1
|
Hong JD, Salom D, Choi EH, Du SW, Tworak A, Smidak R, Gao F, Solano YJ, Zhang J, Kiser PD, Palczewski K. Retinylidene chromophore hydrolysis from mammalian visual and non-visual opsins. J Biol Chem 2024; 300:105678. [PMID: 38272218 PMCID: PMC10877631 DOI: 10.1016/j.jbc.2024.105678] [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: 11/30/2023] [Revised: 01/06/2024] [Accepted: 01/13/2024] [Indexed: 01/27/2024] Open
Abstract
Rhodopsin (Rho) and cone opsins are essential for detection of light. They respond via photoisomerization, converting their Schiff-base-adducted 11-cis-retinylidene chromophores to the all-trans configuration, eliciting conformational changes to activate opsin signaling. Subsequent Schiff-base hydrolysis releases all-trans-retinal, initiating two important cycles that maintain continuous vision-the Rho photocycle and visual cycle pathway. Schiff-base hydrolysis has been thoroughly studied with photoactivated Rho but not with cone opsins. Using established methodology, we directly measured the formation of Schiff-base between retinal chromophores with mammalian visual and nonvisual opsins of the eye. Next, we determined the rate of light-induced chromophore hydrolysis. We found that retinal hydrolysis from photoactivated cone opsins was markedly faster than from photoactivated Rho. Bovine retinal G protein-coupled receptor (bRGR) displayed rapid hydrolysis of its 11-cis-retinylidene photoproduct to quickly supply 11-cis-retinal and re-bind all-trans-retinal. Hydrolysis within bRGR in native retinal pigment epithelium microsomal membranes was >6-times faster than that of bRGR purified in detergent micelles. N-terminal-targeted antibodies significantly slowed bRGR hydrolysis, while C-terminal antibodies had no effect. Our study highlights the much faster photocycle of cone opsins relative to Rho and the crucial role of RGR in chromophore recycling in daylight. By contrast, in our experimental conditions, bovine peropsin did not form pigment in the presence of all-trans-retinal nor with any mono-cis retinal isomers, leaving uncertain the role of this opsin as a light sensor.
Collapse
Affiliation(s)
- John D Hong
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA; Department of Chemistry, University of California Irvine, Irvine, California, USA
| | - David Salom
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA.
| | - Elliot H Choi
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - Samuel W Du
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California Irvine, Irvine, California, USA
| | - Aleksander Tworak
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - Roman Smidak
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - Fangyuan Gao
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - Yasmeen J Solano
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California Irvine, Irvine, California, USA
| | - Jianye Zhang
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - Philip D Kiser
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California Irvine, Irvine, California, USA; Department of Clinical Pharmacy Practice, University of California Irvine, Irvine, California, USA; Research Service, VA Long Beach Healthcare System, Long Beach, California, USA
| | - Krzysztof Palczewski
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA; Department of Chemistry, University of California Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California Irvine, Irvine, California, USA; Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA.
| |
Collapse
|
2
|
Rodgers J, Wright P, Ballister ER, Hughes RB, Storchi R, Wynne J, Martial FP, Lucas RJ. Modulating signalling lifetime to optimise a prototypical animal opsin for optogenetic applications. Pflugers Arch 2023; 475:1387-1407. [PMID: 38036775 PMCID: PMC10730688 DOI: 10.1007/s00424-023-02879-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
Animal opsins are light activated G-protein-coupled receptors, capable of optogenetic control of G-protein signalling for research or therapeutic applications. Animal opsins offer excellent photosensitivity, but their temporal resolution can be limited by long photoresponse duration when expressed outside their native cellular environment. Here, we explore methods for addressing this limitation for a prototypical animal opsin (human rod opsin) in HEK293T cells. We find that the application of the canonical rhodopsin kinase (GRK1)/visual arrestin signal termination mechanism to this problem is complicated by a generalised suppressive effect of GRK1 expression. This attenuation can be overcome using phosphorylation-independent mutants of arrestin, especially when these are tethered to the opsin protein. We further show that point mutations targeting the Schiff base stability of the opsin can also reduce signalling lifetime. Finally, we apply one such mutation (E122Q) to improve the temporal fidelity of restored visual responses following ectopic opsin expression in the inner retina of a mouse model of retinal degeneration (rd1). Our results reveal that these two strategies (targeting either arrestin binding or Schiff-base hydrolysis) can produce more time-delimited opsin signalling under heterologous expression and establish the potential of this approach to improve optogenetic performance.
Collapse
Affiliation(s)
- Jessica Rodgers
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| | - Phillip Wright
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Edward R Ballister
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, 10032, NY, USA
| | - Rebecca B Hughes
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Riccardo Storchi
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Jonathan Wynne
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Franck P Martial
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Robert J Lucas
- Centre for Biological Timing, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
3
|
Hanna K, Nieves J, Dowd C, Bender KO, Sharma P, Singh B, Renz M, Ver Hoeve JN, Cepeda D, Gelfman CM, Riley BE, Grishanin RN. Preclinical evaluation of ADVM-062, a novel intravitreal gene therapy vector for the treatment of blue cone monochromacy. Mol Ther 2023; 31:2014-2027. [PMID: 36932675 PMCID: PMC10362383 DOI: 10.1016/j.ymthe.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/14/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Blue cone monochromacy (BCM) is a rare X-linked retinal disease characterized by the absence of L- and M-opsin in cone photoreceptors, considered a potential gene therapy candidate. However, most experimental ocular gene therapies utilize subretinal vector injection which would pose a risk to the fragile central retinal structure of BCM patients. Here we describe the use of ADVM-062, a vector optimized for cone-specific expression of human L-opsin and administered using a single intravitreal (IVT) injection. Pharmacological activity of ADVM-062 was established in gerbils, whose cone-rich retina naturally lacks L-opsin. A single IVT administration dose of ADVM-062 effectively transduced gerbil cone photoreceptors and produced a de novo response to long-wavelength stimuli. To identify potential first-in-human doses we evaluated ADVM-062 in non-human primates. Cone-specific expression of ADVM-062 in primates was confirmed using ADVM-062.myc, a vector engineered with the same regulatory elements as ADVM-062. Enumeration of human OPN1LW.myc-positive cones demonstrated that doses ≥3 × 1010 vg/eye resulted in transduction of 18%-85% of foveal cones. A Good Laboratory Practice (GLP) toxicology study established that IVT administration of ADVM-062 was well tolerated at doses that could potentially achieve clinically meaningful effect, thus supporting the potential of ADVM-062 as a one-time IVT gene therapy for BCM.
