retinal disease mechanisms originate, is so far unattainable, imposes clear limitations on both modeling/drug screening and transplantation approaches and is a focus of investigation in many labs. Here we dissect different retinal degenerative diseases and analyze how and where retinal organoid technology can contribute the most to developing therapies even with a current limitation and absence of long and functional outer segments, supported by RPE.
Collapse
|
23
|
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
|
24
|
Yokomizo H, Maeda Y, Park K, Clermont AC, Hernandez SL, Fickweiler W, Li Q, Wang CH, Paniagua SM, Simao F, Ishikado A, Sun B, Wu IH, Katagiri S, Pober DM, Tinsley LJ, Avery RL, Feener EP, Kern TS, Keenan HA, Aiello LP, Sun JK, King GL. Retinol binding protein 3 is increased in the retina of patients with diabetes resistant to diabetic retinopathy. Sci Transl Med 2020; 11:11/499/eaau6627. [PMID: 31270273 DOI: 10.1126/scitranslmed.aau6627] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/18/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
Abstract
The Joslin Medalist Study characterized people affected with type 1 diabetes for 50 years or longer. More than 35% of these individuals exhibit no to mild diabetic retinopathy (DR), independent of glycemic control, suggesting the presence of endogenous protective factors against DR in a subpopulation of patients. Proteomic analysis of retina and vitreous identified retinol binding protein 3 (RBP3), a retinol transport protein secreted mainly by the photoreceptors, as elevated in Medalist patients protected from advanced DR. Mass spectrometry and protein expression analysis identified an inverse association between vitreous RBP3 concentration and DR severity. Intravitreal injection and photoreceptor-specific overexpression of RBP3 in rodents inhibited the detrimental effects of vascular endothelial growth factor (VEGF). Mechanistically, our results showed that recombinant RBP3 exerted the therapeutic effects by binding and inhibiting VEGF receptor tyrosine phosphorylation. In addition, by binding to glucose transporter 1 (GLUT1) and decreasing glucose uptake, RBP3 blocked the detrimental effects of hyperglycemia in inducing inflammatory cytokines in retinal endothelial and Müller cells. Elevated expression of photoreceptor-secreted RBP3 may have a role in protection against the progression of DR due to hyperglycemia by inhibiting glucose uptake via GLUT1 and decreasing the expression of inflammatory cytokines and VEGF.
Collapse
Affiliation(s)
- Hisashi Yokomizo
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Yasutaka Maeda
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Kyoungmin Park
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Allen C Clermont
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA
| | | | - Ward Fickweiler
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Qian Li
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Chih-Hao Wang
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | | | - Fabricio Simao
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Atsushi Ishikado
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Bei Sun
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - I-Hsien Wu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Sayaka Katagiri
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - David M Pober
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Liane J Tinsley
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Robert L Avery
- California Retina Consultants, Santa Barbara, CA 93103, USA
| | - Edward P Feener
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy S Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, Irvine, CA 92697, USA
| | - Hillary A Keenan
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Lloyd Paul Aiello
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Jennifer K Sun
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - George L King
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA. .,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
25
|
Yu FJ, Lam TC, Sze AYH, Li KK, Chun RKM, Shan SW, To CH. Alteration of retinal metabolism and oxidative stress may implicate myopic eye growth: Evidence from discovery and targeted proteomics in an animal model. J Proteomics 2020; 221:103684. [PMID: 32061809 DOI: 10.1016/j.jprot.2020.103684] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/01/2020] [Accepted: 02/08/2020] [Indexed: 12/15/2022]
Abstract
Myopia, the most common cause of impaired vision, may induce sight- threatening diseases or ocular complications due to axial elongation. The exact mechanisms underlying myopia development have received much attention and understanding of these is necessary for clinical prevention or therapeutics. In this study, quantitative proteomics using Isotope Coded Protein Label (ICPL) was applied to identify differentially regulated proteins in the retinas of myopic chicks and, from their presence, infer the possible pathogenesis of excessive ocular elongation. Newly hatched white leghorn chicks (n = 15) wore -10D and + 10D lenses bilaterally for 3 and 7 days, respectively, to develop progressive lens-induced myopia (LIM) and hyperopia (LIH). Retinal proteins were quantified with nano-liquid chromatography electrospray ionization coupled with tandem mass spectrometry (nanoLC-ESI-MS/MS). Bioinformatics analysis of differentially regulated proteins revealed that the majority originated from the cytoplasmic region and were related to various metabolic, glycolytic, or oxidative processes. The fold changes of four proteins of interest (vimentin, apolipoprotein A1, interphotoreceptor retinoid binding protein, and glutathione S-transferase) were further confirmed by a novel high-resolution multiple reaction monitoring mass spectrometry (MRM-HR) using a label-free approach. SIGNIFICANCE: Discovery of effective protein biomarkers of myopia has been extensively studied to inhibit myopia progression. This study first applied lens-induced hyperopia and myopia in the same chick to maximize the inter-ocular differences, aiming to discover more protein biomarker candidates. The findings provided new evidence that myopia was metabolism related, accompanied by altered energy generation and oxidative stress at retinal protein levels. The results in the retina were also compared to our previous study in vitreous using ICPL quantitative technology. We have now presented the protein changes in these two adjacent tissues, which may provide extra information of protein changes during ocular growth in myopia.
Collapse
Affiliation(s)
- Feng-Juan Yu
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Thomas Chuen Lam
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| | - Andes Ying-Hon Sze
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - King-Kit Li
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Rachel Ka-Man Chun
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Sze-Wan Shan
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chi-Ho To
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
26
|
Li S, Green JF, Jin M. Impacts of deletion and ichthyosis prematurity syndrome-associated mutations in fatty acid transport protein 4 on the function of RPE65. FEBS Lett 2019; 594:540-552. [PMID: 31595490 DOI: 10.1002/1873-3468.13633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022]
Abstract
The retinal pigment epithelium-specific 65 kDa (RPE65) isomerase plays a pivotal role in photoreceptor survival and function. RPE65-catalyzed synthesis of 11-cis-retinol from all-trans-retinyl esters in the visual cycle is negatively regulated, through a heretofore unknown mechanism, by the fatty acid transport protein FATP4, mutations in which are associated with ichthyosis prematurity syndrome (IPS). Here, we analyzed the interaction between deletion mutants of FATP4 and RPE65 and the impacts of IPS-associated FATP4 mutations on RPE65 expression, 11-cis-retinol synthesis, and all-trans-retinyl ester synthesis. Our results suggest that the interaction between FATP4 and RPE65 contributes to the inhibition of RPE65 function and that IPS-associated nonsense and missense mutations in FATP4 have different effects on the visual cycle.
Collapse
Affiliation(s)
- Songhua Li
- Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - John F Green
- Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Minghao Jin
- Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA, USA.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
27
|
Zhong Z, Rong F, Dai Y, Yibulayin A, Zeng L, Liao J, Wang L, Huang Z, Zhou Z, Chen J. Seven novel variants expand the spectrum of RPE65-related Leber congenital amaurosis in the Chinese population. Mol Vis 2019; 25:204-214. [PMID: 30996589 PMCID: PMC6441358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 03/16/2019] [Indexed: 11/04/2022] Open
Abstract
Purpose To screen RPE65 in 187 families with Leber congenital amaurosis (LCA). Methods Sanger sequencing and/or targeted exome sequencing was employed to identify mutations in the RPE65 gene, and intrafamilial cosegregation analysis if DNA was available. In silico analyses and splicing assay were used to evaluate the variants' pathogenicity. Results Genetic analysis revealed 15 mutations in RPE65 in 14 pedigrees, including one splice-site mutation, one frameshift mutation, three nonsense mutations, and ten missense mutations. Of the mutations identified in RPE65, seven are novel associated with LCA, including five missense variants (c.124C>T, c.149T>C, c.340A>C, c.425A>G, and c.1399C>G) and two indel (insertions or deletions) variants (c.858+1delG and c.1181_1182insT). In vitro splicing assay was performed to evaluate the functional impact on RNA splicing of novel mutations if two of three in silico analyses were predicated to be non-pathogenic at the protein level. Among these 15 variants, 14 were classified as 'pathogenic variants,' and a variant (c.124C>T) was 'variants with uncertain significance' according to the standards and guidelines of the American College of Medical Genetics and Genomics. Conclusions Mutations in RPE65 were responsible for 11 of the cohort of 187 Chinese families with LCA, which expands the spectrum of RPE65-related LCA in the Chinese population and potentially facilitates its clinical implementation.
