1
|
Maritato R, Medugno A, D'Andretta E, De Riso G, Lupo M, Botta S, Marrocco E, Renda M, Sofia M, Mussolino C, Bacci ML, Surace EM. A DNA base-specific sequence interposed between CRX and NRL contributes to RHODOPSIN expression. Sci Rep 2024; 14:26313. [PMID: 39487168 PMCID: PMC11530525 DOI: 10.1038/s41598-024-76664-8] [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: 07/05/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024] Open
Abstract
Gene expression emerges from DNA sequences through the interaction of transcription factors (TFs) with DNA cis-regulatory sequences. In eukaryotes, TFs bind to transcription factor binding sites (TFBSs) with differential affinities, enabling cell-specific gene expression. In this view, DNA enables TF binding along a continuum ranging from low to high affinity depending on its sequence composition; however, it is not known whether evolution has entailed a further level of entanglement between DNA-protein interaction. Here we found that the composition and length (22 bp) of the DNA sequence interposed between the CRX and NRL retinal TFs in the proximal promoter of RHODOPSIN (RHO) largely controls the expression levels of RHO. Mutagenesis of CRX-NRL DNA linking sequences (here termed "DNA-linker") results in uncorrelated gene expression variation. In contrast, mutual exchange of naturally occurring divergent human and mouse Rho cis-regulatory elements conferred similar yet species-specific Rho expression levels. Two orthogonal DNA-binding proteins targeted to the DNA-linker either activate or repress the expression of Rho depending on the DNA-linker orientation relative to the CRX and NRL binding sites. These results argue that, in this instance, DNA itself contributes to CRX and NRL activities through a code based on specific base sequences of a defined length, ultimately determining optimal RHO expression levels.
Collapse
Affiliation(s)
- Rosa Maritato
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Alessia Medugno
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Emanuela D'Andretta
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Giulia De Riso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- AOU Federico II, Naples, Italy
| | - Mariangela Lupo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Salvatore Botta
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Mario Renda
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Martina Sofia
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Enrico Maria Surace
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
2
|
Rutan Woods CT, Makia MS, Lewis TR, Crane R, Zeibak S, Yu P, Kakakhel M, Castillo CM, Arshavsky VY, Naash MI, Al-Ubaidi MR. Downregulation of rhodopsin is an effective therapeutic strategy in ameliorating peripherin-2-associated inherited retinal disorders. Nat Commun 2024; 15:4756. [PMID: 38834544 PMCID: PMC11150396 DOI: 10.1038/s41467-024-48846-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 05/15/2024] [Indexed: 06/06/2024] Open
Abstract
Given the absence of approved treatments for pathogenic variants in Peripherin-2 (PRPH2), it is imperative to identify a universally effective therapeutic target for PRPH2 pathogenic variants. To test the hypothesis that formation of the elongated discs in presence of PRPH2 pathogenic variants is due to the presence of the full complement of rhodopsin in absence of the required amounts of functional PRPH2. Here we demonstrate the therapeutic potential of reducing rhodopsin levels in ameliorating disease phenotype in knockin models for p.Lys154del (c.458-460del) and p.Tyr141Cys (c.422 A > G) in PRPH2. Reducing rhodopsin levels improves physiological function, mitigates the severity of disc abnormalities, and decreases retinal gliosis. Additionally, intravitreal injections of a rhodopsin-specific antisense oligonucleotide successfully enhance the physiological function of photoreceptors and improves the ultrastructure of discs in mutant mice. Presented findings shows that reducing rhodopsin levels is an effective therapeutic strategy for the treatment of inherited retinal degeneration associated with PRPH2 pathogenic variants.
Collapse
Affiliation(s)
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Tylor R Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Stephanie Zeibak
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Paul Yu
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Carson M Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
3
|
Motipally SI, Kolson DR, Guan T, Kolandaivelu S. Aberrant lipid accumulation and retinal pigmental epithelium dysfunction in PRCD-deficient mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584131. [PMID: 38558979 PMCID: PMC10979840 DOI: 10.1101/2024.03.08.584131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Progressive Rod-Cone Degeneration (PRCD) is an integral membrane protein found in photoreceptor outer segment (OS) disc membranes and its function remains unknown. Mutations in Prcd are implicated in Retinitis pigmentosa (RP) in humans and multiple dog breeds. PRCD-deficient models exhibit decreased levels of cholesterol in the plasma. However, potential changes in the retinal cholesterol remain unexplored. In addition, impaired phagocytosis observed in these animal models points to potential deficits in the retinal pigment epithelium (RPE). Here, using a Prcd -/- murine model we investigated the alterations in the retinal cholesterol levels and impairments in the structural and functional integrity of the RPE. Lipidomic and immunohistochemical analyses show a 5-fold increase in the levels of cholesteryl esters (C.Es) and accumulation of neutral lipids in the PRCD-deficient retina, respectively, indicating alterations in total retinal cholesterol. Longitudinal fundus and spectral domain optical coherence tomography (SD-OCT) examinations showed focal lesions and RPE hyperreflectivity. Strikingly, the RPE of Prcd -/- mice exhibited age-related pathological features such as neutral lipid deposits, lipofuscin accumulation, Bruch's membrane (BrM) thickening and drusenoid focal deposits, mirroring an Age-related Macular Degeneration (AMD)-like phenotype. We propose that the extensive lipofuscin accumulation likely impairs lysosomal function, leading to the defective phagocytosis observed in Prcd -/- mice. Our findings support the dysregulation of retinal cholesterol homeostasis in the absence of PRCD. Further, we demonstrate that progressive photoreceptor degeneration in Prcd -/- mice is accompanied by progressive structural and functional deficits in the RPE, which likely exacerbates vision loss over time.
Collapse
|
4
|
Liu Y, Xia P, Yan F, Yuan M, Yuan H, Du Y, Yan J, Song Q, Zhang T, Hu D, Shen Y. Engineered Extracellular Vesicles for Delivery of an IL-1 Receptor Antagonist Promote Targeted Repair of Retinal Degeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302962. [PMID: 37518765 DOI: 10.1002/smll.202302962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/28/2023] [Indexed: 08/01/2023]
Abstract
Retinal degeneration (RD) is an irreversible blinding disease that seriously affects patients' daily activities and mental health. Targeting hyperactivated microglia and regulating polarization are promising strategies for treating the disease. Mesenchymal stem cell (MSC) transplantation is proven to be an effective treatment due to its immunomodulatory and regenerative properties. However, the low efficiency of cell migration and integration of MSCs remains a major obstacle to clinical use. The goal of this study is to develop a nanodelivery system that targets hyperactivated microglia and inhibits their release of proinflammatory factors, to achieve durable neuroprotection. This approach is to engineer extracellular vesicles (EVs) isolated from MSC, modify them with a cyclic RGD (cRGD) peptide on their surface, and load them with an antagonist of the IL-1 receptor, anakinra. Comparing with non-engineered EVs, it is observed that engineered cRGD-EVs exhibit an increased targeting efficiency against hyperactivated microglia and strongly protected photoreceptors in experimental RD cells and animal models. This study provides a strategy to improve drug delivery to degenerated retinas and offers a promising approach to improve the treatment of RD through targeted modulation of the immune microenvironment via engineered cRGD-EVs.
Collapse
Affiliation(s)
- Yizong Liu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Peng Xia
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, P. R. China
| | - Feiyue Yan
- Frontier Science Center of Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, 430071, P. R. China
| | - Man Yuan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Haitao Yuan
- Department of Geriatrics, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, P. R. China
| | - Yuxin Du
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Jiangbo Yan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Qiulin Song
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Tianlu Zhang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Danping Hu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
- Frontier Science Center of Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, 430071, P. R. China
| |
Collapse
|
5
|
Sun C, Ruzycki PA, Chen S. Rho enhancers play unexpectedly minor roles in Rhodopsin transcription and rod cell integrity. Sci Rep 2023; 13:12899. [PMID: 37558693 PMCID: PMC10412641 DOI: 10.1038/s41598-023-39979-6] [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: 12/06/2022] [Accepted: 08/02/2023] [Indexed: 08/11/2023] Open
Abstract
Enhancers function with a basal promoter to control the transcription of target genes. Enhancer regulatory activity is often studied using reporter-based transgene assays. However, unmatched results have been reported when selected enhancers are silenced in situ. In this study, using genomic deletion analysis in mice, we investigated the roles of two previously identified enhancers and the promoter of the Rho gene that codes for the visual pigment rhodopsin. The Rho gene is robustly expressed by rod photoreceptors of the retina, and essential for the subcellular structure and visual function of rod photoreceptors. Mutations in RHO cause severe vision loss in humans. We found that each Rho regulatory region can independently mediate local epigenomic changes, but only the promoter is absolutely required for establishing active Rho chromatin configuration and transcription and maintaining the cell integrity and function of rod photoreceptors. To our surprise, two Rho enhancers that enable strong promoter activation in reporter assays are largely dispensable for Rho expression in vivo. Only small and age-dependent impact is detectable when both enhancers are deleted. Our results demonstrate context-dependent roles of enhancers and highlight the importance of studying functions of cis-regulatory regions in the native genomic context.
Collapse
Affiliation(s)
- Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, USA
| | - Philip A Ruzycki
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, USA.
- Department of Genetics, Washington University, 660 South Euclid Avenue, MSC 8096-0006-11, Saint Louis, MO, 63110, USA.
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University, 660 South Euclid Avenue, MSC 8096-0006-06, Saint Louis, MO, 63110, USA.
| |
Collapse
|
6
|
Lewis TR, Phan S, Castillo CM, Kim KY, Coppenrath K, Thomas W, Hao Y, Skiba NP, Horb ME, Ellisman MH, Arshavsky VY. Photoreceptor disc incisures form as an adaptive mechanism ensuring the completion of disc enclosure. eLife 2023; 12:e89160. [PMID: 37449984 PMCID: PMC10361718 DOI: 10.7554/elife.89160] [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: 05/05/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023] Open
Abstract
The first steps of vision take place within a stack of tightly packed disc-shaped membranes, or 'discs', located in the outer segment compartment of photoreceptor cells. In rod photoreceptors, discs are enclosed inside the outer segment and contain deep indentations in their rims called 'incisures'. The presence of incisures has been documented in a variety of species, yet their role remains elusive. In this study, we combined traditional electron microscopy with three-dimensional electron tomography to demonstrate that incisures are formed only after discs become completely enclosed. We also observed that, at the earliest stage of their formation, discs are not round as typically depicted but rather are highly irregular in shape and resemble expanding lamellipodia. Using genetically manipulated mice and frogs and measuring outer segment protein abundances by quantitative mass spectrometry, we further found that incisure size is determined by the molar ratio between peripherin-2, a disc rim protein critical for the process of disc enclosure, and rhodopsin, the major structural component of disc membranes. While a high perpherin-2 to rhodopsin ratio causes an increase in incisure size and structural complexity, a low ratio precludes incisure formation. Based on these data, we propose a model whereby normal rods express a modest excess of peripherin-2 over the amount required for complete disc enclosure in order to ensure that this important step of disc formation is accomplished. Once the disc is enclosed, the excess peripherin-2 incorporates into the rim to form an incisure.