Collapse
Affiliation(s)
- Kelly Hanna
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | - Julio Nieves
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | - Christine Dowd
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | | | - Pallavi Sharma
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | - Baljit Singh
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | - Mark Renz
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | | | - Diana Cepeda
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA
| | | | - Brigit E Riley
- Adverum Biotechnologies, Inc., Redwood City, CA 94063, USA.
| | | |
Collapse
|
4
|
Daniele LL, Han JY, Samuels IS, Komirisetty R, Mehta N, McCord JL, Yu M, Wang Y, Boesze-Battaglia K, Bell BA, Du J, Peachey NS, Philp NJ. Glucose uptake by GLUT1 in photoreceptors is essential for outer segment renewal and rod photoreceptor survival. FASEB J 2022; 36:e22428. [PMID: 35766190 PMCID: PMC9438481 DOI: 10.1096/fj.202200369r] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/27/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
Photoreceptors consume glucose supplied by the choriocapillaris to support phototransduction and outer segment (OS) renewal. Reduced glucose supply underlies photoreceptor cell death in inherited retinal degeneration and age-related retinal disease. We have previously shown that restricting glucose transport into the outer retina by conditional deletion of Slc2a1 encoding GLUT1 resulted in photoreceptor loss and impaired OS renewal. However, retinal neurons, glia, and the retinal pigment epithelium play specialized, synergistic roles in metabolite supply and exchange, and the cell-specific map of glucose uptake and utilization in the retina is incomplete. In these studies, we conditionally deleted Slc2a1 in a pan-retinal or rod-specific manner to better understand how glucose is utilized in the retina. Using non-invasive ocular imaging, electroretinography, and histochemical and biochemical analyses we show that genetic deletion of Slc2a1 from retinal neurons and Müller glia results in reduced OS growth and progressive rod but not cone photoreceptor cell death. Rhodopsin levels were severely decreased even at postnatal day 20 when OS length was relatively normal. Arrestin levels were not changed suggesting that glucose uptake is required to synthesize membrane glycoproteins. Rod-specific deletion of Slc2a1 resulted in similar changes in OS length and rod photoreceptor cell death. These studies demonstrate that glucose is an essential carbon source for rod photoreceptor cell OS maintenance and viability.
Collapse
Affiliation(s)
- Lauren L. Daniele
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - John Y.S. Han
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Ivy S. Samuels
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Louis Stokes Cleveland VA Medical Center, Cleveland,
OH
| | - Ravikiran Komirisetty
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Nikhil Mehta
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Jessica L. McCord
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Minzhong Yu
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Department of Ophthalmology, Cleveland Clinic Lerner
College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West
Virginia University, Morgantown, WV
- Department of Biochemistry, West Virginia University,
Morgantown, WV
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, Penn Dental
Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brent A. Bell
- Department of Ophthalmology, University of Pennsylvania,
Philadelphia, PA
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West
Virginia University, Morgantown, WV
- Department of Biochemistry, West Virginia University,
Morgantown, WV
| | - Neal S. Peachey
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Louis Stokes Cleveland VA Medical Center, Cleveland,
OH
- Department of Ophthalmology, Cleveland Clinic Lerner
College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Nancy J. Philp
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| |
Collapse
|
5
|
Ma X, Sechrest ER, Fajardo D, Zhu P, Dyka F, Wang Y, Lobanova E, Boye SE, Baehr W, Deng WT. Gene Therapy in Opn1mw-/-/Opn1sw-/- Mice and Implications for Blue Cone Monochromacy Patients with Deletion Mutations. Hum Gene Ther 2022; 33:708-718. [PMID: 35272502 PMCID: PMC9347391 DOI: 10.1089/hum.2021.298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
Blue cone monochromacy (BCM) is a congenital vision disorder affecting both middle-wavelength (M) and long-wavelength (L) cone photoreceptors of the human retina. BCM results from abolished expression of green and red light-sensitive visual pigments expressed in M- and L-cones, respectively. Previously, we showed that gene augmentation therapy to deliver either human L- or M-opsin rescues dorsal M-opsin dominant cone photoreceptors structurally and functionally in treated M-opsin knockout (Opn1mw-/-) mice. Although Opn1mw-/- mice represent a disease model for BCM patients with deletion mutations, at the cellular level, dorsal cones of Opn1mw-/- mice still express low levels of S-opsin, which are different from L- and M-cones of BCM patients carrying a congenital opsin deletion. To determine whether BCM cones lacking complete opsin expression from birth would benefit from AAV-mediated gene therapy, we evaluated the outcome of gene therapy, and determined the therapeutic window and longevity of rescue in a mouse model lacking both M- and S-opsin (Opn1mw-/-/Opn1sw-/-). Our data show that cones of Opn1mw-/-/Opn1sw-/- mice are viable at younger ages but undergo rapid degeneration. AAV-mediated expression of human L-opsin promoted cone outer segment regeneration and rescued cone-mediated function when mice were injected subretinally at 2 months of age or younger. Cone-mediated function and visually guided behavior were maintained for at least 8 months post-treatment. However, when mice were treated at 5 and 7 months of age, the chance and effectiveness of rescue was significantly reduced, although cones were still present in the retina. Crossing Opn1mw-/-/Opn1sw-/- mice with proteasomal activity reporter mice (UbG76V-GFP) did not reveal GFP accumulation in Opn1mw-/-/Opn1sw-/- cones eliminating impaired degradation of ubiquitinated proteins as stress factor contributing to cone loss. Our results demonstrate that AAV-mediated gene augmentation therapy can rescue cone structure and function in a mouse model with a congenital opsin deletion, but also emphasize the importance that early intervention is crucial for successful therapy.
Collapse
Affiliation(s)
- Xiajie Ma
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Emily R. Sechrest
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA
| | - Diego Fajardo
- Division of Cellular and Molecular Therapeutics, Department of Pediatrics; University of Florida, Gainesville, Florida, USA
| | - Ping Zhu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Frank Dyka
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Yixiao Wang
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ekaterina Lobanova
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Shannon E. Boye
- Division of Cellular and Molecular Therapeutics, Department of Pediatrics; University of Florida, Gainesville, Florida, USA
| | - Wolfgang Baehr
- Department of Ophthalmology and Visual Science, University of Utah, Salt Lake City, Utah, USA
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, USA
- Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Wen-Tao Deng
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
6
|
Chen H, Zhang S, Zhang X, Liu H. QR code model: a new possibility for GPCR phosphorylation recognition. Cell Commun Signal 2022; 20:23. [PMID: 35236365 PMCID: PMC8889771 DOI: 10.1186/s12964-022-00832-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/23/2022] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane proteins in the human body and are responsible for accurately transmitting extracellular information to cells. Arrestin is an important member of the GPCR signaling pathway. The main function of arrestin is to assist receptor desensitization, endocytosis and signal transduction. In these processes, the recognition and binding of arrestin to phosphorylated GPCRs is fundamental. However, the mechanism by which arrestin recognizes phosphorylated GPCRs is not fully understood. The GPCR phosphorylation recognition "bar code model" and "flute" model describe the basic process of receptor phosphorylation recognition in terms of receptor phosphorylation sites, arrestin structural changes and downstream signaling. These two models suggest that GPCR phosphorylation recognition is a process involving multiple factors. This process can be described by a "QR code" model in which ligands, GPCRs, G protein-coupled receptor kinase, arrestin, and phosphorylation sites work together to determine the biological functions of phosphorylated receptors. Video Abstract.
Collapse
Affiliation(s)
- Hao Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
7
|
Riedmayr LM, Hinrichsmeyer KS, Karguth N, Böhm S, Splith V, Michalakis S, Becirovic E. dCas9-VPR-mediated transcriptional activation of functionally equivalent genes for gene therapy. Nat Protoc 2022; 17:781-818. [PMID: 35132255 DOI: 10.1038/s41596-021-00666-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022]
Abstract
Many disease-causing genes possess functionally equivalent counterparts, which are often expressed in distinct cell types. An attractive gene therapy approach for inherited disorders caused by mutations in such genes is to transcriptionally activate the appropriate counterpart(s) to compensate for the missing gene function. This approach offers key advantages over conventional gene therapies because it is mutation- and gene size-independent. Here, we describe a protocol for the design, execution and evaluation of such gene therapies using dCas9-VPR. We offer guidelines on how to identify functionally equivalent genes, design and clone single guide RNAs and evaluate transcriptional activation in vitro. Moreover, focusing on inherited retinal diseases, we provide a detailed protocol on how to apply this strategy in mice using dual recombinant adeno-associated virus vectors and how to evaluate its functionality and off-target effects in the target tissue. This strategy is in principle applicable to all organisms that possess functionally equivalent genes suitable for transcriptional activation and addresses pivotal unmet needs in gene therapy with high translational potential. The protocol can be completed in 15-20 weeks.