Collapse
Affiliation(s)
- Zilin Zhong
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China,Department of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Feng Rong
- Kizilsu Kirgiz Autonomous Prefecture People's Hospital, Atushi, Xinjiang, China
| | - Yinghui Dai
- Department of Ophthalmology, the First Affiliated Hospital of Benbu medical college, Benbu, Anhui, China
| | - Alakezi Yibulayin
- Kizilsu Kirgiz Autonomous Prefecture People's Hospital, Atushi, Xinjiang, China
| | - Lin Zeng
- Kizilsu Kirgiz Autonomous Prefecture People's Hospital, Atushi, Xinjiang, China
| | - Jian Liao
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China,Department of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Liefeng Wang
- Department of Biotechnology, Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Zhihua Huang
- School of Basic Medical Sciences, Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Zhenping Zhou
- Kizilsu Kirgiz Autonomous Prefecture People's Hospital, Atushi, Xinjiang, China
| | - Jianjun Chen
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China,Department of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Silvaroli JA, Widjaja-Adhi MAK, Trischman T, Chelstowska S, Horwitz S, Banerjee S, Kiser PD, Blaner WS, Golczak M. Abnormal Cannabidiol Modulates Vitamin A Metabolism by Acting as a Competitive Inhibitor of CRBP1. ACS Chem Biol 2019; 14:434-448. [PMID: 30721022 DOI: 10.1021/acschembio.8b01070] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cellular retinol-binding proteins (CRBPs) facilitate the uptake and intracellular transport of vitamin A. They integrate retinoid metabolism, playing an important role in regulating the synthesis of bioactive vitamin A metabolites. Thus, CRBPs constitute potential pharmacological targets to modulate cellular retinoid status that in turn may have applications in the treatment of certain immunological, metabolic, and ocular disorders. Here we identify abnormal cannabidiol (abn-CBD) as a nonretinoid inhibitor of cellular retinol-binding protein 1 (CRBP1). X-ray crystal structures of CRBP1 in complex with abn-CBD and its derivatives revealed a distinctive mode of protein-ligand interaction and provided a molecular basis for the high affinity and selectivity of this compound. We demonstrated that abn-CBD modulates the flux of retinoids via the retinoid cycle in vivo. Furthermore, the biological activity of abn-CBD was evidenced by its ability to protect against light-induced retinal damage in Balb/cJ mice. Altogether, our findings indicate that targeting selected CRBPs with a small-molecule inhibitor can potentially lead to the development of new therapeutic agents to counteract diseases with etiologies involving imbalance in retinoid metabolism or signaling.
Collapse
Affiliation(s)
| | | | | | | | | | - Surajit Banerjee
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, United States
- Northeastern Collaborative Access Team, Argonne National Laboratory, Argonne, IL, United States
| | - Philip D. Kiser
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, United States
| | - William S. Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | | |
Collapse
|
29
|
Sluch VM, Banks A, Li H, Crowley MA, Davis V, Xiang C, Yang J, Demirs JT, Vrouvlianis J, Leehy B, Hanks S, Hyman AM, Aranda J, Chang B, Bigelow CE, Rice DS. ADIPOR1 is essential for vision and its RPE expression is lost in the Mfrp rd6 mouse. Sci Rep 2018; 8:14339. [PMID: 30254279 PMCID: PMC6156493 DOI: 10.1038/s41598-018-32579-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/10/2018] [Indexed: 12/15/2022] Open
Abstract
The knockout (KO) of the adiponectin receptor 1 (AdipoR1) gene causes retinal degeneration. Here we report that ADIPOR1 protein is primarily found in the eye and brain with little expression in other tissues. Further analysis of AdipoR1 KO mice revealed that these animals exhibit early visual system abnormalities and are depleted of RHODOPSIN prior to pronounced photoreceptor death. A KO of AdipoR1 post-development either in photoreceptors or the retinal pigment epithelium (RPE) resulted in decreased expression of retinal proteins, establishing a role for ADIPOR1 in supporting vision in adulthood. Subsequent analysis of the Mfrprd6 mouse retina demonstrated that these mice are lacking ADIPOR1 in their RPE layer alone, suggesting that loss of ADIPOR1 drives retinal degeneration in this model. Moreover, we found elevated levels of IRBP in both the AdipoR1 KO and the Mfrprd6 models. The spatial distribution of IRBP was also abnormal. This dysregulation of IRBP hypothesizes a role for ADIPOR1 in retinoid metabolism.
Collapse
Affiliation(s)
- Valentin M Sluch
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States.
| | - Angela Banks
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Hui Li
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Maura A Crowley
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Vanessa Davis
- Global Scientific Operations, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Chuanxi Xiang
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Junzheng Yang
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - John T Demirs
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Joanna Vrouvlianis
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Barrett Leehy
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Shawn Hanks
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Alexandra M Hyman
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Jorge Aranda
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | - Chad E Bigelow
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Dennis S Rice
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States.
| |
Collapse
|
30
|
Li S, Sato K, Gordon WC, Sendtner M, Bazan NG, Jin M. Ciliary neurotrophic factor (CNTF) protects retinal cone and rod photoreceptors by suppressing excessive formation of the visual pigments. J Biol Chem 2018; 293:15256-15268. [PMID: 30115683 DOI: 10.1074/jbc.ra118.004008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
The retinal pigment epithelium (RPE)-dependent visual cycle provides 11-cis-retinal to opsins in the photoreceptor outer segments to generate functional visual pigments that initiate phototransduction in response to light stimuli. Both RPE65 isomerase of the visual cycle and the rhodopsin visual pigment have recently been identified as critical players in mediating light-induced retinal degeneration. These findings suggest that the expression and function of RPE65 and rhodopsin need to be coordinately controlled to sustain normal vision and to protect the retina from photodamage. However, the mechanism controlling the development of the retinal visual system remains poorly understood. Here, we show that deficiency in ciliary neurotrophic factor (CNTF) up-regulates the levels of rod and cone opsins accompanied by an increase in the thickness of the outer nuclear layers and the lengths of cone and rod outer segments in the mouse retina. Moreover, retinoid isomerase activity, expression levels of RPE65 and lecithin:retinol acyltransferase (LRAT), which synthesizes the RPE65 substrate, were also significantly increased in the Cntf -/- RPE. Rod a-wave and cone b-wave amplitudes of electroretinograms were increased in Cntf -/- mice, but rod b-wave amplitudes were unchanged compared with those in WT mice. Up-regulated RPE65 and LRAT levels accelerated both the visual cycle rate and recovery rate of rod light sensitivity in Cntf -/- mice. Of note, rods and cones in Cntf -/- mice exhibited hypersusceptibility to light-induced degeneration. These results indicate that CNTF is a common extracellular factor that prevents excessive production of opsins, the photoreceptor outer segments, and 11-cis-retinal to protect rods and cones from photodamage.