Collapse
Affiliation(s)
- Tylor R Lewis
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Sebastien Phan
- National Center for Microscopy and Imaging Research, School of Medicine, University of California, San DiegoLa JollaUnited States
| | - Carson M Castillo
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, School of Medicine, University of California, San DiegoLa JollaUnited States
| | - Kelsey Coppenrath
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus ResourceWoods HoleUnited States
| | - William Thomas
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus ResourceWoods HoleUnited States
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Nikolai P Skiba
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Marko E Horb
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus ResourceWoods HoleUnited States
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, School of Medicine, University of California, San DiegoLa JollaUnited States
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
- Department of Pharmacology and Cancer Biology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
7
|
Lewis TR, Phan S, Castillo CM, Kim KY, Coppenrath K, Thomas W, Hao Y, Skiba NP, Horb ME, Ellisman MH, Arshavsky VY. Photoreceptor disc incisures form as an adaptive mechanism ensuring the completion of disc enclosure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.06.535932. [PMID: 37066355 PMCID: PMC10104153 DOI: 10.1101/2023.04.06.535932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The first steps of vision take place within a stack of tightly packed disc-shaped membranes, or "discs", located in the outer segment compartment of photoreceptor cells. In rod photoreceptors, discs are enclosed inside the outer segment and contain deep indentations in their rims called "incisures". The presence of incisures has been documented in a variety of species, yet their role remains elusive. In this study, we combined traditional electron microscopy with three-dimensional electron tomography to demonstrate that incisures are formed only after discs become completely enclosed. We also observed that, at the earliest stage of their formation, discs are not round as typically depicted but rather are highly irregular in shape and resemble expanding lamellipodia. Using genetically manipulated mice and frogs and measuring outer segment protein abundances by quantitative mass spectrometry, we further found that incisure size is determined by the molar ratio between peripherin-2, a disc rim protein critical for the process of disc enclosure, and rhodopsin, the major structural component of disc membranes. While a high perpherin-2 to rhodopsin ratio causes an increase in incisure size and structural complexity, a low ratio precludes incisure formation. Based on these data, we propose a model whereby normal rods express a modest excess of peripherin-2 over the amount required for complete disc enclosure in order to ensure that this important step of disc formation is accomplished. Once the disc is enclosed, the excess peripherin-2 incorporates into the rim to form an incisure.
Collapse
Affiliation(s)
- Tylor R. Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Sebastien Phan
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA, USA, 92093
| | - Carson M. Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA, USA, 92093
| | - Kelsey Coppenrath
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Woods Hole, MA, USA, 02543
| | - William Thomas
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Woods Hole, MA, USA, 02543
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Nikolai P. Skiba
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Marko E. Horb
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Woods Hole, MA, USA, 02543
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA, USA, 92093
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA, 27710
| |
Collapse
|
8
|
Motipally SI, Kolandaivelu S. Absence of PRCD Leads to Dysregulation in Lipid Homeostasis Resulting in Disorganization of Photoreceptor Outer Segment Structure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:389-394. [PMID: 37440062 DOI: 10.1007/978-3-031-27681-1_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The outer segments of photoreceptors are specialized sensory cilia crucial for light detection. Any disruption that alters outer segment morphology can impair photoreceptor function and therefore vision. Progressive rod-cone degeneration (PRCD) is an integral membrane protein exclusively present in the photoreceptor OS with an unknown function. Multiple mutations in PRCD are linked with retinitis pigmentosa. The most common PRCD mutation observed in both human and multiple dog breeds, PRCD-C2Y, lacks the lipid modification "palmitoylation," which is crucial for protein stability and trafficking to the OS. Previous studies including ours show impaired disc morphogenesis and rhodopsin distributions in the absence of PRCD, but the precise role of PRCD in maintaining OS structure and function remains unclear. In this chapter, we discuss the potential role of PRCD in the maintenance of photoreceptor OS structural and functional integrity.
Collapse
Affiliation(s)
- Sree I Motipally
- Departments of Neuroscience and Biochemistry, Robert C. Byrd Health Sciences Centre, WVU Eye Institute, Morgantown, WV, USA
| | - Saravanan Kolandaivelu
- Departments of Ophthalmology, Visual Sciences and Biochemistry, Robert C. Byrd Health Sciences Centre, WVU Eye Institute, Morgantown, WV, USA.
| |
Collapse
|
9
|
Langouët M, Jolicoeur C, Javed A, Mattar P, Gearhart MD, Daiger SP, Bertelsen M, Tranebjærg L, Rendtorff ND, Grønskov K, Jespersgaard C, Chen R, Sun Z, Li H, Alirezaie N, Majewski J, Bardwell VJ, Sui R, Koenekoop RK, Cayouette M. Mutations in BCOR, a co-repressor of CRX/OTX2, are associated with early-onset retinal degeneration. SCIENCE ADVANCES 2022; 8:eabh2868. [PMID: 36070393 PMCID: PMC9451151 DOI: 10.1126/sciadv.abh2868] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/21/2022] [Indexed: 06/10/2023]
Abstract
Many transcription factors regulating the production, survival, and function of photoreceptor cells have been identified, but little is known about transcriptional co-regulators in retinal health and disease. Here, we show that BCL6 co-repressor (BCOR), a Polycomb repressive complex 1 factor mutated in various cancers, is involved in photoreceptor degenerative diseases. Using proteomics and transcription assays, we report that BCOR interacts with the transcription factors CRX and OTX2 and reduces their ability to activate the promoters of photoreceptor-specific genes. CUT&RUN sequencing further shows that BCOR shares genome-wide binding profiles with CRX/OTX2, consistent with a general co-repression activity. We also identify missense mutations in human BCOR in five families that have no evidence of cancer but present severe early-onset X-linked retinal degeneration. Last, we show that the human BCOR mutants cause degeneration when expressed in the mouse retina and have enhanced repressive activity on OTX2. These results uncover a role for BCOR in photoreceptors in both health and disease.
Collapse
Affiliation(s)
- Maéva Langouët
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Christine Jolicoeur
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Awais Javed
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Pierre Mattar
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Micah D. Gearhart
- Department of Genetics, Cell Biology and Development, Development Biology Center, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Stephen P. Daiger
- EHGED Department, Human Genetics Center, School of Public Health, University of Texas HSC, Houston, TX 77030, USA
| | - Mette Bertelsen
- Department of Clinical Genetics, Rigshospitalet, The Kennedy Centre, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet, The Kennedy Centre, Glostrup, Denmark
| | - Lisbeth Tranebjærg
- Department of Clinical Genetics, Rigshospitalet, The Kennedy Centre, Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Denmark
| | - Nanna D. Rendtorff
- Department of Clinical Genetics, Rigshospitalet, The Kennedy Centre, Copenhagen, Denmark
| | - Karen Grønskov
- Department of Clinical Genetics, Rigshospitalet, The Kennedy Centre, Copenhagen, Denmark
| | - Catherine Jespersgaard
- Department of Clinical Genetics, Rigshospitalet, The Kennedy Centre, Copenhagen, Denmark
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zixi Sun
- Department of Ophthalmology, State Key Laboratory of Complex Severe and Rare diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Hui Li
- Department of Ophthalmology, State Key Laboratory of Complex Severe and Rare diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Najmeh Alirezaie
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Vivian J. Bardwell
- Department of Genetics, Cell Biology and Development, Development Biology Center, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ruifang Sui
- Department of Ophthalmology, State Key Laboratory of Complex Severe and Rare diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Robert K. Koenekoop
- Departments of Pediatric Surgery, Human Genetics, Adult Ophthalmology and the McGill Ocular Genetics Laboratory, McGill University Health Center Research Institute, Montreal, QC, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
10
|
Ziółkowska N, Lewczuk B. Profiles of Rho, Opn4, c-Fos, and Birc5 mRNA expression in Wistar rat retinas exposed to white or monochromatic light. Front Neuroanat 2022; 16:956000. [PMID: 36059433 PMCID: PMC9434339 DOI: 10.3389/fnana.2022.956000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
Abstract
Despite concern over potential retinal damage linked to exposure to light-emitting-diode (LED) light (particularly blue light), it remains unknown how exposure to low-intensity monochromatic LED light affects the expression of rhodopsin (Rho, a photopigment that mediates light-induced retinal degeneration), melanopsin (Opn4, a blue-light sensitive photopigment), c-Fos (associated with retinal damage/degeneration), and Birc5 (anti-apoptotic). This study investigated the mRNA expression profiles of these genes under exposure to white and monochromatic light (blue, red, green) in the retinas of albino rats under a cycle of 12 h of light and 12 h of darkness. In each group, 32 Wistar rats were exposed to one type of monochromatic-LED or white-fluorescent light for 7 day (150 lx). Retinal samples were taken for qPCR analysis and light and electron microscopy. Blue and green light exposure markedly decreased expression of Rho and Opn4 mRNA and increased expression of Birc5 and c-Fos mRNA (P < 0.05). In retinas from the blue-light group, loss and vesiculation of photoreceptor outer segments were visible, but not in retinas from the red-light and control group. Measurements of the photoreceptor inner and outer segments length revealed, that this length was significantly decreased in the blue- and green-light exposure groups (P < 0.02), but not in the red-light exposure group. Increased expression of Birc5 and decreased expression of Rho and Opn4 after exposure to blue and green light may be early responses that help to reduce light-induced retinal damage.