Collapse
Affiliation(s)
- Lisa M Riedmayr
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Klara S Hinrichsmeyer
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nina Karguth
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sybille Böhm
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Victoria Splith
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elvir Becirovic
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
8
|
Phototransduction in Anuran Green Rods: Origins of Extra-Sensitivity. Int J Mol Sci 2021; 22:ijms222413400. [PMID: 34948198 PMCID: PMC8707487 DOI: 10.3390/ijms222413400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 01/13/2023] Open
Abstract
Green rods (GRs) represent a unique type of photoreceptor to be found in the retinas of anuran amphibians. These cells harbor a cone-specific blue-sensitive visual pigment but exhibit morphology of the outer segment typical for classic red rods (RRs), which makes them a perspective model object for studying cone–rod transmutation. In the present study, we performed detailed electrophysiological examination of the light sensitivity, response kinetics and parameters of discrete and continuous dark noise in GRs of the two anuran species: cane toad and marsh frog. Our results confirm that anuran GRs are highly specialized nocturnal vision receptors. Moreover, their rate of phototransduction quenching appeared to be about two-times slower than in RRs, which makes them even more efficient single photon detectors. The operating intensity ranges for two rod types widely overlap supposedly allowing amphibians to discriminate colors in the scotopic region. Unexpectedly for typical cone pigments but in line with some previous reports, the spontaneous isomerization rate of the GR visual pigment was found to be the same as for rhodopsin of RRs. Thus, our results expand the knowledge on anuran GRs and show that these are even more specialized single photon catchers than RRs, which allows us to assign them a status of “super-rods”.
Collapse
|
9
|
Abbas F, Vinberg F. Transduction and Adaptation Mechanisms in the Cilium or Microvilli of Photoreceptors and Olfactory Receptors From Insects to Humans. Front Cell Neurosci 2021; 15:662453. [PMID: 33867944 PMCID: PMC8046925 DOI: 10.3389/fncel.2021.662453] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Sensing changes in the environment is crucial for survival. Animals from invertebrates to vertebrates use both visual and olfactory stimuli to direct survival behaviors including identification of food sources, finding mates, and predator avoidance. In primary sensory neurons there are signal transduction mechanisms that convert chemical or light signals into an electrical response through ligand binding or photoactivation of a receptor, that can be propagated to the olfactory and visual centers of the brain to create a perception of the odor and visual landscapes surrounding us. The fundamental principles of olfactory and phototransduction pathways within vertebrates are somewhat analogous. Signal transduction in both systems takes place in the ciliary sub-compartments of the sensory cells and relies upon the activation of G protein-coupled receptors (GPCRs) to close cyclic nucleotide-gated (CNG) cation channels in photoreceptors to produce a hyperpolarization of the cell, or in olfactory sensory neurons open CNG channels to produce a depolarization. However, while invertebrate phototransduction also involves GPCRs, invertebrate photoreceptors can be either ciliary and/or microvillar with hyperpolarizing and depolarizing responses to light, respectively. Moreover, olfactory transduction in invertebrates may be a mixture of metabotropic G protein and ionotropic signaling pathways. This review will highlight differences of the visual and olfactory transduction mechanisms between vertebrates and invertebrates, focusing on the implications to the gain of the transduction processes, and how they are modulated to allow detection of small changes in odor concentration and light intensity over a wide range of background stimulus levels.
Collapse
Affiliation(s)
- Fatima Abbas
- Vinberg Lab, Department of Ophthalmology and Visual Science, John A. Moran Center, University of Utah, Salt Lake City, UT, United States
| | - Frans Vinberg
- Vinberg Lab, Department of Ophthalmology and Visual Science, John A. Moran Center, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
10
|
Reingruber J, Ingram NT, Griffis KG, Fain GL. A kinetic analysis of mouse rod and cone photoreceptor responses. J Physiol 2020; 598:3747-3763. [PMID: 32557629 PMCID: PMC7484371 DOI: 10.1113/jp279524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Most vertebrate eyes have rods for dim-light vision and cones for brighter light and higher temporal sensitivity. Rods evolved from cone-like precursors through expression of different transduction genes or the same genes at different expression levels, but we do not know which molecular differences were most important. We approached this problem by analysing rod and cone responses with the same model but with different values for model parameters. We showed that, in addition to outer-segment volume, the most important differences between rods and cones are: (1) decreased transduction gain, reflecting smaller amplification in the G-protein cascade; (2) a faster rate of turnover of the second messenger cGMP in darkness; and (3) an accelerated rate of decay of the effector enzyme phosphodiesterase and perhaps also of activated visual pigment. We believe our analysis has identified the principal alterations during evolution responsible for the duplex retina. ABSTRACT Most vertebrates have rod and cone photoreceptors, which differ in their sensitivity and response kinetics. We know that rods evolved from cone-like precursors through the expression of different transduction genes or the same genes at different levels, but we do not know which molecular differences were most important. We have approached this problem in mouse retina by analysing the kinetic differences between rod flash responses and recent voltage-clamp recordings of cone flash responses, using a model incorporating the principal features of photoreceptor transduction. We apply a novel method of analysis using the log-transform of the current, and we ask which of the model's dynamic parameters need be changed to transform the flash response of a rod into that of a cone. The most important changes are a decrease in the gain of the response, reflecting a reduction in amplification of the transduction cascade; an increase in the rate of turnover of cGMP in darkness; and an increase in the rate of decay of activated phosphodiesterase, with perhaps also an increase in the rate of decay of light-activated visual pigment. Although we cannot exclude other differences, and in particular alterations in the Ca2+ economy of the photoreceptors, we believe that we have identified the kinetic parameters principally responsible for the differences in the flash responses of the two kinds of photoreceptors, which were likely during evolution to have resulted in the duplex retina.
Collapse
Affiliation(s)
- Jürgen Reingruber
- Institut de Biologie de l’École Normale Supérieure, 46 rue d’Ulm, 75005 Paris, France
| | - Norianne T. Ingram
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095–7239, USA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| | - Khris G. Griffis
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| | - Gordon L. Fain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095–7239, USA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| |
Collapse
|
11
|
Böhm S, Splith V, Riedmayr LM, Rötzer RD, Gasparoni G, Nordström KJV, Wagner JE, Hinrichsmeyer KS, Walter J, Wahl-Schott C, Fenske S, Biel M, Michalakis S, Becirovic E. A gene therapy for inherited blindness using dCas9-VPR-mediated transcriptional activation. SCIENCE ADVANCES 2020; 6:eaba5614. [PMID: 32875106 PMCID: PMC7438099 DOI: 10.1126/sciadv.aba5614] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/08/2020] [Indexed: 05/08/2023]
Abstract
Catalytically inactive dCas9 fused to transcriptional activators (dCas9-VPR) enables activation of silent genes. Many disease genes have counterparts, which serve similar functions but are expressed in distinct cell types. One attractive option to compensate for the missing function of a defective gene could be to transcriptionally activate its functionally equivalent counterpart via dCas9-VPR. Key challenges of this approach include the delivery of dCas9-VPR, activation efficiency, long-term expression of the target gene, and adverse effects in vivo. Using dual adeno-associated viral vectors expressing split dCas9-VPR, we show efficient transcriptional activation and long-term expression of cone photoreceptor-specific M-opsin (Opn1mw) in a rhodopsin-deficient mouse model for retinitis pigmentosa. One year after treatment, this approach yields improved retinal function and attenuated retinal degeneration with no apparent adverse effects. Our study demonstrates that dCas9-VPR-mediated transcriptional activation of functionally equivalent genes has great potential for the treatment of genetic disorders.