Collapse
Affiliation(s)
- Songhua Li
- From the Neuroscience Center of Excellence and
| | - Kota Sato
- From the Neuroscience Center of Excellence and
| | - William C Gordon
- From the Neuroscience Center of Excellence and.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| | - Michael Sendtner
- the Institute of Clinical Neurobiology, University Hospital Würzburg, D-97078 Würzburg, Germany
| | - Nicolas G Bazan
- From the Neuroscience Center of Excellence and.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| | - Minghao Jin
- From the Neuroscience Center of Excellence and .,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| |
Collapse
|
31
|
Liu YH, Corbett C, Klaska IP, Makinen K, Nickerson JM, Cornall RJ, Kuffova L, Forrester JV. Partial retinal photoreceptor loss in a transgenic mouse model associated with reduced levels of interphotoreceptor retinol binding protein (IRBP, RBP3). Exp Eye Res 2018; 172:54-65. [PMID: 29571629 DOI: 10.1016/j.exer.2018.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Organ-specific transgenic membrane expression of hen egg lysozyme (HEL) as a "neo-self antigen" has been used in several models to study immunological tolerance. In this study we report the changes which occur in the B10.BR mouse retina when membrane-bound HEL is expressed in photoreceptors under the control of the promoter for interphotoreceptor retinoid binding protein (IRBP, RBP3). On direct clinical examination of the single transgenic (sTg-IRBP:HEL) mouse fundus, a low-level increase in retinal degeneration compared to non-transgenic controls was observed, presenting as drusenoid deposits and occasional small patches of atrophy. On histological examination, there was an overall shortening of outer segments and loss of photoreceptor nuclei in sTg-IRBP:HEL mice, which was more pronounced in the retinal periphery, particularly inferiorly. The fundoscopically observed lesions did not correlate with the photoreceptor shortening/loss but appeared to be located at the level of the retinal pigment epithelium/choriocapillaris layer and were an exaggeration in size and number of similar age-related changes found in wild type (WT) mice. In addition, neither the atrophic lesions nor the photoreceptor shortening were associated with common retinal degeneration genes, nor were they caused by exposure to light damage since mice housed at both high and low ambient light levels had similar degrees of retinal degeneration. Instead, sTg-IRBP:HEL mice expressed reduced levels of soluble retinal IRBP compared to WT mice which were present from postnatal day16 (P16) and preceded development of photoreceptor shortening (onset P21). We propose that insertion of the HEL transgene in the photoreceptor membrane disrupted normal photoreceptor function and led to reduced levels of soluble IRBP and retinal thinning. A similar phenotype has been observed in IRBP deficient mice. Despite the retinal thinning, the amount of HEL expressed in the retina was sufficient to act as an autoantigenic target when the mice were crossed to the HEL T cell receptor Tg mouse, since double transgenic (dTg-IRBP:HEL) mice spontaneously developed a severe uveoretinitis with onset at weaning. We suggest that, although membrane expression of foreign transgene products is likely to modify the structure and function of tissues and cells, the technology provides useful models to investigate mechanisms of antigen-specific immunological tolerance.
Collapse
Affiliation(s)
- Yi-Hsia Liu
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Clare Corbett
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Izabela P Klaska
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Institute of Ophthalmology, University College London, London, UK
| | - Kimmo Makinen
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Human Health, Novozymes A/S, Bagsvaerd, Denmark
| | | | | | - Lucia Kuffova
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Department of Ophthalmology, NHS Grampian, Aberdeen, UK
| | - John V Forrester
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; University of Western Australia, Lions Eye Institute, Perth, Western Australia, Australia.
| |
Collapse
|
32
|
Cook JD, Ng SY, Lloyd M, Eddington S, Sun H, Nathans J, Bok D, Radu RA, Travis GH. Peropsin modulates transit of vitamin A from retina to retinal pigment epithelium. J Biol Chem 2017; 292:21407-21416. [PMID: 29109151 DOI: 10.1074/jbc.m117.812701] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/31/2017] [Indexed: 11/06/2022] Open
Abstract
Peropsin is a non-visual opsin in both vertebrate and invertebrate species. In mammals, peropsin is present in the apical microvilli of retinal pigment epithelial (RPE) cells. These structures interdigitate with the outer segments of rod and cone photoreceptor cells. RPE cells play critical roles in the maintenance of photoreceptors, including the recycling of visual chromophore for the opsin visual pigments. Here, we sought to identify the function of peropsin in the mouse eye. To this end, we generated mice with a null mutation in the peropsin gene (Rrh). These mice exhibited normal retinal histology, normal morphology of outer segments and RPE cells, and no evidence of photoreceptor degeneration. Biochemically, Rrh-/- mice had ∼2-fold higher vitamin A (all-trans-retinol (all-trans-ROL)) in the neural retina following a photobleach and 5-fold lower retinyl esters in the RPE. This phenotype was similar to those reported in mice that lack interphotoreceptor retinoid-binding protein (IRBP) or cellular retinol-binding protein, suggesting that peropsin plays a role in the movement of all-trans-ROL from photoreceptors to the RPE. We compared the phenotypes in mice lacking both peropsin and IRBP with those of mice lacking peropsin or IRBP alone and found that the retinoid phenotype was similarly severe in each of these knock-out mice. We conclude that peropsin controls all-trans-ROL movement from the retina to the RPE or may regulate all-trans-ROL storage within the RPE. We propose that peropsin affects light-dependent regulation of all-trans-ROL uptake from photoreceptors into RPE cells through an as yet undefined mechanism.
Collapse
Affiliation(s)
- Jeremy D Cook
- From the Department of Ophthalmology, Stein Eye Institute
| | - Sze Yin Ng
- From the Department of Ophthalmology, Stein Eye Institute
| | - Marcia Lloyd
- From the Department of Ophthalmology, Stein Eye Institute
| | | | - Hui Sun
- From the Department of Ophthalmology, Stein Eye Institute.,Department of Physiology, and
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Neuroscience, and Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, and.,Howard Hughes Medical Institute, Baltimore, Maryland 21205
| | - Dean Bok
- From the Department of Ophthalmology, Stein Eye Institute
| | - Roxana A Radu
- From the Department of Ophthalmology, Stein Eye Institute
| | - Gabriel H Travis
- From the Department of Ophthalmology, Stein Eye Institute, .,Department of Biological Chemistry, School of Medicine, UCLA, Los Angeles, California 90095
| |
Collapse
|
33
|
Malechka VV, Moiseyev G, Takahashi Y, Shin Y, Ma JX. Impaired Rhodopsin Generation in the Rat Model of Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2222-2231. [PMID: 28734946 PMCID: PMC5809515 DOI: 10.1016/j.ajpath.2017.06.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 12/11/2022]
Abstract
Diabetic retinopathy is a common complication of diabetes mellitus. Diabetic patients experience functional deficits in dark adaptation, contrast sensitivity, and color perception before microvascular pathologies become apparent. Herein, we evaluated early changes in neural retinal function and in retinoid metabolism in the eye in diabetes. Streptozotocin-induced diabetic rats showed decreased a- and b-wave amplitudes of scotopic and photopic electroretinography responses 4 months after diabetes induction compared to nondiabetic controls. Although Western blot analysis revealed no difference in opsin expression, rhodopsin content was decreased in diabetic retinas, as shown by a difference in absorbance. Consistently, levels of 11-cis-retinal, the chromophore for visual pigments, were significantly lower in diabetic retinas compared to those in controls, suggesting a retinoid deficiency. Among visual cycle proteins, interphotoreceptor retinoid-binding protein and stimulated by retinoic acid 6 protein showed significantly lower levels in diabetic rats than those in nondiabetic controls. Similarly, serum levels of retinol-binding protein 4 and retinoids were significantly lower in diabetic rats. Overall, these results suggest that retinoid metabolism in the eye is impaired in type 1 diabetes, which leads to deficient generation of visual pigments and neural retinal dysfunction in early diabetes.