Collapse
Affiliation(s)
- Natalia Ziółkowska
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | |
Collapse
|
11
|
Pöge M, Mahamid J, Imanishi SS, Plitzko JM, Palczewski K, Baumeister W. Determinants shaping the nanoscale architecture of the mouse rod outer segment. eLife 2021; 10:e72817. [PMID: 34931611 PMCID: PMC8758146 DOI: 10.7554/elife.72817] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/09/2021] [Indexed: 12/02/2022] Open
Abstract
The unique membrane organization of the rod outer segment (ROS), the specialized sensory cilium of rod photoreceptor cells, provides the foundation for phototransduction, the initial step in vision. ROS architecture is characterized by a stack of identically shaped and tightly packed membrane disks loaded with the visual receptor rhodopsin. A wide range of genetic aberrations have been reported to compromise ROS ultrastructure, impairing photoreceptor viability and function. Yet, the structural basis giving rise to the remarkably precise arrangement of ROS membrane stacks and the molecular mechanisms underlying genetically inherited diseases remain elusive. Here, cryo-electron tomography (cryo-ET) performed on native ROS at molecular resolution provides insights into key structural determinants of ROS membrane architecture. Our data confirm the existence of two previously observed molecular connectors/spacers which likely contribute to the nanometer-scale precise stacking of the ROS disks. We further provide evidence that the extreme radius of curvature at the disk rims is enforced by a continuous supramolecular assembly composed of peripherin-2 (PRPH2) and rod outer segment membrane protein 1 (ROM1) oligomers. We suggest that together these molecular assemblies constitute the structural basis of the highly specialized ROS functional architecture. Our Cryo-ET data provide novel quantitative and structural information on the molecular architecture in ROS and substantiate previous results on proposed mechanisms underlying pathologies of certain PRPH2 mutations leading to blindness.
Collapse
Affiliation(s)
- Matthias Pöge
- Max Planck Institute of Biochemistry, Department of Molecular Structural BiologyMartinsriedGermany
| | - Julia Mahamid
- Max Planck Institute of Biochemistry, Department of Molecular Structural BiologyMartinsriedGermany
| | - Sanae S Imanishi
- Eugene and Marilyn Glick Eye Institute and the Department of Ophthalmology, Indiana University School of MedicineyIndianapolisUnited States
| | - Jürgen M Plitzko
- Max Planck Institute of Biochemistry, Department of Molecular Structural BiologyMartinsriedGermany
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute and the Department of Ophthalmology, Center for Translational Vision Research, Department of Physiology & Biophysics, Department of Chemistry, Department of Molecular Biology and BiochemistryIrvineUnited States
| | - Wolfgang Baumeister
- Max Planck Institute of Biochemistry, Department of Molecular Structural BiologyMartinsriedGermany
| |
Collapse
|
12
|
Zhang HJ, Liu XB, Chen XM, Kong QH, Liu YS, So KF, Chen JS, Xu Y, Mi XS, Tang SB. Lutein delays photoreceptor degeneration in a mouse model of retinitis pigmentosa. Neural Regen Res 2021; 17:1596-1603. [PMID: 34916446 PMCID: PMC8771084 DOI: 10.4103/1673-5374.330622] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Retinitis pigmentosa is a retinal disease characterized by photoreceptor degeneration. There is currently no effective treatment for retinitis pigmentosa. Although a mixture of lutein and other antioxidant agents has shown promising effects in protecting the retina from degeneration, the role of lutein alone remains unclear. In this study, we administered intragastric lutein to Pde6brd10 model mice, which display degeneration of retinal photoreceptors, on postnatal days 17 (P17) to P25, when rod apoptosis reaches peak. Lutein at the optimal protective dose of 200 mg/kg promoted the survival of photoreceptors compared with vehicle control. Lutein increased rhodopsin expression in rod cells and opsin expression in cone cells, in line with an increased survival rate of photoreceptors. Functionally, lutein improved visual behavior, visual acuity, and retinal electroretinogram responses in Pde6brd10 mice. Mechanistically, lutein reduced the expression of glial fibrillary acidic protein in Müller glial cells. The results of this study confirm the ability of lutein to postpone photoreceptor degeneration by reducing reactive gliosis of Müller cells in the retina and exerting anti-inflammatory effects. This study was approved by the Laboratory Animal Ethics Committee of Jinan University (approval No. LACUC-20181217-02) on December 17, 2018.
Collapse
Affiliation(s)
- Hui-Jun Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University; Department of Ophthalmology, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Xiao-Bin Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiong-Min Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Qi-Hang Kong
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Yu-Sang Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province; Aier Academician Station, Changsha, Hunan Province; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education, Guangzhou, Guangdong Province; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jian-Su Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province; Laboratory of Retinal Cell Biology, Aier Eye Institute; Aier Academician Station, Changsha, Hunan Province, China
| | - Ying Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province; Aier Academician Station, Changsha, Hunan Province; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education, Guangzhou, Guangdong Province; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xue-Song Mi
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province; Aier Academician Station, Changsha, Hunan Province, China
| | - Shi-Bo Tang
- Laboratory of Retinal Cell Biology, Aier Eye Institute; Aier Academician Station, Changsha, Hunan Province, China
| |
Collapse
|
13
|
Liu X, Jia R, Meng X, Li Y, Yang L. Retinal degeneration in humanized mice expressing mutant rhodopsin under the control of the endogenous murine promoter. Exp Eye Res 2021; 215:108893. [PMID: 34919893 DOI: 10.1016/j.exer.2021.108893] [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: 04/12/2021] [Revised: 10/31/2021] [Accepted: 12/08/2021] [Indexed: 11/04/2022]
Abstract
RHO is one of the most common genetic causes of autosomal dominant retinitis Pigmentosa (adRP) and there is no effective therapy for this disease. While rapidly developed CRISPR/Cas9 gene editing technology presents a promising therapeutic strategy to treat adRP. A large number of studies for treating adRP using CRISPR/Cas9 have been performed based on transgenic mouse models which are affected with adRP caused by mutant mouse rhodopsin allele, the counterpart of human rhodopsin. Recently, some RHO humanized mouse models like T17M, P23H are generated, which permit testing of the therapeutic effect of CRISPR/Cas9 in preclinical in vivo systems, without putting humans at risk. While available humanized mouse models are few compared to the number of known RHO mutations, but it is time-consuming and costly to build humanized mice for each mutation. We wonder whether a humanized mouse model having several mutations simultaneously can be developed, although which rarely occurs in patients, to investigate the therapeutic effect of CRISPR/Cas9 for RHO-mediated adRP in preclinical in vivo systems. Homology directed repair strategy combing with CRISPR/Cas9 was employed to introduce human RHO genomic fragment containing the replacement of mouse exon1(mE1) after the start codon to mE5 before the stop codon and all introns by the human counterparts. The human rhodopsin could express under the control of the endogenous murine promoter both transcriptionally and translationally in vivo. Human rhodopsin in humanized mouse lines (without mutation) could replace murine rhodopsin morphologically and functionally. While human rhodopsin containing T17M, G51D, G114R, R135W and P171R mutations simultaneously in mutant humanized (Mut-Rhowt/hum and Mut-Rhohum/hum) mouse lines caused retinal degeneration. Mut-Rhohum/hum mice suffered from severe retinal degeneration with defective formation of rod outer segment, leaving nonrecordable electroretinogram (ERG) at 3 months. Mut- Rhowt/hum mice had a slower rate of photoreceptors loss. In 7-month-old Mut- Rhowt/hum mice, statistically reduced scotopic ERG responses were visible compared with age-matched WT mice, but the shortened outer segment and thinner outer nuclear layer could be observed from 3 months. From 7 months to 9 months, significantly abnormal scotopic ERG responses were visible and photoreceptors loss were also obvious in 9-month-old Mut-Rhowt/hum mice. In 12-month-old Mut- Rhowt/hum mice, statistically reduced scotopic and photopic ERG responses and retinal degeneration throughout the retina were visible. Because scotopic responses were more affected than photopic responses in mutant humanized mice, demonstrating that rods dysfunction was more severe than cones dysfunction and deteriorated earlier, the pattern of retinal degeneration caused by mutant human rhodopsin was a typical rod-cone decay. Immunocytochemistry in cells indicated human rhodopsin proteins with 5 mutations aggregated in the cytoplasm and were also retained in the endoplasmic reticulum. The mutant human rhodopsin also accumulated in rod inner segments and cellular bodies in vivo. In conclusion, our humanized models provide excellent opportunities to study the human rhodopsin expression patterns. Our mutant humanized heterozygotes can provide opportunities to explore gene editing therapies via CRISPR/Cas9 for these five mutations in preclinical studies, it is time-saving and cost-effective.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Ruixuan Jia
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Xiang Meng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Ying Li
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Liping Yang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
14
|
Temporal Contrast Sensitivity Increases despite Photoreceptor Degeneration in a Mouse Model of Retinitis Pigmentosa. eNeuro 2021; 8:ENEURO.0020-21.2021. [PMID: 33509952 PMCID: PMC8059883 DOI: 10.1523/eneuro.0020-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 11/21/2022] Open
Abstract
The detection of temporal variations in amplitude of light intensity, or temporal contrast sensitivity (TCS), depends on the kinetics of rod photoresponse recovery. Uncharacteristically fast rod recovery kinetics are facets of both human patients and transgenic animal models with a P23H rhodopsin mutation, a prevalent cause of retinitis pigmentosa (RP). Here, we show that mice with this mutation (RhoP23H/+) exhibit an age-dependent and illumination-dependent enhancement in TCS compared with controls. At retinal illumination levels producing ≥1000 R*/rod/s or more, postnatal day 30 (P30) RhoP23H/+ mice exhibit a 1.2-fold to 2-fold increase in retinal and optomotor TCS relative to controls in response to flicker frequencies of 3, 6, and 12 Hz despite significant photoreceptor degeneration and loss of flash electroretinogram (ERG) b-wave amplitude. Surprisingly, the TCS of RhoP23H/+ mice further increases as degeneration advances. Enhanced TCS is also observed in a second model (rhodopsin heterozygous mice, Rho+/-) with fast rod recovery kinetics and no apparent retinal degeneration. In both mouse models, enhanced TCS is explained quantitatively by a comprehensive model that includes photoresponse recovery kinetics, density and collecting area of degenerating rods. Measurement of TCS may be a non-invasive early diagnostic tool indicative of rod dysfunction in some forms of retinal degenerative disease.