Collapse
Affiliation(s)
- Sybille Böhm
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Victoria Splith
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lisa Maria Riedmayr
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - René Dominik Rötzer
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | | | - Johanna Elisabeth Wagner
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Klara Sonnie Hinrichsmeyer
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | | | - Stefanie Fenske
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Ophthalmology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elvir Becirovic
- Center for Integrated Protein Science Munich CIPSM, Munich, Germany
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
12
|
Ingram NT, Sampath AP, Fain GL. Voltage-clamp recordings of light responses from wild-type and mutant mouse cone photoreceptors. J Gen Physiol 2019; 151:1287-1299. [PMID: 31562185 PMCID: PMC6829558 DOI: 10.1085/jgp.201912419] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/15/2019] [Accepted: 08/30/2019] [Indexed: 01/16/2023] Open
Abstract
We describe the first extensive study of voltage-clamp current responses of cone photoreceptors in unlabeled, dark-adapted mouse retina using only the position and appearance of cone somata as a guide. Identification was confirmed from morphology after dye filling. Photocurrents recorded from wild-type mouse cones were biphasic with a fast cone component and a slower rod component. The rod component could be eliminated with dim background light and was not present in mouse lines lacking the rod transducin-α subunit (Gnat1-/- ) or connexin 36 (Cx36-/- ). Cones from Gnat1-/- or Cx36-/- mice had resting membrane potentials between -45 and -55 mV, peak photocurrents of 20-25 picoamps (pA) at a membrane potential Vm = -50 mV, sensitivities 60-70 times smaller than rods, and a total membrane capacitance two to four times greater than rods. The rate of activation (amplification constant) was largely independent of the brightness of the flash and was 1-2 s-2, less than half that of rods. The role of Ca2+-dependent transduction modulation was investigated by recording from cones in mice lacking rod transducin (Gnat1), recoverin, and/or the guanylyl-cyclase-activating proteins (GCAPs). In confirmation of previous results, responses of Gnat1-/- ;Gcaps-/- cones and triple-mutant Gnat1-/- ;Gcaps-/- ;Rv-/- cones recovered more slowly both to light flashes and steps and were more sensitive than cones expressing the GCAPs. Cones from all four mouse lines showed significant recovery and escaped saturation even in bright background light. This recovery occurred too rapidly to be caused by pigment bleaching or metaII decay and appears to reflect some modulation of response inactivation in addition to those produced by recoverin and the GCAPs. Our experiments now make possible a more detailed understanding of the cellular physiology of mammalian cone photoreceptors and the role of conductances in the inner and outer segment in producing cone light responses.
Collapse
Affiliation(s)
- Norianne T Ingram
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA
| | - Alapakkam P Sampath
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA
| | - Gordon L Fain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
13
|
Disruption of Rhodopsin Dimerization in Mouse Rod Photoreceptors by Synthetic Peptides Targeting Dimer Interface. Methods Mol Biol 2019; 1753:115-128. [PMID: 29564785 DOI: 10.1007/978-1-4939-7720-8_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Synthetic peptides derived from transmembrane segments of G protein-coupled receptors (GPCR) are used to disrupt GPCR dimer interface. This peptide competition technique is an effective approach to map the dimer interface of GPCR and its functional significance. Here we present a technique to deliver synthetic transmembrane peptides to living mouse rod photoreceptors to disrupt rhodopsin (a prototypical member of Class A GPCRs) dimer formation in the endoplasmic reticulum (ER). We have shown that rhodopsin helix H1- or H8-peptide caused mislocalization of rhodopsin to the perinuclear endoplasmic reticulum (ER).
Collapse
|
14
|
Vinberg F, Kefalov VJ. Investigating the Ca 2+-dependent and Ca 2+-independent mechanisms for mammalian cone light adaptation. Sci Rep 2018; 8:15864. [PMID: 30367097 PMCID: PMC6203770 DOI: 10.1038/s41598-018-34073-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Vision is mediated by two types of photoreceptors: rods, enabling vision in dim light; and cones, which function in bright light. Despite many similarities in the components of their respective phototransduction cascades, rods and cones have distinct sensitivity, response kinetics, and adaptation capacity. Cones are less sensitive and have faster responses than rods. In addition, cones can function over a wide range of light conditions whereas rods saturate in moderately bright light. Calcium plays an important role in regulating phototransduction and light adaptation of rods and cones. Notably, the two dominant Ca2+-feedbacks in rods and cones are driven by the identical calcium-binding proteins: guanylyl cyclase activating proteins 1 and 2 (GCAPs), which upregulate the production of cGMP; and recoverin, which regulates the inactivation of visual pigment. Thus, the mechanisms producing the difference in adaptation capacity between rods and cones have remained poorly understood. Using GCAPs/recoverin-deficient mice, we show that mammalian cones possess another Ca2+-dependent mechanism promoting light adaptation. Surprisingly, we also find that, unlike in mouse rods, a unique Ca2+-independent mechanism contributes to cone light adaptation. Our findings point to two novel adaptation mechanisms in mouse cones that likely contribute to the great adaptation capacity of cones over rods.
Collapse
Affiliation(s)
- Frans Vinberg
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA. .,John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, USA.
| | - Vladimir J Kefalov
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
15
|
Martinecz A, Niitsuma M. Fractional integral-like processing in retinal cones reduces noise and improves adaptation. PLoS One 2018; 13:e0205099. [PMID: 30286168 PMCID: PMC6171915 DOI: 10.1371/journal.pone.0205099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 08/21/2018] [Indexed: 11/17/2022] Open
Abstract
In the human retina, rod and cone cells detect incoming light with a molecule called rhodopsin. After rhodopsin molecules are activated (by photon impact), these molecules activate the rest of the signalling process for a brief period of time until they are deactivated by a multistage process. First, active rhodopsin is phosphorylated multiple times. Following this, they are further inhibited by the binding of molecules called arrestins. Finally, they decay into opsins. The time required for each of these stages becomes progressively longer, and each stage further reduces the activity of rhodopsin. However, while this deactivation process itself is well researched, the roles of the above stages in signal (and image) processing are poorly understood. In this paper, we will show that the activity of rhodopsin molecules during the deactivation process can be described as the fractional integration of an incoming signal. Furthermore, we show how this affects an image; specifically, the effect of fractional integration in video and signal processing and how it reduces noise and the improves adaptability under different lighting conditions. Our experimental results provide a better understanding of vertebrate and human vision, and why the rods and cones of the retina differ from the light detectors in cameras.