Collapse
Affiliation(s)
- Volha V Malechka
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Gennadiy Moiseyev
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Yusuke Takahashi
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Younghwa Shin
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jian-Xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
34
|
Chen C, Adler L, Goletz P, Gonzalez-Fernandez F, Thompson DA, Koutalos Y. Interphotoreceptor retinoid-binding protein removes all- trans-retinol and retinal from rod outer segments, preventing lipofuscin precursor formation. J Biol Chem 2017; 292:19356-19365. [PMID: 28972139 DOI: 10.1074/jbc.m117.795187] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/22/2017] [Indexed: 11/06/2022] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) is a specialized lipophilic carrier that binds the all-trans and 11-cis isomers of retinal and retinol, and this facilitates their transport between photoreceptors and cells in the retina. One of these retinoids, all-trans-retinal, is released in the rod outer segment by photoactivated rhodopsin after light excitation. Following its release, all-trans-retinal is reduced by the retinol dehydrogenase RDH8 to all-trans-retinol in an NADPH-dependent reaction. However, all-trans-retinal can also react with outer segment components, sometimes forming lipofuscin precursors, which after conversion to lipofuscin accumulate in the lysosomes of the retinal pigment epithelium and display cytotoxic effects. Here, we have imaged the fluorescence of all-trans-retinol, all-trans-retinal, and lipofuscin precursors in real time in single isolated mouse rod photoreceptors. We found that IRBP removes all-trans-retinol from individual rod photoreceptors in a concentration-dependent manner. The rate constant for retinol removal increased linearly with IRBP concentration with a slope of 0.012 min-1 μm-1 IRBP also removed all-trans-retinal, but with much less efficacy, indicating that the reduction of retinal to retinol promotes faster clearance of the photoisomerized rhodopsin chromophore. The presence of physiological IRBP concentrations in the extracellular medium resulted in lower levels of all-trans-retinal and retinol in rod outer segments following light exposure. It also prevented light-induced lipofuscin precursor formation, but it did not remove precursors that were already present. These findings reveal an important and previously unappreciated role of IRBP in protecting the photoreceptor cells against the cytotoxic effects of accumulated all-trans-retinal.
Collapse
Affiliation(s)
- Chunhe Chen
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Leopold Adler
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Patrice Goletz
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Federico Gonzalez-Fernandez
- the Departments of Ophthalmology and Pathology, University of Mississippi and G. V. (Sonny) Montgomery Veterans Affairs Medical Centers, Jackson, Mississippi 39216, and
| | - Debra A Thompson
- the Departments of Ophthalmology and Visual Sciences, and Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, Michigan 48105
| | - Yiannis Koutalos
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425,
| |
Collapse
|
35
|
Pharmacological Amelioration of Cone Survival and Vision in a Mouse Model for Leber Congenital Amaurosis. J Neurosci 2017; 36:5808-19. [PMID: 27225770 DOI: 10.1523/jneurosci.3857-15.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 04/20/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED RPE65, an abundant membrane-associate protein in the retinal pigment epithelium (RPE), is a key retinoid isomerase of the visual cycle necessary for generating 11-cis-retinal that functions not only as a molecular switch for activating cone and rod visual pigments in response to light stimulation, but also as a chaperone for normal trafficking of cone opsins to the outer segments. Many mutations in RPE65 are associated with Leber congenital amaurosis (LCA). A R91W substitution, the most frequent LCA-associated mutation, results in a severe decrease in protein level and enzymatic activity of RPE65, causing cone opsin mislocalization and early cone degeneration in the mutation knock-in mouse model of LCA. Here we show that R91W RPE65 undergoes ubiquitination-dependent proteasomal degradation in the knock-in mouse RPE due to misfolding. The 26S proteasome non-ATPase regulatory subunit 13 mediated degradation specifically of misfolded R91W RPE65. The mutation disrupted membrane-association and colocalization of RPE65 with lecithin:retinol acyltransferase (LRAT) that provides the hydrophobic substrate for RPE65. Systemic administration of sodium 4-phenylbutyrate (PBA), a chemical chaperone, increased protein stability, enzymatic activity, membrane-association, and colocalization of R91W RPE65 with LRAT. This rescue effect increased synthesis of 11-cis-retinal and 9-cis-retinal, a functional iso-chromophore of the visual pigments, led to alleviation of S-opsin mislocalization and cone degeneration in the knock-in mice. Importantly, PBA-treatment also improved cone-mediated vision in the mutant mice. These results indicate that PBA, a U.S. Food and Drug Administration-approved safe oral medication, may provide a noninvasive therapeutic intervention that delays daylight vision loss in patients with RPE65 mutations. SIGNIFICANCE STATEMENT LCA is a severe early onset retinal dystrophy. Recent clinical trials of gene therapy have implicated the need of an alternative or combination therapy to improve cone survival and function in patients with LCA caused by RPE65 mutations. Using a mouse model carrying the most frequent LCA-associated mutation (R91W), we found that the mutant RPE65 underwent ubiquitination-dependent proteasomal degradation due to misfolding. Treatment of the mice with a chemical chaperone partially corrected stability, enzymatic activity, and subcellular localization of R91W RPE65, which was also accompanied by improvement of cone survival and vision. These findings identify an in vivo molecular pathogenic mechanism for R91W mutation and provide a feasible pharmacological approach that can delay vision loss in patients with RPE65 mutations.
Collapse
|
36
|
Sun D, Sahu B, Gao S, Schur RM, Vaidya AM, Maeda A, Palczewski K, Lu ZR. Targeted Multifunctional Lipid ECO Plasmid DNA Nanoparticles as Efficient Non-viral Gene Therapy for Leber's Congenital Amaurosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624218 PMCID: PMC5363681 DOI: 10.1016/j.omtn.2017.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Development of a gene delivery system with high efficiency and a good safety profile is essential for successful gene therapy. Here we developed a targeted non-viral delivery system using a multifunctional lipid ECO for treating Leber’s congenital amaurosis type 2 (LCA2) and tested this in a mouse model. ECO formed stable nanoparticles with plasmid DNA (pDNA) at a low amine to phosphate (N/P) ratio and mediated high gene transfection efficiency in ARPE-19 cells because of their intrinsic properties of pH-sensitive amphiphilic endosomal escape and reductive cytosolic release (PERC). All-trans-retinylamine, which binds to interphotoreceptor retinoid-binding protein (IRBP), was incorporated into the nanoparticles via a polyethylene glycol (PEG) spacer for targeted delivery of pDNA into the retinal pigmented epithelium. The targeted ECO/pDNA nanoparticles provided high GFP expression in the RPE of 1-month-old Rpe65−/− mice after subretinal injection. Such mice also exhibited a significant increase in electroretinographic activity, and this therapeutic effect continued for at least 120 days. A safety study in wild-type BALB/c mice indicated no irreversible retinal damage following subretinal injection of these targeted nanoparticles. All-trans-retinylamine-modified ECO/pDNA nanoparticles provide a promising non-viral platform for safe and effective treatment of RPE-specific monogenic eye diseases such as LCA2.