Collapse
|
15
|
Sechrest ER, Murphy J, Senapati S, Goldberg AFX, Park PSH, Kolandaivelu S. Loss of PRCD alters number and packaging density of rhodopsin in rod photoreceptor disc membranes. Sci Rep 2020; 10:17885. [PMID: 33087780 PMCID: PMC7577997 DOI: 10.1038/s41598-020-74628-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023] Open
Abstract
Progressive rod-cone degeneration (PRCD) is a small protein localized to photoreceptor outer segment (OS) disc membranes. Several mutations in PRCD are linked to retinitis pigmentosa (RP) in canines and humans, and while recent studies have established that PRCD is required for high fidelity disc morphogenesis, its precise role in this process remains a mystery. To better understand the part which PRCD plays in disease progression as well as its contribution to photoreceptor OS disc morphogenesis, we generated a Prcd-KO animal model using CRISPR/Cas9. Loss of PRCD from the retina results in reduced visual function accompanied by slow rod photoreceptor degeneration. We observed a significant decrease in rhodopsin levels in Prcd-KO retina prior to photoreceptor degeneration. Furthermore, ultrastructural analysis demonstrates that rod photoreceptors lacking PRCD display disoriented and dysmorphic OS disc membranes. Strikingly, atomic force microscopy reveals that many disc membranes in Prcd-KO rod photoreceptor neurons are irregular, containing fewer rhodopsin molecules and decreased rhodopsin packing density compared to wild-type discs. This study strongly suggests an important role for PRCD in regulation of rhodopsin incorporation and packaging density into disc membranes, a process which, when dysregulated, likely gives rise to the visual defects observed in patients with PRCD-associated RP.
Collapse
Affiliation(s)
- Emily R Sechrest
- Department of Pharmaceutical Sciences, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA.,Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA
| | - Joseph Murphy
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA.,Department of Biochemistry, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Saravanan Kolandaivelu
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA. .,Department of Biochemistry, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA.
| |
Collapse
|
16
|
Abstract
Numerous rhodopsin mutations have been implicated in night blindness and retinal degeneration, often with unclear etiology. D190N-rhodopsin (D190N-Rho) is a well-known inherited human mutation causing retinitis pigmentosa. Both higher-than-normal spontaneous-isomerization activity and misfolding/mistargeting of the mutant protein have been proposed as causes of the disease, but neither explanation has been thoroughly examined. We replaced wild-type rhodopsin (WT-Rho) in RhoD190N/WT mouse rods with a largely "functionally silenced" rhodopsin mutant to isolate electrical responses triggered by D190N-Rho activity, and found that D190N-Rho at the single-molecule level indeed isomerizes more frequently than WT-Rho by over an order of magnitude. Importantly, however, this higher molecular dark activity does not translate into an overall higher cellular dark noise, owing to diminished D190N-Rho content in the rod outer segment. Separately, we found that much of the degeneration and shortened outer-segment length of RhoD190N/WT mouse rods was not averted by ablating rod transducin in phototransduction-also consistent with D190N-Rho's higher isomerization activity not being the primary cause of disease. Instead, the low pigment content, shortened outer-segment length, and a moderate unfolded protein response implicate protein misfolding as the major pathogenic problem. Finally, D190N-Rho also provided some insight into the mechanism of spontaneous pigment excitation.
Collapse
|
17
|
Lewis TR, Shores CR, Cady MA, Hao Y, Arshavsky VY, Burns ME. The F220C and F45L rhodopsin mutations identified in retinitis pigmentosa patients do not cause pathology in mice. Sci Rep 2020; 10:7538. [PMID: 32371886 PMCID: PMC7200662 DOI: 10.1038/s41598-020-64437-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 11/12/2022] Open
Abstract
Retinitis pigmentosa is a retinal degenerative disease that leads to blindness through photoreceptor loss. Rhodopsin is the most frequently mutated protein in this disease. While many rhodopsin mutations have well-understood consequences that lead to cell death, the disease association of several rhodopsin mutations identified in retinitis pigmentosa patients, including F220C and F45L, has been disputed. In this study, we generated two knockin mouse lines bearing each of these mutations. We did not observe any photoreceptor degeneration in either heterozygous or homozygous animals of either line. F220C mice exhibited minor disruptions of photoreceptor outer segment dimensions without any mislocalization of outer segment proteins, whereas photoreceptors of F45L mice were normal. Suction electrode recordings from individual photoreceptors of both mutant lines showed normal flash sensitivity and photoresponse kinetics. Taken together, these data suggest that neither the F220C nor F45L mutation has pathological consequences in mice and, therefore, may not be causative of retinitis pigmentosa in humans.
Collapse
Affiliation(s)
- Tylor R Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Camilla R Shores
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, United States
| | - Martha A Cady
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Marie E Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, United States. .,Center for Neuroscience and Department of Ophthalmology & Vision Science, University of California, Davis, CA, 95616, United States.
| |
Collapse
|
18
|
Hauzman E. Adaptations and evolutionary trajectories of the snake rod and cone photoreceptors. Semin Cell Dev Biol 2020; 106:86-93. [PMID: 32359892 DOI: 10.1016/j.semcdb.2020.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
Abstract
Most vertebrates have duplex retinas, with two classes of photoreceptors, rods and cones. In the group of Snakes, however, distinct patterns of retinal morphology are associated with transitions between diurnal-nocturnal habits and reflect important adaptations of their visual system. Pure-cone, pure-rod and duplex retinas were described in different species, and this variability led Gordon Walls (1934) to formulate the transmutation theory, which suggests that rods and cones are not fixed entities, but can assume transitional states. Three opsin genes are expressed in retinas of most snake species, lws, rh1, and sws1, and recent studies have shown that the rhodopsin gene, rh1, is expressed in pure-cone retinas of diurnal snakes. This expression raised many questions about the nature of transmutation and functional aspects of the rhodopsin in a cone-like photoreceptor. Extreme differences in the retinal architecture of diurnal and nocturnal snakes also highlight the complexity of adaptations of their visual structures, which might have contributed to the adaptive radiation of this group and will be discussed in this review.
Collapse
Affiliation(s)
- Einat Hauzman
- Department of Experimental Psychology, Psychology Institute, University of São Paulo, Av. Professor Mello Moraes, 1721, Bloco A - D9. Butantã, São Paulo, CEP. 05508-030, Brazil.
| |
Collapse
|
19
|
Local, nonlinear effects of cGMP and Ca2+ reduce single photon response variability in retinal rods. PLoS One 2019; 14:e0225948. [PMID: 31805112 PMCID: PMC6894879 DOI: 10.1371/journal.pone.0225948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/14/2019] [Indexed: 11/26/2022] Open
Abstract
The single photon response (SPR) in vertebrate photoreceptors is inherently variable due to several stochastic events in the phototransduction cascade, the main one being the shutoff of photoactivated rhodopsin. Deactivation is driven by a random number of steps, each of random duration with final quenching occurring after a random delay. Nevertheless, variability of the SPR is relatively low, making the signal highly reliable. Several biophysical and mathematical mechanisms contributing to variability suppression have been examined by the authors. Here we investigate the contribution of local depletion of cGMP by PDE*, the non linear dependence of the photocurrent on cGMP, Ca2+ feedback by making use of a fully space resolved (FSR) mathematical model, applied to two species (mouse and salamander), by varying the cGMP diffusion rate severalfold and rod outer segment diameter by an order of magnitude, and by introducing new, more refined, and time dependent variability functionals. Globally well stirred (GWS) models, and to a lesser extent transversally well stirred models (TWS), underestimate the role of nonlinearities and local cGMP depletion in quenching the variability of the circulating current with respect to fully space resolved models (FSR). These distortions minimize the true extent to which SPR is stabilized by locality in cGMP depletion, nonlinear effects linking cGMP to current, and Ca2+ feedback arising from the physical separation of E* from the ion channels located on the outer shell, and the diffusion of these second messengers in the cytoplasm.
Collapse
|
20
|
Wang J, Xu D, Zhu T, Zhou Y, Chen X, Wang F, Zhang J, Tian H, Gao F, Zhang J, Jin C, Xu J, Lu L, Liu Q, Xu GT. Identification of two novel RHO mutations in Chinese retinitis pigmentosa patients. Exp Eye Res 2019; 188:107726. [DOI: 10.1016/j.exer.2019.107726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 11/29/2022]
|
21
|
Bocchero U, Tam BM, Chiu CN, Torre V, Moritz OL. Electrophysiological Changes During Early Steps of Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2019; 60:933-943. [PMID: 30840038 DOI: 10.1167/iovs.18-25347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The rhodopsin mutation P23H is responsible for a significant portion of autosomal-dominant retinitis pigmentosa, a disorder characterized by rod photoreceptor death. The mechanisms of toxicity remain unclear; previous studies implicate destabilization of P23H rhodopsin during light exposure, causing decreased endoplasmic reticulum (ER) exit and ER stress responses. Here, we probed phototransduction in Xenopus laevis rods expressing bovine P23H rhodopsin, in which retinal degeneration is inducible by light exposure, in order to examine early physiological changes that occur during retinal degeneration. Methods We recorded single-cell and whole-retina responses to light stimuli using electrophysiology. Moreover, we monitored morphologic changes in rods after different periods of light exposure. Results Initially, P23H rods had almost normal photoresponses, but following a brief light exposure varying from 4 to 32 photoisomerizations per disc, photoresponses became irreversibly prolonged. In intact retinas, rods began to shed OS fragments after a rod-saturating exposure of 12 minutes, corresponding to approximately 10 to 100 times more photoisomerizations. Conclusions Our results indicate that in P23H rods light-induced degeneration occurs in at least two stages, the first involving impairment of phototransduction and the second involving initiation of morphologic changes.
Collapse
Affiliation(s)
- Ulisse Bocchero
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Beatrice M Tam
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colette N Chiu
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vincent Torre
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Orson L Moritz
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Wright ZC, Loskutov Y, Murphy D, Stoilov P, Pugacheva E, Goldberg AFX, Ramamurthy V. ADP-Ribosylation Factor-Like 2 (ARL2) regulates cilia stability and development of outer segments in rod photoreceptor neurons. Sci Rep 2018; 8:16967. [PMID: 30446707 PMCID: PMC6240099 DOI: 10.1038/s41598-018-35395-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/28/2018] [Indexed: 01/31/2023] Open
Abstract
Photoreceptor cells are specialized neurons with a sensory cilium carrying an elaborate membrane structure, the outer segment (OS). Inherited mutations in genes involved in ciliogenesis frequently result in OS malformation and blindness. ADP-ribosylation factor-like 2 (ARL2) has recently been implicated in OS formation through its association with Binder of ARL2 (BART or ARL2BP), a protein linked to inherited blinding disease. To test the role of ARL2 in vision we created a transgenic mouse model expressing a tagged-dominant active form of human ARL2 (ARL2-Q70L) under a rod-specific promoter. Transgenic ARL2-Q70L animals exhibit reduced photoreceptor cell function as early as post-natal day 16 and progressive rod degeneration. We attribute loss of photoreceptor function to the defective OS morphogenesis in the ARL2-Q70L transgenic model. ARL2-Q70L expression results in shortened inner and outer segments, shortened and mislocalized axonemes and cytoplasmic accumulation of rhodopsin. In conclusion, we show that ARL2-Q70L is crucial for photoreceptor neuron sensory cilium development. Future research will expand upon our hypothesis that ARL2-Q70L mutant interferes with microtubule maintenance and tubulin regulation resulting in impaired growth of the axoneme and elaboration of the photoreceptor outer segment.