Collapse
Affiliation(s)
- Antal Martinecz
- Department of Precision Mechanics, Chuo University, Tokyo, Japan
| | - Mihoko Niitsuma
- Department of Precision Mechanics, Chuo University, Tokyo, Japan
| |
Collapse
|
16
|
Vinberg F, Chen J, Kefalov VJ. Regulation of calcium homeostasis in the outer segments of rod and cone photoreceptors. Prog Retin Eye Res 2018; 67:87-101. [PMID: 29883715 DOI: 10.1016/j.preteyeres.2018.06.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/30/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022]
Abstract
Calcium plays important roles in the function and survival of rod and cone photoreceptor cells. Rapid regulation of calcium in the outer segments of photoreceptors is required for the modulation of phototransduction that drives the termination of the flash response as well as light adaptation in rods and cones. On a slower time scale, maintaining proper calcium homeostasis is critical for the health and survival of photoreceptors. Decades of work have established that the level of calcium in the outer segments of rods and cones is regulated by a dynamic equilibrium between influx via the transduction cGMP-gated channels and extrusion via rod- and cone-specific Na+/Ca2+, K+ exchangers (NCKXs). It had been widely accepted that the only mechanism for extrusion of calcium from rod outer segments is via the rod-specific NCKX1, while extrusion from cone outer segments is driven exclusively by the cone-specific NCKX2. However, recent evidence from mice lacking NCKX1 and NCKX2 have challenged that notion and have revealed a more complex picture, including a NCKX-independent mechanism in rods and two separate NCKX-dependent mechanisms in cones. This review will focus on recent findings on the molecular mechanisms of extrusion of calcium from the outer segments of rod and cone photoreceptors, and the functional and structural changes in photoreceptors when normal extrusion is disrupted.
Collapse
Affiliation(s)
- Frans Vinberg
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA; John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Jeannie Chen
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Vladimir J Kefalov
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
17
|
Morshedian A, Fain GL. The evolution of rod photoreceptors. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0074. [PMID: 28193819 DOI: 10.1098/rstb.2016.0074] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2016] [Indexed: 12/29/2022] Open
Abstract
Photoreceptors in animals are generally of two kinds: the ciliary or c-type and the rhabdomeric or r-type. Although ciliary photoreceptors are found in many phyla, vertebrates seem to be unique in having two distinct kinds which together span the entire range of vision, from single photons to bright light. We ask why the principal photoreceptors of vertebrates are ciliary and not rhabdomeric, and how rods evolved from less sensitive cone-like photoreceptors to produce our duplex retina. We suggest that the principal advantage of vertebrate ciliary receptors is that they use less ATP than rhabdomeric photoreceptors. This difference may have provided sufficient selection pressure for the development of a completely ciliary eye. Although many of the details of rod evolution are still uncertain, present evidence indicates that (i) rods evolved very early before the split between the jawed and jawless vertebrates, (ii) outer-segment discs make no contribution to rod sensitivity but may have evolved to increase the efficiency of protein renewal, and (iii) evolution of the rod was incremental and multifaceted, produced by the formation of several novel protein isoforms and by changes in protein expression, with no one alteration having more than a few-fold effect on transduction activation or inactivation.This article is part of the themed issue 'Vision in dim light'.
Collapse
Affiliation(s)
- Ala Morshedian
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA 90095-7239, USA
| | - Gordon L Fain
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA 90095-7239, USA .,Department of Ophthalmology and Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095-7000, USA
| |
Collapse
|
18
|
Indrischek H, Prohaska SJ, Gurevich VV, Gurevich EV, Stadler PF. Uncovering missing pieces: duplication and deletion history of arrestins in deuterostomes. BMC Evol Biol 2017; 17:163. [PMID: 28683816 PMCID: PMC5501109 DOI: 10.1186/s12862-017-1001-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 06/19/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The cytosolic arrestin proteins mediate desensitization of activated G protein-coupled receptors (GPCRs) via competition with G proteins for the active phosphorylated receptors. Arrestins in active, including receptor-bound, conformation are also transducers of signaling. Therefore, this protein family is an attractive therapeutic target. The signaling outcome is believed to be a result of structural and sequence-dependent interactions of arrestins with GPCRs and other protein partners. Here we elucidated the detailed evolution of arrestins in deuterostomes. RESULTS Identity and number of arrestin paralogs were determined searching deuterostome genomes and gene expression data. In contrast to standard gene prediction methods, our strategy first detects exons situated on different scaffolds and then solves the problem of assigning them to the correct gene. This increases both the completeness and the accuracy of the annotation in comparison to conventional database search strategies applied by the community. The employed strategy enabled us to map in detail the duplication- and deletion history of arrestin paralogs including tandem duplications, pseudogenizations and the formation of retrogenes. The two rounds of whole genome duplications in the vertebrate stem lineage gave rise to four arrestin paralogs. Surprisingly, visual arrestin ARR3 was lost in the mammalian clades Afrotheria and Xenarthra. Duplications in specific clades, on the other hand, must have given rise to new paralogs that show signatures of diversification in functional elements important for receptor binding and phosphate sensing. CONCLUSION The current study traces the functional evolution of deuterostome arrestins in unprecedented detail. Based on a precise re-annotation of the exon-intron structure at nucleotide resolution, we infer the gain and loss of paralogs and patterns of conservation, co-variation and selection.
Collapse
Affiliation(s)
- Henrike Indrischek
- Computational EvoDevo Group, Department of Computer Science, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany.
- Bioinformatics Group, Department of Computer Science, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany.
- Interdisciplinary Center for Bioinformatics, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany.
| | - Sonja J Prohaska
- Computational EvoDevo Group, Department of Computer Science, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany
- Interdisciplinary Center for Bioinformatics, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave, Nashville, TN 37232, USA
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave, Nashville, TN 37232, USA
| | - Peter F Stadler
- Bioinformatics Group, Department of Computer Science, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany
- Interdisciplinary Center for Bioinformatics, Universität Leipzig, Härtelstraße 16-18, Leipzig, D-04107, Germany
- Max Planck Institute for Mathematics in the Sciences, Inselstraße 22, Leipzig, D-04103, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, Leipzig, D-04103, Germany
- Department of Theoretical Chemistry, University of Vienna, Währinger Straße 17, Vienna, A-1090, Austria
- Center for non-coding RNA in Technology and Health, Grønegårdsvej 3, Frederiksberg C, DK-1870, Denmark
- Santa Fe Institute, 1399 Hyde Park Rd., Santa Fe, NM 87501, USA
| |
Collapse
|
19
|
Sato S, Kefalov VJ. cis Retinol oxidation regulates photoreceptor access to the retina visual cycle and cone pigment regeneration. J Physiol 2016; 594:6753-6765. [PMID: 27385534 PMCID: PMC5108915 DOI: 10.1113/jp272831] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/04/2016] [Indexed: 01/21/2023] Open
Abstract