Collapse
Affiliation(s)
- Da Sun
- Case Center for Biomolecular Engineering and Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Bhubanananda Sahu
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Songqi Gao
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44140, USA
| | - Rebecca M Schur
- Case Center for Biomolecular Engineering and Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Amita M Vaidya
- Case Center for Biomolecular Engineering and Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Krzysztof Palczewski
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44140, USA
| | - Zheng-Rong Lu
- Case Center for Biomolecular Engineering and Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
37
|
Markand S, Baskin NL, Chakraborty R, Landis E, Wetzstein SA, Donaldson KJ, Priyadarshani P, Alderson SE, Sidhu CS, Boatright JH, Iuvone PM, Pardue MT, Nickerson JM. IRBP deficiency permits precocious ocular development and myopia. Mol Vis 2016; 22:1291-1308. [PMID: 27829784 PMCID: PMC5082647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/25/2016] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Interphotoreceptor retinoid-binding protein (IRBP) is abundant in the subretinal space and binds retinoids and lipophilic molecules. The expression of IRBP begins precociously early in mouse eye development. IRBP-deficient (KO) mice show less cell death in the inner retinal layers of the retina before eyelid opening compared to wild-type C57BL/6J (WT) controls and eventually develop profound myopia. Thus, IRBP may play a role in eye development before visually-driven phenomena. We report comparative observations during the course of the natural development of eyes in WT and congenic IRBP KO mice that suggest IRBP is necessary at the early stages of mouse eye development for correct function and development to exist in later stages. METHODS We observed the natural development of congenic WT and IRBP KO mice, monitoring several markers of eye size and development, including haze and clarity of optical components in the eye, eye size, axial length, immunohistological markers of differentiation and eye development, visually guided behavior, and levels of a putative eye growth stop signal, dopamine. We conducted these measurements at several ages. Slit-lamp examinations were conducted at post-natal day (P)21. Fundus and spectral domain optical coherence tomography (SD-OCT) images were compared at P15, P30, P45, and P80. Enucleated eyes from P5 to P10 were measured for weight, and ocular dimensions were measured with a noncontact light-emitting diode (LED) micrometer. We counted the cells that expressed tyrosine hydroxylase (TH-positive cells) at P23-P36 using immunohistochemistry on retinal flatmounts. High-performance liquid chromatography (HPLC) was used to analyze the amounts of dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) at P7-P60. Monocular form deprivation in the right eye was induced using head-mounted goggles from P28 to P56. RESULTS Eye elongation and eye size in the IRBP KO mice began to increase at P7 compared to the WT mice. This difference increased until P12, and the difference was maintained thereafter. SD-OCT images in live mice confirmed previously reported retinal thinning of the outer nuclear layer in the IRBP KO mice compared to the WT mice from P15 to P80. Slit-lamp and fundoscopy examination outcomes did not differ between the WT and KO mice. SD-OCT measurements of the optical axis components showed that the only factor contributing to excess optical axis length was the depth of the vitreous body. No other component of optical axis length (including corneal thickness, anterior chamber depth, and lens thickness) was different from that of the WT mouse. The refractive power of the IRBP KO mice did not change in response to form deprivation. The number of retinal TH-positive cells was 28% greater in the IRBP KO retinas compared to the WT mice at P30. No significant differences were observed in the steady-state retinal DA or DOPAC levels or in the DOPAC/DA ratios between the WT and IRBP KO mice. CONCLUSIONS The IRBP KO mouse eye underwent precocious development and rapid eye size growth temporally about a day sooner than the WT mouse eye. Eye size began to differ between the WT and KO mice before eyelid opening, indicating no requirement for focus-dependent vision, and suggesting a developmental abnormality in the IRBP KO mouse eye that precedes form vision-dependent emmetropization. Additionally, the profoundly myopic KO eye did not respond to form deprivation compared to the non-deprived contralateral eye. Too much growth occurred in some parts of the eye, possibly upsetting a balance among size, differentiation, and focus-dependent growth suppression. Thus, the loss of IRBP may simply cause growth that is too rapid, possibly due to a lack of sequestration or buffering of morphogens that normally would bind to IRBP but are unbound in the IRBP KO eye. Despite the development of profound myopia, the DA levels in the IRBP KO mice were not statistically different from those in the WT mice, even with the excess of TH-positive cells in the IRBP KO mice compared to the WT mice. Overall, these data suggest that abnormal eye elongation in the IRBP KO mouse is independent of, precedes, and is epistatic to the process(es) of visually-driven refractive development.
Collapse
Affiliation(s)
- Shanu Markand
- Department of Ophthalmology, Emory University, Atlanta, GA
| | | | - Ranjay Chakraborty
- Department of Ophthalmology, Emory University, Atlanta, GA,Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA
| | - Erica Landis
- Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA,Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA
| | | | | | | | | | | | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, GA,Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA
| | - P. Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, GA,Department of Pharmacology, Emory University, Atlanta, GA
| | - Machelle T. Pardue
- Department of Ophthalmology, Emory University, Atlanta, GA,Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA,Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA,Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | | |
Collapse
|
38
|
Lee M, Li S, Sato K, Jin M. Interphotoreceptor Retinoid-Binding Protein Mitigates Cellular Oxidative Stress and Mitochondrial Dysfunction Induced by All-trans-Retinal. Invest Ophthalmol Vis Sci 2016; 57:1553-62. [PMID: 27046120 PMCID: PMC4824376 DOI: 10.1167/iovs.15-18551] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Point and null mutations in interphotoreceptor retinoid-binding protein (IRBP) cause retinal dystrophy in affected patients and IRBP-deficient mice with unknown mechanism. This study investigated whether IRBP protects cells from damages induced by all-trans-retinal (atRAL), which was increased in the Irbp−/− retina. Methods Wild-type and Irbp−/− mice retinal explants in buffer with or without purified IBRP were exposed to 800 lux light for different times and subjected to retinoid analysis by high-performance liquid chromatography. Purity of IRBP was determined by Coomassie Brilliant Blue staining and immunoblot analysis. Cellular damages induced by atRAL in the presence or absence of IRBP were evaluated in the mouse photoreceptor-derived 661W cells. Cell viability and death were measured by 3-(4,5-dimethyl-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) and TUNEL assays. Expression and modification levels of retinal proteins were determined by immunoblot analysis. Intracellular reactive oxygen species (ROS) and nitric oxide (NO) were detected with fluorogenic dyes and confocal microscopy. Mitochondrial membrane potential was analyzed by using JC-1 fluorescent probe and a flow cytometer. Results Content of atRAL in Irbp−/− retinal explants exposed to light for 40 minutes was significantly higher than that in wild-type retinas under the same light conditions. All-trans-retinal caused increase in cell death, tumor necrosis factor activation, and Adam17 upregulation in 661W cells. NADPH oxidase-1 (NOX1) upregulation, ROS generation, NO-mediated protein S-nitrosylation, and mitochondrial dysfunction were also observed in 661W cells treated with atRAL. These cytotoxic effects were significantly attenuated in the presence of IRBP. Conclusions Interphotoreceptor retinoid-binding protein is required for preventing accumulation of retinal atRAL, which causes inflammation, oxidative stress, and mitochondrial dysfunction of the cells.