Collapse
Affiliation(s)
- Zachary C Wright
- Departments of Ophthalmology, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Yuriy Loskutov
- Departments of Biochemistry, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Daniel Murphy
- Departments of Biochemistry, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Peter Stoilov
- Departments of Biochemistry, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Elena Pugacheva
- Departments of Biochemistry, West Virginia University, Morgantown, West Virginia, 26506, USA
| | | | - Visvanathan Ramamurthy
- Departments of Ophthalmology, West Virginia University, Morgantown, West Virginia, 26506, USA. .,Departments of Biochemistry, West Virginia University, Morgantown, West Virginia, 26506, USA. .,Center for Neuroscience, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, 26506, USA.
| |
Collapse
|
23
|
Crespo C, Soroldoni D, Knust E. A novel transgenic zebrafish line for red opsin expression in outer segments of photoreceptor cells. Dev Dyn 2018; 247:951-959. [PMID: 29603474 PMCID: PMC6099204 DOI: 10.1002/dvdy.24631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Opsins are a group of light-sensitive proteins present in photoreceptor cells, which convert the energy of photons into electrochemical signals, thus allowing vision. Given their relevance, we aimed to visualize the two red opsins at subcellular scale in photoreceptor cells. RESULTS We generated a novel Zebrafish BAC transgenic line, which express fluorescently tagged, full-length Opsin 1 long-wave-sensitive 1 (Opn1lw1) and full-length Opsin 1 long-wave-sensitive 2 (Opn1lw2) under the control of their endogenous promoters. Both fusion proteins are localized in the outer segments of photoreceptor cells. During development, Opn1lw2-mKate2 is detected from the initial formation of outer segments onward. In contrast, Opn1lw1-mNeonGreen is first detected in juvenile Zebrafish at about 2 weeks postfertilization, and both opsins continue to be expressed throughout adulthood. It is important to note that the presence of the transgene did not significantly alter the size of outer segments. CONCLUSIONS We have generated a transgenic line that mimics the endogenous expression pattern of Opn1lw1 and Opn1lw2 in the developing and adult retina. In contrast to existing lines, our transgene design allows to follow protein localization. Hence, we expect that these lines could act as useful real-time reporters to directly measure phenomena in retinal development and disease models. Developmental Dynamics 247:951-959, 2018. © 2018 The Authors Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Cátia Crespo
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Elisabeth Knust
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
24
|
Vinberg F, Peshenko IV, Chen J, Dizhoor AM, Kefalov VJ. Guanylate cyclase-activating protein 2 contributes to phototransduction and light adaptation in mouse cone photoreceptors. J Biol Chem 2018; 293:7457-7465. [PMID: 29549122 DOI: 10.1074/jbc.ra117.001574] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/12/2018] [Indexed: 12/14/2022] Open
Abstract
Light adaptation of photoreceptor cells is mediated by Ca2+-dependent mechanisms. In darkness, Ca2+ influx through cGMP-gated channels into the outer segment of photoreceptors is balanced by Ca2+ extrusion via Na+/Ca2+, K+ exchangers (NCKXs). Light activates a G protein signaling cascade, which closes cGMP-gated channels and decreases Ca2+ levels in photoreceptor outer segment because of continuing Ca2+ extrusion by NCKXs. Guanylate cyclase-activating proteins (GCAPs) then up-regulate cGMP synthesis by activating retinal membrane guanylate cyclases (RetGCs) in low Ca2+ This activation of RetGC accelerates photoresponse recovery and critically contributes to light adaptation of the nighttime rod and daytime cone photoreceptors. In mouse rod photoreceptors, GCAP1 and GCAP2 both contribute to the Ca2+-feedback mechanism. In contrast, only GCAP1 appears to modulate RetGC activity in mouse cones because evidence of GCAP2 expression in cones is lacking. Surprisingly, we found that GCAP2 is expressed in cones and can regulate light sensitivity and response kinetics as well as light adaptation of GCAP1-deficient mouse cones. Furthermore, we show that GCAP2 promotes cGMP synthesis and cGMP-gated channel opening in mouse cones exposed to low Ca2+ Our biochemical model and experiments indicate that GCAP2 significantly contributes to the activation of RetGC1 at low Ca2+ when GCAP1 is not present. Of note, in WT mouse cones, GCAP1 dominates the regulation of cGMP synthesis. We conclude that, under normal physiological conditions, GCAP1 dominates the regulation of cGMP synthesis in mouse cones, but if its function becomes compromised, GCAP2 contributes to the regulation of phototransduction and light adaptation of cones.
Collapse
Affiliation(s)
- Frans Vinberg
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Igor V Peshenko
- Pennsylvania College of Optometry, Salus University, Elkins Park, Pennsylvania 19027
| | - Jeannie Chen
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California 90033
| | - Alexander M Dizhoor
- Pennsylvania College of Optometry, Salus University, Elkins Park, Pennsylvania 19027
| | - Vladimir J Kefalov
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110.
| |
Collapse
|
25
|
Athanasiou D, Aguila M, Bellingham J, Li W, McCulley C, Reeves PJ, Cheetham ME. The molecular and cellular basis of rhodopsin retinitis pigmentosa reveals potential strategies for therapy. Prog Retin Eye Res 2018; 62:1-23. [PMID: 29042326 PMCID: PMC5779616 DOI: 10.1016/j.preteyeres.2017.10.002] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/03/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
Inherited mutations in the rod visual pigment, rhodopsin, cause the degenerative blinding condition, retinitis pigmentosa (RP). Over 150 different mutations in rhodopsin have been identified and, collectively, they are the most common cause of autosomal dominant RP (adRP). Mutations in rhodopsin are also associated with dominant congenital stationary night blindness (adCSNB) and, less frequently, recessive RP (arRP). Recessive RP is usually associated with loss of rhodopsin function, whereas the dominant conditions are a consequence of gain of function and/or dominant negative activity. The in-depth characterisation of many rhodopsin mutations has revealed that there are distinct consequences on the protein structure and function associated with different mutations. Here we categorise rhodopsin mutations into seven discrete classes; with defects ranging from misfolding and disruption of proteostasis, through mislocalisation and disrupted intracellular traffic to instability and altered function. Rhodopsin adRP offers a unique paradigm to understand how disturbances in photoreceptor homeostasis can lead to neuronal cell death. Furthermore, a wide range of therapies have been tested in rhodopsin RP, from gene therapy and gene editing to pharmacological interventions. The understanding of the disease mechanisms associated with rhodopsin RP and the development of targeted therapies offer the potential of treatment for this currently untreatable neurodegeneration.
Collapse
Affiliation(s)
| | - Monica Aguila
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - James Bellingham
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Wenwen Li
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Caroline McCulley
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Philip J Reeves
- School of Biological Sciences, University of Essex, Wivenhoe Park, Essex CO4 3SQ, UK.
| | | |
Collapse
|
26
|
Feehan JM, Chiu CN, Stanar P, Tam BM, Ahmed SN, Moritz OL. Modeling Dominant and Recessive Forms of Retinitis Pigmentosa by Editing Three Rhodopsin-Encoding Genes in Xenopus Laevis Using Crispr/Cas9. Sci Rep 2017; 7:6920. [PMID: 28761125 PMCID: PMC5537283 DOI: 10.1038/s41598-017-07153-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/27/2017] [Indexed: 11/11/2022] Open
Abstract
The utility of Xenopus laevis, a common research subject for developmental biology, retinal physiology, cell biology, and other investigations, has been limited by lack of a robust gene knockout or knock-down technology. Here we describe manipulation of the X. laevis genome using CRISPR/Cas9 to model the human disorder retinitis pigmentosa, and to introduce point mutations or exogenous DNA sequences. We introduced and characterized in-frame and out-of-frame insertions and deletions in three genes encoding rhodopsin by co-injection of Cas9 mRNA, eGFP mRNA, and single guide RNAs into fertilized eggs. Deletions were characterized by direct sequencing and cloning; phenotypes were assessed by assays of rod opsin in retinal extracts, and confocal microscopy of cryosectioned and immunolabeled contralateral eyes. We obtained germline transmission of editing to F1 offspring. In-frame deletions frequently caused dominant retinal degeneration associated with rhodopsin biosynthesis defects, while frameshift phenotypes were consistent with knockout. We inserted eGFP or point mutations into rhodopsin genes by co-injection of repair fragments with homology to the Cas9 target sites. Our techniques can produce high frequency gene editing in X. laevis, permitting analysis in the F0 generation, and advancing the utility of X. laevis as a subject for biological research and disease modeling.
Collapse
Affiliation(s)
- Joanna M Feehan
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V5Z 3N9
- The Sainsbury Laboratory, Colney Ln, Norwich Research Park, Norwich, Norfolk, UK, NR4 7UH
| | - Colette N Chiu
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V5Z 3N9
| | - Paloma Stanar
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V5Z 3N9
| | - Beatrice M Tam
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V5Z 3N9
| | - Sheikh N Ahmed
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V5Z 3N9
| | - Orson L Moritz
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada, V5Z 3N9.
| |
Collapse
|
27
|
May-Simera H, Nagel-Wolfrum K, Wolfrum U. Cilia - The sensory antennae in the eye. Prog Retin Eye Res 2017; 60:144-180. [PMID: 28504201 DOI: 10.1016/j.preteyeres.2017.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022]
Abstract
Cilia are hair-like projections found on almost all cells in the human body. Originally believed to function merely in motility, the function of solitary non-motile (primary) cilia was long overlooked. Recent research has demonstrated that primary cilia function as signalling hubs that sense environmental cues and are pivotal for organ development and function, tissue hoemoestasis, and maintenance of human health. Cilia share a common anatomy and their diverse functional features are achieved by evolutionarily conserved functional modules, organized into sub-compartments. Defects in these functional modules are responsible for a rapidly growing list of human diseases collectively termed ciliopathies. Ocular pathogenesis is common in virtually all classes of syndromic ciliopathies, and disruptions in cilia genes have been found to be causative in a growing number of non-syndromic retinal dystrophies. This review will address what is currently known about cilia contribution to visual function. We will focus on the molecular and cellular functions of ciliary proteins and their role in the photoreceptor sensory cilia and their visual phenotypes. We also highlight other ciliated cell types in tissues of the eye (e.g. lens, RPE and Müller glia cells) discussing their possible contribution to disease progression. Progress in basic research on the cilia function in the eye is paving the way for therapeutic options for retinal ciliopathies. In the final section we describe the latest advancements in gene therapy, read-through of non-sense mutations and stem cell therapy, all being adopted to treat cilia dysfunction in the retina.