KEY POINTS This study explores the nature of the cis retinol that Müller cells in the retina provide to cones for the regeneration of their visual pigment. We report that the retina visual cycle provides cones exclusively with 11-cis chromophore in both salamander and mouse and show that this selectivity is dependent on the 11-cis-specific cellular retinaldehyde binding protein (CRALBP) present in Müller cells. Even though salamander blue cones and green rods share the same visual pigment, only blue cones but not green rods are able to dark-adapt in the retina following a bleach and to use exogenous 9-cis retinol for pigment regeneration, suggesting that access to the retina visual cycle is cone-specific and pigment-independent. Our results show that the retina produces 11-cis retinol that can be oxidized and used for pigment regeneration and dark adaptation selectively in cones and not in rods. ABSTRACT Chromophore supply by the retinal Müller cells (retina visual cycle) supports the efficient pigment regeneration required for cone photoreceptor function in bright light. Surprisingly, a large fraction of the chromophore produced by dihydroceramide desaturase-1, the putative all-trans retinol isomerase in Müller cells, appears to be 9-cis retinol. In contrast, the canonical retinal pigment epithelium (RPE) visual cycle produces exclusively 11-cis retinal. Here, we used the different absorption spectra of 9-cis and 11-cis pigments to identify the isoform of the chromophore produced by the visual cycle of the intact retina. We found that the spectral sensitivity of salamander and mouse cones dark-adapted in the isolated retina (with only the retina visual cycle) was similar to that of cones dark-adapted in the intact eye (with both the RPE and retina visual cycles) and consistent with pure 11-cis pigment composition. However, in mice lacking the cellular retinaldehyde binding protein (CRALBP), cone spectral sensitivity contained a substantial 9-cis component. Thus, the retina visual cycle provides cones exclusively with 11-cis chromophore and this process is mediated by the 11-cis selective CRALBP in Müller cells. Finally, despite sharing the same pigment, salamander blue cones, but not green rods, recovered their sensitivity in the isolated retina. Exogenous 9-cis retinol produced robust sensitivity recovery in bleached red and blue cones but not in red and green rods, suggesting that cis retinol oxidation restricts access to the retina visual cycle to cones.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Ophthalmology and Visual SciencesWashington University School of MedicineSaint LouisMO63110USA
| | - Vladimir J. Kefalov
- Department of Ophthalmology and Visual SciencesWashington University School of MedicineSaint LouisMO63110USA
| |
Collapse
|
20
|
Zhou Z, Vinberg F, Schottler F, Doggett TA, Kefalov VJ, Ferguson TA. Autophagy supports color vision. Autophagy 2016; 11:1821-32. [PMID: 26292183 DOI: 10.1080/15548627.2015.1084456] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cones comprise only a small portion of the photoreceptors in mammalian retinas. However, cones are vital for color vision and visual perception, and their loss severely diminishes the quality of life for patients with retinal degenerative diseases. Cones function in bright light and have higher demand for energy than rods; yet, the mechanisms that support the energy requirements of cones are poorly understood. One such pathway that potentially could sustain cones under basal and stress conditions is macroautophagy. We addressed the role of macroautophagy in cones by examining how the genetic block of this pathway affects the structural integrity, survival, and function of these neurons. We found that macroautophagy was not detectable in cones under normal conditions but was readily observed following 24 h of fasting. Consistent with this, starvation induced phosphorylation of AMPK specifically in cones indicating cellular starvation. Inhibiting macroautophagy in cones by deleting the essential macroautophagy gene Atg5 led to reduced cone function following starvation suggesting that cones are sensitive to systemic changes in nutrients and activate macroautophagy to maintain their function. ATG5-deficiency rendered cones susceptible to light-induced damage and caused accumulation of damaged mitochondria in the inner segments, shortening of the outer segments, and degeneration of all cone types, revealing the importance of mitophagy in supporting cone metabolic needs. Our results demonstrate that macroautophagy supports the function and long-term survival of cones providing for their unique metabolic requirements and resistance to stress. Targeting macroautophagy has the potential to preserve cone-mediated vision during retinal degenerative diseases.
Collapse
Affiliation(s)
- Zhenqing Zhou
- a Department of Ophthalmology and Visual Sciences, Washington University in St. Louis; School of Medicine ; St. Louis , MO USA
| | - Frans Vinberg
- a Department of Ophthalmology and Visual Sciences, Washington University in St. Louis; School of Medicine ; St. Louis , MO USA
| | - Frank Schottler
- a Department of Ophthalmology and Visual Sciences, Washington University in St. Louis; School of Medicine ; St. Louis , MO USA
| | - Teresa A Doggett
- a Department of Ophthalmology and Visual Sciences, Washington University in St. Louis; School of Medicine ; St. Louis , MO USA
| | - Vladimir J Kefalov
- a Department of Ophthalmology and Visual Sciences, Washington University in St. Louis; School of Medicine ; St. Louis , MO USA
| | - Thomas A Ferguson
- a Department of Ophthalmology and Visual Sciences, Washington University in St. Louis; School of Medicine ; St. Louis , MO USA
| |
Collapse
|
21
|
Sakurai K, Vinberg F, Wang T, Chen J, Kefalov VJ. The Na(+)/Ca(2+), K(+) exchanger 2 modulates mammalian cone phototransduction. Sci Rep 2016; 6:32521. [PMID: 27580676 PMCID: PMC5007492 DOI: 10.1038/srep32521] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/04/2016] [Indexed: 12/15/2022] Open
Abstract
Calcium ions (Ca2+) modulate the phototransduction cascade of vertebrate cone photoreceptors to tune gain, inactivation, and light adaptation. In darkness, the continuous current entering the cone outer segment through cGMP-gated (CNG) channels is carried in part by Ca2+, which is then extruded back to the extracellular space. The mechanism of Ca2+ extrusion from mammalian cones is not understood. The dominant view has been that the cone-specific isoform of the Na+/Ca2+, K+ exchanger, NCKX2, is responsible for removing Ca2+ from their outer segments. However, indirect evaluation of cone function in NCKX2-deficient (Nckx2−/−) mice by electroretinogram recordings revealed normal photopic b-wave responses. This unexpected result suggested that NCKX2 may not be involved in the Ca2+ homeostasis of mammalian cones. To address this controversy, we examined the expression of NCKX2 in mouse cones and performed transretinal recordings from Nckx2−/− mice to determine the effect of NCKX2 deletion on cone function directly. We found that Nckx2−/− cones exhibit compromised phototransduction inactivation, slower response recovery and delayed background adaptation. We conclude that NCKX2 is required for the maintenance of efficient Ca2+ extrusion from mouse cones. However, surprisingly, Nckx2−/− cones adapted normally in steady background light, indicating the existence of additional Ca2+-extruding mechanisms in mammalian cones.
Collapse
Affiliation(s)
- Keisuke Sakurai
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO 63110, USA
| | - Frans Vinberg
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO 63110, USA
| | - Tian Wang
- Zilkha Neurogenetic Institute, Department of Cell and Neurobiology &Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jeannie Chen
- Zilkha Neurogenetic Institute, Department of Cell and Neurobiology &Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO 63110, USA
| |
Collapse
|
22
|
Abstract
It is a deeply engrained notion that the visual pigment rhodopsin signals light as a monomer, even though many G protein-coupled receptors are now known to exist and function as dimers. Nonetheless, recent studies (albeit all in vitro) have suggested that rhodopsin and its chromophore-free apoprotein, R-opsin, may indeed exist as a homodimer in rod disk membranes. Given the overwhelmingly strong historical context, the crucial remaining question, therefore, is whether pigment dimerization truly exists naturally and what function this dimerization may serve. We addressed this question in vivo with a unique mouse line (S-opsin(+)Lrat(-/-)) expressing, transgenically, short-wavelength-sensitive cone opsin (S-opsin) in rods and also lacking chromophore to exploit the fact that cone opsins, but not R-opsin, require chromophore for proper folding and trafficking to the photoreceptor's outer segment. In R-opsin's absence, S-opsin in these transgenic rods without chromophore was mislocalized; in R-opsin's presence, however, S-opsin trafficked normally to the rod outer segment and produced functional S-pigment upon subsequent chromophore restoration. Introducing a competing R-opsin transmembrane helix H1 or helix H8 peptide, but not helix H4 or helix H5 peptide, into these transgenic rods caused mislocalization of R-opsin and S-opsin to the perinuclear endoplasmic reticulum. Importantly, a similar peptide-competition effect was observed even in WT rods. Our work provides convincing evidence for visual pigment dimerization in vivo under physiological conditions and for its role in pigment maturation and targeting. Our work raises new questions regarding a potential mechanistic role of dimerization in rhodopsin signaling.