Collapse
|
39
|
Sarfare S, Dacquay Y, Askari S, Nusinowitz S, Hubschman JP. Biocompatibility of a Synthetic Biopolymer for the Treatment of Rhegmatogenous Retinal Detachment. ACTA ACUST UNITED AC 2016; 6. [PMID: 26744635 DOI: 10.4172/2155-9570.1000475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The aim of this study is to evaluate the retinal safety and toxicity of a novel synthetic biopolymer to be used as a patch to treat rhegmatogenous retinal detachment. METHODS Thirty one adult wild type albino mice were divided in 2 groups. In Group A (n=9) 0.2 μl balanced salt solution (BSS) and in Group B (n=22), 0.2 μl biopolymer was injected in the subretinal space. Trans-scleral subretinal injection was performed in one eye and the fellow eye was used as control. In both groups, in vivo color fundus photography, electroretinogram (ERG), spectral domain optical coherence tomography (SD-OCT) were performed before injection and at days 7 and 14 post-intervention. Histological analysis was performed following euthanization at days 1, 7 and 21 post-injection. RESULTS The biopolymer was visualized in the subretinal space in vivo by SD-OCT and post-life by histology up to 1 week after the injection. There were no significant differences in ERG parameters between the two groups at 1 and 2 weeks post-injection. Minimal inflammatory response and loss of photoreceptor cells was only observed in the immediate proximity of the site of scleral perforation, which was similar in both groups. Overall integrity of the outer, inner retina and retinal pigment epithelial (RPE) layers was unaffected by the presence of the biopolymer in the subretinal space. CONCLUSIONS Functional and histological evaluation suggests that the synthetic biopolymer is non-inflammatory and non-toxic to the eye. It may represent a safe therapeutic agent in the future, for the treatment of rhegmatogenous retinal detachment.
Collapse
Affiliation(s)
- Shanta Sarfare
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Yann Dacquay
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Syed Askari
- Medicus Biosciences, 2528 Qume Drive, Unit 1, San José, California 95131, USA
| | - Steven Nusinowitz
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Jean-Pierre Hubschman
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
40
|
Jin M, Li S, Hu J, Jin HH, Jacobson SG, Bok D. Functional Rescue of Retinal Degeneration-Associated Mutant RPE65 Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 854:525-32. [PMID: 26427455 PMCID: PMC5623592 DOI: 10.1007/978-3-319-17121-0_70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
More than 100 different mutations in the RPE65 gene are associated with inherited retinal degeneration. Although some missense mutations have been shown to abolish isomerase activity of RPE65, the molecular bases leading to loss of function and retinal degeneration remain incompletely understood. Here we show that several missense mutations resulted in significant decrease in expression level of RPE65 in the human retinal pigment epithelium cells. The 26S proteasome non-ATPase regulatory subunit 13, a newly identified negative regulator of RPE65, mediated degradation of mutant RPE65s, which were misfolded and formed aggregates in the cells. Many mutations, including L22P, T101I, and L408P, were mapped on nonactive sites of RPE65. Enzyme activities of these mutant RPE65s were significantly rescued at low temperature, whereas mutant RPE65s with a distinct active site mutation could not be rescued under the same conditions. 4-phenylbutyrate (PBA) displayed a significant synergistic effect on the low temperature-mediated rescue of the mutant RPE65s. Our results suggest that a low temperature eye mask and PBA, a FDA-approved oral medicine, may provide a promising "protein repair therapy" that can enhance the efficacy of gene therapy for delaying retinal degeneration caused by RPE65 mutations.
Collapse
Affiliation(s)
- Minghao Jin
- Department of Ophthalmology and Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier St. Suite D, 70112, New Orleans, LA, USA.
| | - Songhua Li
- Department of Ophthalmology and Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier St. Suite D, 70112, New Orleans, LA, USA.
| | - Jane Hu
- Jules Stein Eye Institute, University of California, 90095, Los Angeles, CA, USA.
| | - Heather H Jin
- Department of Biology, Washington University, 63130, St. Louis, MO, USA.
| | - Samuel G Jacobson
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA.
| | - Dean Bok
- Jules Stein Eye Institute, University of California, 90095, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Abstract
Visual systems detect light by monitoring the effect of photoisomerization of a chromophore on the release of a neurotransmitter from sensory neurons, known as rod and cone photoreceptor cells in vertebrate retina. In all known visual systems, the chromophore is 11-cis-retinal complexed with a protein, called opsin, and photoisomerization produces all-trans-retinal. In mammals, regeneration of 11-cis-retinal following photoisomerization occurs by a thermally driven isomerization reaction. Additional reactions are required during regeneration to protect cells from the toxicity of aldehyde forms of vitamin A that are essential to the visual process. Photochemical and phototransduction reactions in rods and cones are identical; however, reactions of the rod and cone visual pigment regeneration cycles differ, and perplexingly, rod and cone regeneration cycles appear to use different mechanisms to overcome the energy barrier involved in converting all-trans- to 11-cis-retinoid. Abnormal processing of all-trans-retinal in the rod regeneration cycle leads to retinal degeneration, suggesting that excessive amounts of the retinoid itself or its derivatives are toxic. This line of reasoning led to the development of various approaches to modifying the activity of the rod visual cycle as a possible therapeutic approach to delay or prevent retinal degeneration in inherited retinal diseases and perhaps in the dry form of macular degeneration (geographic atrophy). In spite of great progress in understanding the functioning of rod and cone regeneration cycles at a molecular level, resolution of a number of remaining puzzling issues will offer insight into the amelioration of several blinding retinal diseases.
Collapse
|
42
|
Yang GQ, Chen T, Tao Y, Zhang ZM. Recent advances in the dark adaptation investigations. Int J Ophthalmol 2015; 8:1245-52. [PMID: 26682182 DOI: 10.3980/j.issn.2222-3959.2015.06.31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/28/2015] [Indexed: 12/15/2022] Open
Abstract
Dark adaptation is a highly sensitive neural function and may be the first symptom of many status including the physiologic and pathologic entity, suggesting that it could be instrumental for diagnose. However, shortcomings such as the lack of standardized parameters, the long duration of examination, and subjective randomness would substantially impede the use of dark adaptation in clinical work. In this review we summarize the recent research about the dark adaptation, including two visual cycles-canonical and cone-specific visual cycle, affecting factors and the methods for measuring dark adaptation. In the opinions of authors, intensive investigations are needed to be done for the widely use of this significant visual function in clinic.
Collapse
Affiliation(s)
- Guo-Qing Yang
- Department of Clinical Aerospace Medicine, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Tao Chen
- Department of Clinical Aerospace Medicine, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Ye Tao
- Department of Ophthalmology, Beidaihe Hospital of PLA, Beidaihe 066100, Hebei Province, China
| | - Zuo-Ming Zhang
- Department of Clinical Aerospace Medicine, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
43
|
Kolesnikov AV, Maeda A, Tang PH, Imanishi Y, Palczewski K, Kefalov VJ. Retinol dehydrogenase 8 and ATP-binding cassette transporter 4 modulate dark adaptation of M-cones in mammalian retina. J Physiol 2015; 593:4923-41. [PMID: 26350353 DOI: 10.1113/jp271285] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS This study explores the molecular mechanisms that regulate the recycling of chromophore required for pigment regeneration in mammalian cones. We report that two chromophore binding proteins, retinol dehydrogenase 8 (RDH8) and photoreceptor-specific ATP-binding cassette transporter (ABCA4) accelerate the dark adaptation of cones, first, directly, by facilitating the processing of chromophore in cones, and second, indirectly, by accelerating the turnover of chromophore in rods, which is then recycled and delivered to both rods and cones. Preventing competition with the rods by knocking out rhodopsin accelerated cone dark adaptation, demonstrating the interplay between rod and cone pigment regeneration driven by the retinal pigment epithelium (RPE). This novel interdependence of rod and cone pigment regeneration should be considered when developing therapies targeting the recycling of chromophore for rods, and evaluating residual cone function should be a critical test for such regimens targeting the RPE. ABSTRACT Rapid recycling of visual chromophore and regeneration of the visual pigment are critical for the continuous function of mammalian cone photoreceptors in daylight vision. However, the molecular mechanisms modulating the supply of visual chromophore to cones have remained unclear. Here we explored the roles of two chromophore-binding proteins, retinol dehydrogenase 8 (RDH8) and photoreceptor-specific ATP-binding cassette transporter 4 (ABCA4), in dark adaptation of mammalian cones. We report that young adult RDH8/ABCA4-deficient mice have normal M-cone morphology but reduced visual acuity and photoresponse amplitudes. Notably, the deletion of RDH8 and ABCA4 suppressed the dark adaptation of M-cones driven by both the intraretinal visual cycle and the retinal pigmented epithelium (RPE) visual cycle. This delay can be caused by two separate mechanisms: direct involvement of RDH8 and ABCA4 in cone chromophore processing, and an indirect effect from the delayed recycling of chromophore by the RPE due to its slow release from RDH8/ABCA4-deficient rods. Intriguingly, our data suggest that RDH8 could also contribute to the oxidation of cis-retinoids in cones, a key reaction of the retina visual cycle. Finally, we dissected the roles of rod photoreceptors and RPE for dark adaptation of M-cones. We found that rods suppress, whereas RPE promotes, cone dark adaptation. Thus, therapeutic approaches targeting the RPE visual cycle could have adverse effects on the function of cones, making the evaluation of residual cone function a critical test for regimens targeting the RPE.