Collapse
Affiliation(s)
- Helen May-Simera
- Institute of Molecular Physiology, Cilia Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany.
| |
Collapse
|
28
|
Bhattacharyya N, Darren B, Schott RK, Tropepe V, Chang BSW. Cone-like rhodopsin expressed in the all cone retina of the colubrid pine snake as a potential adaptation to diurnality. J Exp Biol 2017; 220:2418-2425. [DOI: 10.1242/jeb.156430] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/13/2017] [Indexed: 12/12/2022]
Abstract
Colubridae is the largest and most diverse family of snakes, with visual systems that reflect this diversity, encompassing a variety of retinal photoreceptor organizations. The transmutation theory proposed by Walls postulates that photoreceptors could evolutionarily transition between cell types in squamates, but few studies have tested this theory. Recently, evidence for transmutation and rod-like machinery in an all cone retina has been identified in a diurnal garter snake (Thamnophis), and it appears that the rhodopsin gene at least may be widespread among colubrid snakes. However, functional evidence supporting transmutation beyond the existence of the rhodopsin gene remains rare. We examined the all cone retina of another colubrid, Pituophis melanoleucus, thought to be more secretive/burrowing than Thamnophis. We found that P. melanoleucus expresses two cone opsins (SWS1, LWS) and rhodopsin (RH1) within the eye. Immunohistochemistry localized rhodopsin to the outer segment of photoreceptors in the all-cone retina of the snake and all opsin genes produced functional visual pigments when expressed in vitro. Consistent with other studies, we found that P. melanoleucus rhodopsin is extremely blue-shifted. Surprisingly, P. melanoleucus rhodopsin reacted with hydroxylamine, a typical cone opsin characteristic. These results support the idea that the rhodopsin-containing photoreceptors of P. melanoleucus are the products of evolutionary transmutation from rod ancestors, and suggests that this phenomenon may be widespread in colubrid snakes. We hypothesize that transmutation may be an adaptation for diurnal, brighter-light vision, which could result in increased spectral sensitivity and chromatic discrimination with the potential for colour vision.
Collapse
Affiliation(s)
- Nihar Bhattacharyya
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Benedict Darren
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ryan K. Schott
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vincent Tropepe
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto ON, M5T 3A9, Canada
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario, Canada
| | - Belinda S. W. Chang
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Chakraborty D, Conley SM, Pittler SJ, Naash MI. Role of RDS and Rhodopsin in Cngb1-Related Retinal Degeneration. Invest Ophthalmol Vis Sci 2016; 57:787-97. [PMID: 26934134 PMCID: PMC4777275 DOI: 10.1167/iovs.15-18516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Rod photoreceptor outer segment (OS) morphogenesis, structural integrity, and proper signal transduction rely on critical proteins found in the different OS membrane domains (e.g., plasma, disc, and disc rim membrane). Among these key elements are retinal degeneration slow (RDS, also known as peripherin-2), rhodopsin, and the beta subunit of the cyclic nucleotide gated channel (CNGB1a), which have been found to interact in a complex. The purpose of this study was to evaluate the potential interplay between these three proteins by examining retinal disease phenotypes in animal models expressing varying amounts of CNGB1a, rhodopsin, and RDS. Methods Outer segment trafficking, retinal function, and photoreceptor structure were evaluated using knockout mouse lines. Results Eliminating Cngb1 and reducing RDS leads to additive defects in RDS expression levels and rod electroretinogram (ERG) function, (e.g., Cngb1−/−/rds+/− versus rds+/− or Cngb1−/−) but not to additive defects in rod ultrastructure. These additive effects also manifested in cone function: Photopic ERG responses were significantly lower in the Cngb1−/−/rds+/− versus rds+/− or Cngb1−/−, suggesting that eliminating Cngb1 can accelerate the cone degeneration that usually presents later in the rds+/−. This was not the case with rhodopsin; reducing rhodopsin levels in concert with eliminating CNGB1a did not lead to phenotypes more severe than those observed in the Cngb1 knockout alone. Conclusions These data support a role for RDS as the core component of a multiprotein plasma membrane-rim-disc complex that has both a structural role in photoreceptor OS formation and maintenance and a functional role in orienting proteins for optimal signal transduction.
Collapse
Affiliation(s)
- Dibyendu Chakraborty
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Steven J Pittler
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, Texas, United States
| |
Collapse
|
30
|
Goldberg AFX, Moritz OL, Williams DS. Molecular basis for photoreceptor outer segment architecture. Prog Retin Eye Res 2016; 55:52-81. [PMID: 27260426 DOI: 10.1016/j.preteyeres.2016.05.003] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/27/2016] [Accepted: 05/29/2016] [Indexed: 01/11/2023]
Abstract
To serve vision, vertebrate rod and cone photoreceptors must detect photons, convert the light stimuli into cellular signals, and then convey the encoded information to downstream neurons. Rods and cones are sensory neurons that each rely on specialized ciliary organelles to detect light. These organelles, called outer segments, possess elaborate architectures that include many hundreds of light-sensitive membranous disks arrayed one atop another in precise register. These stacked disks capture light and initiate the chain of molecular and cellular events that underlie normal vision. Outer segment organization is challenged by an inherently dynamic nature; these organelles are subject to a renewal process that replaces a significant fraction of their disks (up to ∼10%) on a daily basis. In addition, a broad range of environmental and genetic insults can disrupt outer segment morphology to impair photoreceptor function and viability. In this chapter, we survey the major progress that has been made for understanding the molecular basis of outer segment architecture. We also discuss key aspects of organelle lipid and protein composition, and highlight distributions, interactions, and potential structural functions of key OS-resident molecules, including: kinesin-2, actin, RP1, prominin-1, protocadherin 21, peripherin-2/rds, rom-1, glutamic acid-rich proteins, and rhodopsin. Finally, we identify key knowledge gaps and challenges that remain for understanding how normal outer segment architecture is established and maintained.
Collapse
Affiliation(s)
- Andrew F X Goldberg
- Eye Research Institute, Oakland University, 417 Dodge Hall, Rochester, MI, 48309, USA.
| | - Orson L Moritz
- Department of Ophthalmology & Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - David S Williams
- Department of Ophthalmology and Jules Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| |
Collapse
|
31
|
Evolutionary transformation of rod photoreceptors in the all-cone retina of a diurnal garter snake. Proc Natl Acad Sci U S A 2015; 113:356-61. [PMID: 26715746 DOI: 10.1073/pnas.1513284113] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Vertebrate retinas are generally composed of rod (dim-light) and cone (bright-light) photoreceptors with distinct morphologies that evolved as adaptations to nocturnal/crepuscular and diurnal light environments. Over 70 years ago, the "transmutation" theory was proposed to explain some of the rare exceptions in which a photoreceptor type is missing, suggesting that photoreceptors could evolutionarily transition between cell types. Although studies have shown support for this theory in nocturnal geckos, the origins of all-cone retinas, such as those found in diurnal colubrid snakes, remain a mystery. Here we investigate the evolutionary fate of the rods in a diurnal garter snake and test two competing hypotheses: (i) that the rods, and their corresponding molecular machinery, were lost or (ii) that the rods were evolutionarily modified to resemble, and function, as cones. Using multiple approaches, we find evidence for a functional and unusually blue-shifted rhodopsin that is expressed in small single "cones." Moreover, these cones express rod transducin and have rod ultrastructural features, providing strong support for the hypothesis that they are not true cones, as previously thought, but rather are modified rods. Several intriguing features of garter snake rhodopsin are suggestive of a more cone-like function. We propose that these cone-like rods may have evolved to regain spectral sensitivity and chromatic discrimination as a result of ancestral losses of middle-wavelength cone opsins in early snake evolution. This study illustrates how sensory evolution can be shaped not only by environmental constraints but also by historical contingency in forming new cell types with convergent functionality.
Collapse
|
32
|
Nemet I, Tian G, Imanishi Y. Organization of cGMP sensing structures on the rod photoreceptor outer segment plasma membrane. Channels (Austin) 2015; 8:528-35. [PMID: 25616687 DOI: 10.4161/19336950.2014.973776] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A diffusion barrier segregates the plasma membrane of the rod photoreceptor outer segment into 2 domains; one which is optimized for the conductance of ions in the phototransduction cascade and another for disk membrane synthesis. We propose the former to be named "phototransductive plasma membrane domain," and the latter to be named "disk morphogenic plasma membrane domain." Within the phototransductive plasma membrane, cGMP-gated channels are concentrated in striated membrane features, which are proximally located to the sites of active cGMP production within the disk membranes. For proper localization of cGMP-gated channel to the phototransductive plasma membrane, the glutamic acid-rich protein domain encoded in the β subunit plays a critical role. Quantitative study suggests that the disk morphogenic domain likely plays an important role in enriching rhodopsin prior to its sequestration into closed disk membranes. Thus, this and our previous studies provide new insight into the mechanism that spatially organizes the vertebrate phototransduction cascade.
Collapse
Key Words
- CNGA1
- CNGA1, cyclic nucleotide gated channel α-1
- CNGB1
- CNGB1, cyclic nucleotide gated channel β-1
- Dend2, Dendra2
- GARP, glutamic acid-rich protein
- GC, guanylate cyclase
- GCAP, guanylate cyclase activating protein
- GPCR, G protein-coupled receptor
- IS, inner segment
- OS, outer segment
- PDE6, phosphodiesterase 6
- Rho, rhodopsin
- cyclic nucleotide gated channel
- morphogenesis
- photoreceptor
- retina
- rhodopsin
Collapse
Affiliation(s)
- Ina Nemet
- a Department of Pharmacology; School of Medicine ; Case Western Reserve University ; Cleveland , OH USA
| | | | | |
Collapse
|
33
|
Rakshit T, Park PSH. Impact of reduced rhodopsin expression on the structure of rod outer segment disc membranes. Biochemistry 2015; 54:2885-94. [PMID: 25881629 DOI: 10.1021/acs.biochem.5b00003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rhodopsin is the light receptor embedded in rod outer segment (ROS) disc membranes of photoreceptor cells that initiates vision via phototransduction. The relationship between rhodopsin expression and the formation of membrane structures in the ROS is unclear but important to better understand both normal function and pathological conditions. To determine the impact of reduced rhodopsin expression on the structure of ROS discs and the supramolecular organization of rhodopsin, ROS disc membrane samples from heterozygous rhodopsin knockout mice were examined by atomic force microscopy. Similar to rhodopsin in wild-type mice, rhodopsin formed nanodomains in ROS disc membranes of heterozygous knockout mice. The reduced rhodopsin expression in heterozygous knockout mice resulted in ROS disc membranes that were smaller compared to those in wild-type mice at all ages tested. Changes in ROS disc membrane properties were observed between 4 and 6 weeks of age in heterozygous knockout mice that were not present in age-matched wild-type mice. In 4 week old mice, the number and density of rhodopsin in ROS disc membranes was lower than that in age-matched wild-type mice. In contrast, 6 and 8 week old mice had more rhodopsin molecules present in disc membranes compared to 4 week old mice, which resulted in rhodopsin densities similar to those found in age-matched wild-type mice. Thus, mechanisms appear to be present that maintain a constant density of rhodopsin within ROS disc membranes even when reducing the expression of the light receptor by about half. These adaptive mechanisms, however, only occur after 4 weeks of age.