Collapse
|
23
|
Ingram NT, Sampath AP, Fain GL. Why are rods more sensitive than cones? J Physiol 2016; 594:5415-26. [PMID: 27218707 DOI: 10.1113/jp272556] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022] Open
Abstract
One hundred and fifty years ago Max Schultze first proposed the duplex theory of vision, that vertebrate eyes have two types of photoreceptor cells with differing sensitivity: rods for dim light and cones for bright light and colour detection. We now know that this division is fundamental not only to the photoreceptors themselves but to the whole of retinal and visual processing. But why are rods more sensitive, and how did the duplex retina first evolve? Cells resembling cones are very old, first appearing among cnidarians; the emergence of rods was a key step in the evolution of the vertebrate eye. Many transduction proteins have different isoforms in rods and cones, and others are expressed at different levels. Moreover rods and cones have a different anatomy, with only rods containing membranous discs enclosed by the plasma membrane. These differences must be responsible for the difference in absolute sensitivity, but which are essential? Recent research particularly expressing cone proteins in rods or changing the level of expression seem to show that many of the molecular differences in the activation and decay of the response may have each made a small contribution as evolution proceeded stepwise with incremental increases in sensitivity. Rod outer-segment discs were not essential and developed after single-photon detection. These experiments collectively provide a new understanding of the two kinds of photoreceptors and help to explain how gene duplication and the formation of rod-specific proteins produced the duplex retina, which has remained remarkably constant in physiology from amphibians to man.
Collapse
Affiliation(s)
- Norianne T Ingram
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095-7239, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA, 90095-7000, USA
| | - Gordon L Fain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095-7239, USA. .,Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA, 90095-7000, USA.
| |
Collapse
|
24
|
Zang J, Keim J, Kastenhuber E, Gesemann M, Neuhauss SCF. Recoverin depletion accelerates cone photoresponse recovery. Open Biol 2016; 5:rsob.150086. [PMID: 26246494 PMCID: PMC4554923 DOI: 10.1098/rsob.150086] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The neuronal Ca2+-binding protein Recoverin has been shown to regulate phototransduction termination in mammalian rods. Here we identify four recoverin genes in the zebrafish genome, rcv1a, rcv1b, rcv2a and rcv2b, and investigate their role in modulating the cone phototransduction cascade. While Recoverin-1b is only found in the adult retina, the other Recoverins are expressed throughout development in all four cone types, except Recoverin-1a, which is expressed only in rods and UV cones. Applying a double flash electroretinogram (ERG) paradigm, downregulation of Recoverin-2a or 2b accelerates cone photoresponse recovery, albeit at different light intensities. Exclusive recording from UV cones via spectral ERG reveals that knockdown of Recoverin-1a alone has no effect, but Recoverin-1a/2a double-knockdowns showed an even shorter recovery time than Recoverin-2a-deficient larvae. We also showed that UV cone photoresponse kinetics depend on Recoverin-2a function via cone-specific kinase Grk7a. This is the first in vivo study demonstrating that cone opsin deactivation kinetics determine overall photoresponse shut off kinetics.
Collapse
Affiliation(s)
- Jingjing Zang
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Jennifer Keim
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Edda Kastenhuber
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Matthias Gesemann
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Stephan C F Neuhauss
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| |
Collapse
|
25
|
Exchange of Cone for Rod Phosphodiesterase 6 Catalytic Subunits in Rod Photoreceptors Mimics in Part Features of Light Adaptation. J Neurosci 2015; 35:9225-35. [PMID: 26085644 DOI: 10.1523/jneurosci.3563-14.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED Despite the expression of homologous phototransduction components, the molecular basis for differences in light-evoked responses between rod and cone photoreceptors remains unclear. We examined the role of cGMP phosphodiesterase (PDE6) in this difference by expressing cone PDE6 (PDE6C) in rd1/rd1 rods lacking rod PDE6 (PDE6AB) using transgenic mice. The expression of PDE6C rescues retinal degeneration observed in rd1/rd1 rods. Double-transgenic rods (PDE6C++) were compared with rd1/+ rods based on similar PDE6 expression. PDE6C increased the basal PDE activity and speeded the rate-limiting step for phototransduction deactivation, causing rod photoresponses to appear light adapted, with reduced dark current and sensitivity and faster response kinetics. When PDE6C++ and rd1/+ rods were exposed to similar background light, rd1/+ rods displayed greater desensitization. These results indicate an increased spontaneous activity and faster deactivation of PDE6C compared with PDE6AB in darkness, but that background light increases steady PDE6C activity to a lesser extent. In addition to accelerating the recovery of the photoresponse, faster PDE6C deactivation may blunt the rise in background-induced steady PDE6C activity. Therefore, higher basal PDE6C activity and faster deactivation together partially account for faster and less sensitive cone photoresponses in darkness, whereas a reduced rise of steady PDE6C activity in background light may allow cones to avoid saturation. SIGNIFICANCE STATEMENT Cones are the primary photoreceptors responsible for most of our visual experience. Cone light responses are less sensitive and display speeded responses compared with rods. Despite the fact that rods and cones use a G-protein signaling cascade with similar organization, the mechanistic basis for these differences remains unclear. Here, we examined the role of distinct isoforms of PDE6, the effector enzyme in phototransduction, in these differences. We developed a transgenic mouse model that expresses cone PDE6 in rods and show that the cone PDE6 isoform is partially responsible for the difference in sensitivity and response kinetics between rods and cones.
Collapse
|
26
|
Vinberg F, Wang T, Molday RS, Chen J, Kefalov VJ. A new mouse model for stationary night blindness with mutant Slc24a1 explains the pathophysiology of the associated human disease. Hum Mol Genet 2015; 24:5915-29. [PMID: 26246500 DOI: 10.1093/hmg/ddv319] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/31/2015] [Indexed: 11/12/2022] Open
Abstract
Mutations that affect calcium homeostasis (Ca(2+)) in rod photoreceptors are linked to retinal degeneration and visual disorders such as retinitis pigmentosa and congenital stationary night blindness (CSNB). It is thought that the concentration of Ca(2+) in rod outer segments is controlled by a dynamic balance between influx via cGMP-gated (CNG) channels and extrusion via Na(+)/Ca(2+), K(+) exchangers (NCKX1). The extrusion-driven lowering of rod [Ca(2+)]i following light exposure controls their light adaptation and response termination. Mutant NCKX1 has been linked to autosomal-recessive stationary night blindness. However, whether NCKX1 contributes to light adaptation has not been directly tested and the mechanisms by which human NCKX1 mutations cause night blindness are not understood. Here, we report that the deletion of NCKX1 in mice results in malformed outer segment disks, suppressed expression and function of rod CNG channels and a subsequent 100-fold reduction in rod responses, while preserving normal cone responses. The compensating loss of CNG channel function in the absence of NCKX1-mediated Ca(2+) extrusion may prevent toxic Ca(2+) buildup and provides an explanation for the stationary nature of the associated disorder in humans. Surprisingly, the lack of NCKX1 did not compromise rod background light adaptation, suggesting additional Ca(2+)-extruding mechanisms exist in these cells.
Collapse
Affiliation(s)
- Frans Vinberg
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Tian Wang
- Cell and Neurobiology, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA and
| | - Robert S Molday
- Biochemistry/Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jeannie Chen
- Cell and Neurobiology, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA and
| | - Vladimir J Kefalov
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA,
| |
Collapse
|
27
|
Hofmann L, Palczewski K. Advances in understanding the molecular basis of the first steps in color vision. Prog Retin Eye Res 2015; 49:46-66. [PMID: 26187035 DOI: 10.1016/j.preteyeres.2015.07.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 01/05/2023]
Abstract
Serving as one of our primary environmental inputs, vision is the most sophisticated sensory system in humans. Here, we present recent findings derived from energetics, genetics and physiology that provide a more advanced understanding of color perception in mammals. Energetics of cis-trans isomerization of 11-cis-retinal accounts for color perception in the narrow region of the electromagnetic spectrum and how human eyes can absorb light in the near infrared (IR) range. Structural homology models of visual pigments reveal complex interactions of the protein moieties with the light sensitive chromophore 11-cis-retinal and that certain color blinding mutations impair secondary structural elements of these G protein-coupled receptors (GPCRs). Finally, we identify unsolved critical aspects of color tuning that require future investigation.