Collapse
Affiliation(s)
- Alexander V Kolesnikov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peter H Tang
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.,Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yoshikazu Imanishi
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Krzysztof Palczewski
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| |
Collapse
|
44
|
Ghosh D, Haswell KM, Sprada M, Gonzalez-Fernandez F. Structure of zebrafish IRBP reveals fatty acid binding. Exp Eye Res 2015; 140:149-158. [PMID: 26344741 DOI: 10.1016/j.exer.2015.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 08/22/2015] [Accepted: 08/31/2015] [Indexed: 10/23/2022]
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) has a remarkable role in targeting and protecting all-trans and 11-cis retinol, and 11-cis retinal during the rod and cone visual cycles. Little is known about how the correct retinoid is efficiently delivered and removed from the correct cell at the required time. It has been proposed that different fatty composition at that the outer-segments and retinal-pigmented epithelium have an important role is regulating the delivery and uptake of the visual cycle retinoids at the cell-interphotoreceptor-matrix interface. Although this suggests intriguing mechanisms for the role of local fatty acids in visual-cycle retinoid trafficking, nothing is known about the structural basis of IRBP-fatty acid interactions. Such regulation may be mediated through IRBP's unusual repeating homologous modules, each containing about 300 amino acids. We have been investigating structure-function relationships of Zebrafish IRBP (zIRBP), which has only two tandem modules (z1 and z2), as a model for the more complex four-module mammalian IRBP's. Here we report the first X-ray crystal structure of a teleost IRBP, and the only structure with a bound ligand. The X-ray structure of z1, determined at 1.90 Å resolution, reveals a two-domain organization of the module (domains A and B). A deep hydrophobic pocket with a single bound molecule of oleic acid was identified within the N-terminal domain A. In fluorescence titrations assays, oleic acid displaced all-trans retinol from zIRBP. Our study, which provides the first structure of an IRBP with bound ligand, supports a potential role for fatty acids in regulating retinoid binding.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| | - Karen M Haswell
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Molly Sprada
- SUNY Eye Institute, State University of New York, USA
| | - Federico Gonzalez-Fernandez
- Medical Research & Development Service, G.V. (Sonny) Veterans Affairs Medical Center, Jackson, MS, USA; Departments of Ophthalmology and Pathology, University of Mississippi Medical Center, Jackson, MS, USA; SUNY Eye Institute, State University of New York, USA.
| |
Collapse
|
45
|
Kaylor JJ, Radu RA, Bischoff N, Makshanoff J, Hu J, Lloyd M, Eddington S, Bianconi T, Bok D, Travis GH. Diacylglycerol O-acyltransferase type-1 synthesizes retinyl esters in the retina and retinal pigment epithelium. PLoS One 2015; 10:e0125921. [PMID: 25974161 PMCID: PMC4431840 DOI: 10.1371/journal.pone.0125921] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/13/2015] [Indexed: 11/19/2022] Open
Abstract
Retinyl esters represent an insoluble storage form of vitamin A and are substrates for the retinoid isomerase (Rpe65) in cells of the retinal pigment epithelium (RPE). The major retinyl-ester synthase in RPE cells is lecithin:retinol acyl-transferase (LRAT). A second palmitoyl coenzyme A-dependent retinyl-ester synthase activity has been observed in RPE homogenates but the protein responsible has not been identified. Here we show that diacylglycerol O-acyltransferase-1 (DGAT1) is expressed in multiple cells of the retina including RPE and Müller glial cells. DGAT1 catalyzes the synthesis of retinyl esters from multiple retinol isomers with similar catalytic efficiencies. Loss of DGAT1 in dgat1 -/- mice has no effect on retinal anatomy or the ultrastructure of photoreceptor outer-segments (OS) and RPE cells. Levels of visual chromophore in dgat1 -/- mice were also normal. However, the normal build-up of all-trans-retinyl esters (all-trans-RE’s) in the RPE during the first hour after a deep photobleach of visual pigments in the retina was not seen in dgat1 -/- mice. Further, total retinyl-ester synthase activity was reduced in both dgat1 -/- retina and RPE.
Collapse
Affiliation(s)
- Joanna J. Kaylor
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| | - Roxana A. Radu
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Nicholas Bischoff
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jacob Makshanoff
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jane Hu
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Marcia Lloyd
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Shannan Eddington
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tran Bianconi
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Dean Bok
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Gabriel H. Travis
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
46
|
Simó R, Hernández C. Novel approaches for treating diabetic retinopathy based on recent pathogenic evidence. Prog Retin Eye Res 2015; 48:160-80. [PMID: 25936649 DOI: 10.1016/j.preteyeres.2015.04.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/13/2015] [Accepted: 04/21/2015] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy remains as a leading cause of blindness in developed countries. Current treatments target late stages of DR when vision has already been significantly affected. A better understanding of the pathogenesis of DR would permit the development of more efficient preventional/interventional strategies against early stages of DR. In this article a critical review of the state of the art of this issue is provided along with a discussion of problems which have yet to be overcome. Neuroprotection as a new approach for the treatment of the early stages of DR has been particularly emphasized. The development and progression of DR is not homogeneous and, apart from blood glucose levels and blood pressure, it depends on genetic factors which remain to be elucidated. In addition, the role of the pathogenic pathways is not the same in all patients. All these factors should be taken into account in the near future when an individualized oriented treatment for DR could become feasible. The new techniques in retinal imaging acquisition, the identification of useful circulating biomarkers and the individualized analysis of biological samples could facilitate the development of early and personalized therapy in the setting of DR. Finally, it should be noted that only a coordinated action among ophthalmologists, diabetologists, basic researchers, experts in pharmaco-economics and health care providers addressed to the design of rational strategies targeting prevention and the early stages of DR will be effective in reducing the burden and improving the clinical outcome of this devastating complication of diabetes.