Collapse
Affiliation(s)
- Tatini Rakshit
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
34
|
Azevedo AW, Doan T, Moaven H, Sokal I, Baameur F, Vishnivetskiy SA, Homan KT, Tesmer JJG, Gurevich VV, Chen J, Rieke F. C-terminal threonines and serines play distinct roles in the desensitization of rhodopsin, a G protein-coupled receptor. eLife 2015; 4. [PMID: 25910054 PMCID: PMC4438306 DOI: 10.7554/elife.05981] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/23/2015] [Indexed: 12/31/2022] Open
Abstract
Rod photoreceptors generate measurable responses to single-photon activation of individual molecules of the G protein-coupled receptor (GPCR), rhodopsin. Timely rhodopsin desensitization depends on phosphorylation and arrestin binding, which quenches G protein activation. Rhodopsin phosphorylation has been measured biochemically at C-terminal serine residues, suggesting that these residues are critical for producing fast, low-noise responses. The role of native threonine residues is unclear. We compared single-photon responses from rhodopsin lacking native serine or threonine phosphorylation sites. Contrary to expectation, serine-only rhodopsin generated prolonged step-like single-photon responses that terminated abruptly and randomly, whereas threonine-only rhodopsin generated responses that were only modestly slower than normal. We show that the step-like responses of serine-only rhodopsin reflect slow and stochastic arrestin binding. Thus, threonine sites play a privileged role in promoting timely arrestin binding and rhodopsin desensitization. Similar coordination of phosphorylation and arrestin binding may more generally permit tight control of the duration of GPCR activity. DOI:http://dx.doi.org/10.7554/eLife.05981.001 ‘Rod’ cells in the eye enable us to see in starlight. Inside these cells, a protein called rhodopsin is activated by light, which leads to an electrical signal being produced that travels to the brain. The duration of the electrical signal depends on the time it takes for the rhodopsin to be deactivated. Rhodopsin is a member of a large class of receptor proteins known as G protein-coupled receptors that regulate many processes throughout the body. Previous studies have shown that rhodopsin is deactivated by the attachment of phosphate groups to the protein. This allows another protein called arrestin to bind to rhodopsin. The phosphates can be attached to particular amino acids—the building blocks of proteins—at one end of rhodopsin. Three of these are a type of amino acid called serine. Previous work has shown that light increases the speed at which phosphate groups are added to these serines, suggesting that they are important for producing rapid electrical signals. The other three amino acids are of a different type—called threonine—but it is less clear what role they play in deactivating rhodopsin. Here, Azevedo et al. studied mutant forms of rhodopsin that were missing the serines or threonines in mice. The experiments show that loss of the serines only slightly slowed the electrical signals. However, loss of the threonines resulted in much slower electrical signals that ended at random times. This was due to rhodopsin being less able to bind to arrestin. Azevedo et al. propose a new model for how rhodopsin is deactivated. Once light activates the protein, phosphate groups are rapidly added to the serines, which begins to lower the activity of rhodopsin. However, it is the slower addition of phosphates to the threonines that is essential to promote arrestin binding and fully deactivate the protein. Other proteins belonging to the G protein-coupled receptor family also have these serines and threonines, and thus, may be regulated in a similar way. DOI:http://dx.doi.org/10.7554/eLife.05981.002
Collapse
Affiliation(s)
- Anthony W Azevedo
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Thuy Doan
- Department of Ophthalmology, University of Washington, Seattle, United States
| | - Hormoz Moaven
- Departments of Cell & Neurobiology and Ophthalmology, Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, United States
| | - Iza Sokal
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Faiza Baameur
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, United States
| | - Sergey A Vishnivetskiy
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, United States
| | - Kristoff T Homan
- Life Sciences Institute, Departments of Pharmacology and Biological Chemistry, University of Michigan, Ann Arbor, United States
| | - John J G Tesmer
- Life Sciences Institute, Departments of Pharmacology and Biological Chemistry, University of Michigan, Ann Arbor, United States
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, United States
| | - Jeannie Chen
- Departments of Cell & Neurobiology and Ophthalmology, Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, United States
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| |
Collapse
|
35
|
Gunkel M, Schöneberg J, Alkhaldi W, Irsen S, Noé F, Kaupp UB, Al-Amoudi A. Higher-order architecture of rhodopsin in intact photoreceptors and its implication for phototransduction kinetics. Structure 2015; 23:628-38. [PMID: 25728926 DOI: 10.1016/j.str.2015.01.015] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/16/2015] [Accepted: 01/22/2015] [Indexed: 12/23/2022]
Abstract
The visual pigment rhodopsin belongs to the family of G protein-coupled receptors that can form higher oligomers. It is controversial whether rhodopsin forms oligomers and whether oligomers are functionally relevant. Here, we study rhodopsin organization in cryosections of dark-adapted mouse rod photoreceptors by cryoelectron tomography. We identify four hierarchical levels of organization. Rhodopsin forms dimers; at least ten dimers form a row. Rows form pairs (tracks) that are aligned parallel to the disk incisures. Particle-based simulation shows that the combination of tracks with fast precomplex formation, i.e. rapid association and dissociation between inactive rhodopsin and the G protein transducin, leads to kinetic trapping: rhodopsin first activates transducin from its own track, whereas recruitment of transducin from other tracks proceeds more slowly. The trap mechanism could produce uniform single-photon responses independent of rhodopsin lifetime. In general, tracks might provide a platform that coordinates the spatiotemporal interaction of signaling molecules.
Collapse
Affiliation(s)
- Monika Gunkel
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Johannes Schöneberg
- Computational Molecular Biology Group, Freie Universität Berlin, Arnimallee 6, 14195 Berlin, Germany
| | - Weaam Alkhaldi
- German Center of Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Stephan Irsen
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Frank Noé
- Computational Molecular Biology Group, Freie Universität Berlin, Arnimallee 6, 14195 Berlin, Germany
| | - U Benjamin Kaupp
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| | - Ashraf Al-Amoudi
- German Center of Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| |
Collapse
|
36
|
Sarfare S, McKeown AS, Messinger J, Rubin G, Wei H, Kraft TW, Pittler SJ. Overexpression of rod photoreceptor glutamic acid rich protein 2 (GARP2) increases gain and slows recovery in mouse retina. Cell Commun Signal 2014; 12:67. [PMID: 25323447 PMCID: PMC4207353 DOI: 10.1186/s12964-014-0067-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/02/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The rod photoreceptor cGMP-gated cation channel, consisting of three α- and one β subunit, controls ion flow into the rod outer segment (ROS). In addition to the β-subunit, the Cngb1 locus encodes an abundant soluble protein, GARP2 that binds stoichiometrically to rod photoreceptor cGMP phosphodiesterase type 6 (PDE6). To examine the in vivo functional role of GARP2 we generated opsin promoter-driven transgenic mice overexpressing GARP2 three-fold specifically in rod photoreceptors. RESULTS In the GARP2 overexpressing transgenic mice (tg), the endogenous channel β-subunit, cGMP phosphodiesterase α-subunit, peripherin2/RDS and guanylate cyclase I were present at WT levels and were properly localized within the ROS. While localized properly within ROS, two proteins cGMP phosphodiesterase α-subunit (1.4-fold) and cGMP-gated cation channel α-subunit (1.2-fold) were moderately, but significantly elevated. Normal stratification of all retinal layers was observed, and ROS were stable in numbers but were 19% shorter than WT. Analysis of the photoresponse using electroretinography (ERG) showed that tg mice exhibit no change in sensitivity indicating overall normal rod function, however two parameters of the photoresponse significantly differed from WT responses. Fitting of the rising phase of the ERG a-wave to an accepted model of phototransduction showed a two-fold increase in phototransduction gain in the tg mice. The increase in gain was confirmed in isolated retinal tissue and by suction electrode recordings of individual rod photoreceptor cells. A measure of response recovery, the dominant time constant (τD) was elevated 69% in isolated retina compared to WT, indicating slower shutoff of the photoresponse. CONCLUSIONS GARP2 may participate in regulating visual signal transduction through a previously unappreciated role in regulating phototransduction gain and recovery.
Collapse
|
37
|
Whited AM, Park PSH. Nanodomain organization of rhodopsin in native human and murine rod outer segment disc membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:26-34. [PMID: 25305340 DOI: 10.1016/j.bbamem.2014.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/25/2014] [Accepted: 10/01/2014] [Indexed: 01/31/2023]
Abstract
Biological membranes display distinct domains that organize membrane proteins and signaling molecules to facilitate efficient and reliable signaling. The organization of rhodopsin, a G protein-coupled receptor, in native rod outer segment disc membranes was investigated by atomic force microscopy. Atomic force microscopy revealed that rhodopsin is arranged into domains of variable size, which we refer to herein as nanodomains, in native membranes. Quantitative analysis of 150 disc membranes revealed that the physical properties of nanodomains are conserved in humans and mice and that the properties of individual disc membranes can be variable. Examining the variable properties of disc membranes revealed some of the factors contributing to the size of rod outer segment discs and the formation of nanodomains in the membrane. The diameter of rod outer segment discs was dependent on the number of rhodopsin molecules incorporated into the membrane but independent of the spatial density of rhodopsin. The number of nanodomains present in a single disc was also dependent on the number of rhodopsin molecules incorporated into the membrane. The size of the nanodomains was largely independent of the number or spatial density of rhodopsin in the membrane.