Collapse
Affiliation(s)
- Rafael Simó
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas) and Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain.
| | - Cristina Hernández
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas) and Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain.
| |
Collapse
|
47
|
Zhu L, Shen W, Lyons B, Wang Y, Zhou F, Gillies MC. Dysregulation of inter-photoreceptor retinoid-binding protein (IRBP) after induced Müller cell disruption. J Neurochem 2015; 133:909-18. [PMID: 25692504 DOI: 10.1111/jnc.13075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 11/29/2022]
Abstract
Reduced expression of a ~150 kDa protein was unexpectedly observed while investigating Norrin protein in a transgenic murine model in which Müller cells can be selectively and inducibly disrupted. Isolation of this unknown protein via ion exchange and hydrophobic interaction chromatography followed by Tandem mass spectrometry identified it as Inter-photoreceptor retinoid-binding protein (IRBP). Significantly reduced IRBP mRNA expression was observed at the early and late stages after Müller cell disruption. IRBP protein expression was also consistently reduced to 5.7% of the control level as early as 1 week after Müller cell disruption. This down-regulation of IRBP was accompanied by focal hyperfluorescent dots and cytotoxic N-retinylidene-N-retinylethanolamine (A2E) accumulation. In vitro treatment of cone photoreceptor cell lines with conditioned medium collected from stressed Müller cells suggested that Müller cells regulated photoreceptors expression of IRBP via secreted factor(s). In vivo studies suggested that one of these secreted factors was tumour necrosis factor alpha (TNFα). These findings suggest that dysregulation of IRBP expression caused by Müller cell dysfunction may be an important early event in photoreceptor degeneration in some retinal diseases. This study reports down-regulation of inter-photoreceptor retinoid-binding protein (IRBP) in photoreceptors and retinoid cycle derangement after Müller cell disruption in a transgenic mouse model. The findings indicate that Müller cells communicate with photoreceptors in response to stress by secreting soluble protein factor(s). We propose that down-regulation of IRBP may represent an early and novel pathogenic mechanism in degenerative retinal diseases.
Collapse
Affiliation(s)
- Ling Zhu
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Weiyong Shen
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Brian Lyons
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Ying Wang
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, the University of Sydney, Sydney, NSW, Australia
| | - Mark C Gillies
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
48
|
Gonzalez-Fernandez F, Betts-Obregon B, Yust B, Mimun J, Sung D, Sardar D, Tsin AT. Interphotoreceptor retinoid-binding protein protects retinoids from photodegradation. Photochem Photobiol 2015; 91:371-8. [PMID: 25565073 DOI: 10.1111/php.12416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022]
Abstract
Retinol degrades rapidly in light into a variety of photoproducts. It is remarkable that visual cycle retinoids can evade photodegradation as they are exchanged between the photoreceptors, retinal pigment epithelium and Müller glia. Within the interphotoreceptor matrix, all-trans retinol, 11-cis retinol and retinal are bound by interphotoreceptor retinoid-binding protein (IRBP). Apart from its role in retinoid trafficking and targeting, could IRBP have a photoprotective function? HPLC was used to evaluate the ability of IRBP to protect all-trans and 11-cis retinols from photodegradation when exposed to incandescent light (0 to 8842 μW cm(-2)); time periods of 0-60 min, and bIRBP: retinol molar ratios of 1:1 to 1:5. bIRBP afforded a significant prevention of both all-trans and 11-cis retinol to rapid photodegradation. The effect was significant over the entire light intensity range tested, and extended to the bIRBP: retinol ratio 1:5. In view of the continual exposure of the retina to light, and the high oxidative stress in the outer retina, our results suggest IRBP may have an important protective role in the visual cycle by reducing photodegradation of all-trans and 11-cis retinols. This role of IRBP is particularly relevant in the high flux conditions of the cone visual cycle.
Collapse
Affiliation(s)
- Federico Gonzalez-Fernandez
- Medical Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS; Departments of Ophthalmology & Pathology, University of Mississippi School of Medicine, Jackson, MS; SUNY Eye Institute, State University of New York, Buffalo, NY
| | | | | | | | | | | | | |
Collapse
|
49
|
Xue Y, Shen SQ, Jui J, Rupp AC, Byrne LC, Hattar S, Flannery JG, Corbo JC, Kefalov VJ. CRALBP supports the mammalian retinal visual cycle and cone vision. J Clin Invest 2015; 125:727-38. [PMID: 25607845 DOI: 10.1172/jci79651] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/11/2014] [Indexed: 11/17/2022] Open
Abstract
Mutations in the cellular retinaldehyde-binding protein (CRALBP, encoded by RLBP1) can lead to severe cone photoreceptor-mediated vision loss in patients. It is not known how CRALBP supports cone function or how altered CRALBP leads to cone dysfunction. Here, we determined that deletion of Rlbp1 in mice impairs the retinal visual cycle. Mice lacking CRALBP exhibited M-opsin mislocalization, M-cone loss, and impaired cone-driven visual behavior and light responses. Additionally, M-cone dark adaptation was largely suppressed in CRALBP-deficient animals. While rearing CRALBP-deficient mice in the dark prevented the deterioration of cone function, it did not rescue cone dark adaptation. Adeno-associated virus-mediated restoration of CRALBP expression specifically in Müller cells, but not retinal pigment epithelial (RPE) cells, rescued the retinal visual cycle and M-cone sensitivity in knockout mice. Our results identify Müller cell CRALBP as a key component of the retinal visual cycle and demonstrate that this pathway is important for maintaining normal cone-driven vision and accelerating cone dark adaptation.
Collapse
|
50
|
Li S, Izumi T, Hu J, Jin HH, Siddiqui AAA, Jacobson SG, Bok D, Jin M. Rescue of enzymatic function for disease-associated RPE65 proteins containing various missense mutations in non-active sites. J Biol Chem 2014; 289:18943-56. [PMID: 24849605 PMCID: PMC4081934 DOI: 10.1074/jbc.m114.552117] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Over 70 different missense mutations, including a dominant mutation, in RPE65 retinoid isomerase are associated with distinct forms of retinal degeneration; however, the disease mechanisms for most of these mutations have not been studied. Although some mutations have been shown to abolish enzyme activity, the molecular mechanisms leading to the loss of enzymatic function and retinal degeneration remain poorly understood. Here we show that the 26 S proteasome non-ATPase regulatory subunit 13 (PSMD13), a newly identified negative regulator of RPE65, plays a critical role in regulating pathogenicity of three mutations (L22P, T101I, and L408P) by mediating rapid degradation of mutated RPE65s via a ubiquitination- and proteasome-dependent non-lysosomal pathway. These mutant RPE65s were misfolded and formed aggregates or high molecular complexes via disulfide bonds. Interaction of PSMD13 with mutant RPE65s promoted degradation of misfolded but not properly folded mutant RPE65s. Many mutations, including L22P, T101I, and L408P, were mapped on non-active sites. Although their activities were very low, these mutant RPE65s were catalytically active and could be significantly rescued at low temperature, whereas mutant RPE65s with a distinct active site mutation could not be rescued under the same conditions. Sodium 4-phenylbutyrate and glycerol displayed a significant synergistic effect on the low temperature rescue of the mutant RPE65s by promoting proper folding, reducing aggregation, and increasing membrane association. Our results suggest that a low temperature eye mask and sodium 4-phenylbutyrate, a United States Food and Drug Administration-approved oral medicine, may provide a promising "protein repair therapy" that can enhance the efficacy of gene therapy by reducing the cytotoxic effect of misfolded mutant RPE65s.
Collapse
Affiliation(s)
- Songhua Li
- From the Department of Ophthalmology and Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Tadahide Izumi
- Graduate Center for Toxicology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Jane Hu
- Jules Stein Eye Institute and Department of Neurobiology, University of California, Los Angeles, California 90095
| | - Heather H Jin
- Department of Biology, Washington University, St. Louis, Missouri 63130
| | | | - Samuel G Jacobson
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Dean Bok
- Jules Stein Eye Institute and Department of Neurobiology, University of California, Los Angeles, California 90095
| | - Minghao Jin
- From the Department of Ophthalmology and Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112,
| |
Collapse
|