Collapse
Affiliation(s)
- Allison M Whited
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
38
|
Chakraborty D, Conley SM, Al-Ubaidi MR, Naash MI. Initiation of rod outer segment disc formation requires RDS. PLoS One 2014; 9:e98939. [PMID: 24897172 PMCID: PMC4045911 DOI: 10.1371/journal.pone.0098939] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/08/2014] [Indexed: 11/23/2022] Open
Abstract
Rod outer segment (OS) morphogenesis involves assembly of flattened discs circumscribed by a hairpin-like rim, however, the role of the rim and rim proteins such as retinal degeneration slow (RDS) and its homologue rod OS membrane protein-1 (ROM-1) in this process remains unclear. Here we show that without RDS, no disc/OS formation occurs, while without rhodopsin, small OS structures form containing aligned nascent discs. In the absence of both rhodopsin and RDS, RDS-associated degeneration is slowed, and ROM-1 is stabilized and trafficked to the OS. These animals (rho-/-/rds-/-) exhibit OSs slightly better than those lacking only RDS, but still without signs of disc formation. These results clearly demonstrate that OS morphogenesis is initiated by RDS-mediated rim formation, a process ROM-1 cannot recapitulate, with subsequent disc growth mediated by rhodopsin. The critical role of RDS in this process helps explain why photoreceptors are so sensitive to varied RDS levels, and why mutations in RDS cause debilitating retinal disease.
Collapse
Affiliation(s)
- Dibyendu Chakraborty
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Muayyad R. Al-Ubaidi
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Muna I. Naash
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
39
|
Wen XH, Dizhoor AM, Makino CL. Membrane guanylyl cyclase complexes shape the photoresponses of retinal rods and cones. Front Mol Neurosci 2014; 7:45. [PMID: 24917784 PMCID: PMC4040495 DOI: 10.3389/fnmol.2014.00045] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/01/2014] [Indexed: 12/02/2022] Open
Abstract
In vertebrate rods and cones, photon capture by rhodopsin leads to the destruction of cyclic GMP (cGMP) and the subsequent closure of cyclic nucleotide gated ion channels in the outer segment plasma membrane. Replenishment of cGMP and reopening of the channels limit the growth of the photon response and are requisite for its recovery. In different vertebrate retinas, there may be as many as four types of membrane guanylyl cyclases (GCs) for cGMP synthesis. Ten neuronal Ca2+ sensor proteins could potentially modulate their activities. The mouse is proving to be an effective model for characterizing the roles of individual components because its relative simplicity can be reduced further by genetic engineering. There are two types of GC activating proteins (GCAPs) and two types of GCs in mouse rods, whereas cones express one type of GCAP and one type of GC. Mutant mouse rods and cones bereft of both GCAPs have large, long lasting photon responses. Thus, GCAPs normally mediate negative feedback tied to the light-induced decline in intracellular Ca2+ that accelerates GC activity to curtail the growth and duration of the photon response. Rods from other mutant mice that express a single GCAP type reveal how the two GCAPs normally work together as a team. Because of its lower Ca2+ affinity, GCAP1 is the first responder that senses the initial decrease in Ca2+ following photon absorption and acts to limit response amplitude. GCAP2, with a higher Ca2+ affinity, is recruited later during the course of the photon response as Ca2+ levels continue to decline further. The main role of GCAP2 is to provide for a timely response recovery and it is particularly important after exposure to very bright light. The multiplicity of GC isozymes and GCAP homologs in the retinas of other vertebrates confers greater flexibility in shaping the photon responses in order to tune visual sensitivity, dynamic range and frequency response.
Collapse
Affiliation(s)
- Xiao-Hong Wen
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary and Harvard Medical School Boston, MA, USA
| | - Alexander M Dizhoor
- Department of Basic Sciences Research and Pennsylvania College of Optometry, Salus University Elkins Park, PA, USA
| | - Clint L Makino
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary and Harvard Medical School Boston, MA, USA
| |
Collapse
|
40
|
Cideciyan AV, Hufnagel RB, Carroll J, Sumaroka A, Luo X, Schwartz SB, Dubra A, Land M, Michaelides M, Gardner JC, Hardcastle AJ, Moore AT, Sisk RA, Ahmed ZM, Kohl S, Wissinger B, Jacobson SG. Human cone visual pigment deletions spare sufficient photoreceptors to warrant gene therapy. Hum Gene Ther 2013; 24:993-1006. [PMID: 24067079 DOI: 10.1089/hum.2013.153] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human X-linked blue-cone monochromacy (BCM), a disabling congenital visual disorder of cone photoreceptors, is a candidate disease for gene augmentation therapy. BCM is caused by either mutations in the red (OPN1LW) and green (OPN1MW) cone photoreceptor opsin gene array or large deletions encompassing portions of the gene array and upstream regulatory sequences that would predict a lack of red or green opsin expression. The fate of opsin-deficient cone cells is unknown. We know that rod opsin null mutant mice show rapid postnatal death of rod photoreceptors. Using in vivo histology with high-resolution retinal imaging, we studied a cohort of 20 BCM patients (age range 5-58) with large deletions in the red/green opsin gene array. Already in the first years of life, retinal structure was not normal: there was partial loss of photoreceptors across the central retina. Remaining cone cells had detectable outer segments that were abnormally shortened. Adaptive optics imaging confirmed the existence of inner segments at a spatial density greater than that expected for the residual blue cones. The evidence indicates that human cones in patients with deletions in the red/green opsin gene array can survive in reduced numbers with limited outer segment material, suggesting potential value of gene therapy for BCM.
Collapse
Affiliation(s)
- Artur V Cideciyan
- 1 Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA 19104
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Prokai-Tatrai K, Xin H, Nguyen V, Szarka S, Blazics B, Prokai L, Koulen P. 17β-estradiol eye drops protect the retinal ganglion cell layer and preserve visual function in an in vivo model of glaucoma. Mol Pharm 2013; 10:3253-61. [PMID: 23841874 DOI: 10.1021/mp400313u] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neuroprotection in glaucoma as a curative strategy complementary to current therapies to lower intraocular pressure (IOP) is highly desirable. This study was designed to investigate neuroprotection by 17β-estradiol (E2) to prevent retinal ganglion cell (RGC) death in a glaucoma model of surgically elevated IOP in rats. We found that daily treatment with E2-containing eye drops resulted in significant E2 concentration in the retina with concomitant profound neuroprotective therapeutic benefits, even in the presence of continually elevated IOP. The number of apoptotic cells in the RGC layer was significantly decreased in the E2-treated group, when compared to the vehicle-treated controls. Deterioration in visual acuity in these animals was also markedly prevented. Using mass spectrometry-based proteomics, beneficial changes in the expression of several proteins implicated in the maintenance of retinal health were also found in the retina of E2-treated animals. On the other hand, systemic side effects could not be avoided with the eye drops, as confirmed by the measured high circulating estrogen levels and through the assessment of the uterus representing a typical hormone-sensitive peripheral organ. Collectively, the demonstrated significant neuroprotective effect of topical E2 in the selected animal model of glaucoma provides a clear rationale for further studies aiming at targeting E2 into the eye while avoiding systemic E2 exposure to diminish undesirable off-target side effects.
Collapse
Affiliation(s)
- Katalin Prokai-Tatrai
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center , 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | | | | | | | | | | | | |
Collapse
|
42
|
Pearring JN, Salinas RY, Baker SA, Arshavsky VY. Protein sorting, targeting and trafficking in photoreceptor cells. Prog Retin Eye Res 2013; 36:24-51. [PMID: 23562855 DOI: 10.1016/j.preteyeres.2013.03.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/22/2013] [Accepted: 03/26/2013] [Indexed: 01/24/2023]
Abstract
Vision is the most fundamental of our senses initiated when photons are absorbed by the rod and cone photoreceptor neurons of the retina. At the distal end of each photoreceptor resides a light-sensing organelle, called the outer segment, which is a modified primary cilium highly enriched with proteins involved in visual signal transduction. At the proximal end, each photoreceptor has a synaptic terminal, which connects this cell to the downstream neurons for further processing of the visual information. Understanding the mechanisms involved in creating and maintaining functional compartmentalization of photoreceptor cells remains among the most fascinating topics in ocular cell biology. This review will discuss how photoreceptor compartmentalization is supported by protein sorting, targeting and trafficking, with an emphasis on the best-studied cases of outer segment-resident proteins.
Collapse
Affiliation(s)
- Jillian N Pearring
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Photoreceptors are exquisitely adapted to transform light stimuli into electrical signals that modulate neurotransmitter release. These cells are organized into several compartments including the unique outer segment (OS). Its whole function is to absorb light and transduce this signal into a change of membrane potential. Another compartment is the inner segment where much of metabolism and regulation of membrane potential takes place and that connects the OS and synapse. The synapse is the compartment where changes in membrane potentials are relayed to other neurons in the retina via release of neurotransmitter. The composition of the plasma membrane surrounding these compartments varies to accommodate their specific functions. In this chapter, we discuss the organization of the plasma membrane emphasizing the protein composition of each region as it relates to visual signaling. We also point out examples where mutations in these proteins cause visual impairment.
Collapse
Affiliation(s)
- Sheila A Baker
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| | | |
Collapse
|
44
|
Price BA, Sandoval IM, Chan F, Nichols R, Roman-Sanchez R, Wensel TG, Wilson JH. Rhodopsin gene expression determines rod outer segment size and rod cell resistance to a dominant-negative neurodegeneration mutant. PLoS One 2012; 7:e49889. [PMID: 23185477 PMCID: PMC3503812 DOI: 10.1371/journal.pone.0049889] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 10/17/2012] [Indexed: 01/26/2023] Open
Abstract
Two outstanding unknowns in the biology of photoreceptors are the molecular determinants of cell size, which is remarkably uniform among mammalian species, and the mechanisms of rod cell death associated with inherited neurodegenerative blinding diseases such as retinitis pigmentosa. We have addressed both questions by performing an in vivo titration with rhodopsin gene copies in genetically engineered mice that express only normal rhodopsin or an autosomal dominant allele, encoding rhodopsin with a disease-causing P23H substitution. The results reveal that the volume of the rod outer segment is proportional to rhodopsin gene expression; that P23H-rhodopsin, the most common rhodopsin gene disease allele, causes cell death via a dominant-negative mechanism; and that long term survival of rod cells carrying P23H-rhodopsin can be achieved by increasing the levels of wild type rhodopsin. These results point to promising directions in gene therapy for autosomal dominant neurodegenerative diseases caused by dominant-negative mutations.
Collapse
Affiliation(s)
- Brandee A. Price
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ivette M. Sandoval
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fung Chan
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ralph Nichols
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ramon Roman-Sanchez
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Graduate Program in Cellular and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Theodore G. Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States of America
- Graduate Program in Cellular and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - John H. Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Graduate Program in Cellular and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|