Collapse
Affiliation(s)
- Lukas Hofmann
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Krzysztof Palczewski
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
28
|
Jiang L, Wei Y, Ronquillo CC, Marc RE, Yoder BK, Frederick JM, Baehr W. Heterotrimeric kinesin-2 (KIF3) mediates transition zone and axoneme formation of mouse photoreceptors. J Biol Chem 2015; 290:12765-78. [PMID: 25825494 DOI: 10.1074/jbc.m115.638437] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Indexed: 11/06/2022] Open
Abstract
Anterograde intraflagellar transport (IFT) employing kinesin-2 molecular motors has been implicated in trafficking of photoreceptor outer segment proteins. We generated embryonic retina-specific (prefix "emb") and adult tamoxifen-induced (prefix "tam") deletions of KIF3a and IFT88 in adult mice to study photoreceptor ciliogenesis and protein trafficking. In (emb)Kif3a(-/-) and in (emb)Ift88(-/-) mice, basal bodies failed to extend transition zones (connecting cilia) with outer segments, and visual pigments mistrafficked. In contrast, (tam)Kif3a(-/-) and (tam)Ift88(-/-) photoreceptor axonemes disintegrated slowly post-induction, starting distally, but rhodopsin and cone pigments trafficked normally for more than 2 weeks, a time interval during which the outer segment is completely renewed. The results demonstrate that visual pigments transport to the retinal outer segment despite removal of KIF3 and IFT88, and KIF3-mediated anterograde IFT is responsible for photoreceptor transition zone and axoneme formation.
Collapse
Affiliation(s)
- Li Jiang
- From the Departments of Ophthalmology and Visual Sciences and
| | - Yuxiao Wei
- From the Departments of Ophthalmology and Visual Sciences and
| | | | - Robert E Marc
- From the Departments of Ophthalmology and Visual Sciences and
| | - Bradley K Yoder
- the Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, and
| | | | - Wolfgang Baehr
- From the Departments of Ophthalmology and Visual Sciences and the Department of Biology, University of Utah, Salt Lake City, Utah 84112 Neurobiology and Anatomy, University of Utah Health Science Center, Salt Lake City, Utah 84132,
| |
Collapse
|
29
|
Activation and quenching of the phototransduction cascade in retinal cones as inferred from electrophysiology and mathematical modeling. Mol Vis 2015; 21:244-63. [PMID: 25866462 PMCID: PMC4392649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/05/2015] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To experimentally identify and quantify factors responsible for the lower sensitivity of retinal cones compared to rods. METHODS Electrical responses of frog rods and fish (Carassius) cones to short flashes of light were recorded using the suction pipette technique. A fast solution changer was used to apply a solution that fixed intracellular Ca2+ concentration at the prestimulus level, thereby disabling Ca2+ feedback, to the outer segment (OS). The results were analyzed with a specially designed mathematical model of phototransduction. The model included all basic processes of activation and quenching of the phototransduction cascade but omitted unnecessary mechanistic details of each step. RESULTS Judging from the response versus intensity curves, Carassius cones were two to three orders of magnitude less sensitive than frog rods. There was a large scatter in sensitivity among individual cones, with red-sensitive cones being on average approximately two times less sensitive than green-sensitive ones. The scatter was mostly due to different signal amplification, since the kinetic parameters of the responses among cones were far less variable than sensitivity. We argue that the generally accepted definition of the biochemical amplification in phototransduction cannot be used for comparing amplification in rods and cones, since it depends on an irrelevant factor, that is, the cell's volume. We also show that the routinely used simplified parabolic curve fitting to an initial phase of the response leads to a few-fold underestimate of the amplification. We suggest a new definition of the amplification that only includes molecular parameters of the cascade activation, and show how it can be derived from experimental data. We found that the mathematical model with unrestrained parameters can yield an excellent fit to experimental responses. However, the fits with wildly different sets of parameters can be virtually indistinguishable, and therefore cannot provide meaningful data on underlying mechanisms. Based on results of Ca2+-clamp experiments, we developed an approach to strongly constrain the values of many key parameters that set the time course and sensitivity of the photoresponse (such as the dark turnover rate of cGMP, rates of turnoffs of the photoactivated visual pigment and phosphodiesterase, and kinetics of Ca2+ feedback). We show that applying these constraints to our mathematical model enables accurate determination of the biochemical amplification in phototransduction. It appeared that, contrary to many suggestions, maximum biochemical amplification derived for "best" Carassius cones was as high as in frog rods. On the other hand, all turnoff and recovery reactions in cones proceeded approximately 10 times faster than in rods. CONCLUSIONS The main cause of the differing sensitivity of rods and cones is cones' ability to terminate their photoresponse faster.
Collapse
|
30
|
Sakurai K, Chen J, Khani SC, Kefalov VJ. Regulation of mammalian cone phototransduction by recoverin and rhodopsin kinase. J Biol Chem 2015; 290:9239-50. [PMID: 25673692 DOI: 10.1074/jbc.m115.639591] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Indexed: 11/06/2022] Open
Abstract
Cone photoreceptors function under daylight conditions and are essential for color perception and vision with high temporal and spatial resolution. A remarkable feature of cones is that, unlike rods, they remain responsive in bright light. In rods, light triggers a decline in intracellular calcium, which exerts a well studied negative feedback on phototransduction that includes calcium-dependent inhibition of rhodopsin kinase (GRK1) by recoverin. Rods and cones share the same isoforms of recoverin and GRK1, and photoactivation also triggers a calcium decline in cones. However, the molecular mechanisms by which calcium exerts negative feedback on cone phototransduction through recoverin and GRK1 are not well understood. Here, we examined this question using mice expressing various levels of GRK1 or lacking recoverin. We show that although GRK1 is required for the timely inactivation of mouse cone photoresponse, gradually increasing its expression progressively delays the cone response recovery. This surprising result is in contrast with the known effect of increasing GRK1 expression in rods. Notably, the kinetics of cone responses converge and become independent of GRK1 levels for flashes activating more than ∼1% of cone pigment. Thus, mouse cone response recovery in bright light is independent of pigment phosphorylation and likely reflects the spontaneous decay of photoactivated visual pigment. We also find that recoverin potentiates the sensitivity of cones in dim light conditions but does not contribute to their capacity to function in bright light.
Collapse
Affiliation(s)
- Keisuke Sakurai
- From the Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jeannie Chen
- the Zilkha Neurogenetic Institute, Department of Cell and Neurobiology & Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, and
| | - Shahrokh C Khani
- the Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Vladimir J Kefalov
- From the Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110,
| |
Collapse
|
31
|
Isayama T, Chen Y, Kono M, Fabre E, Slavsky M, DeGrip WJ, Ma JX, Crouch RK, Makino CL. Coexpression of three opsins in cone photoreceptors of the salamander Ambystoma tigrinum. J Comp Neurol 2014; 522:2249-65. [PMID: 24374736 DOI: 10.1002/cne.23531] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/24/2013] [Accepted: 12/20/2013] [Indexed: 12/12/2022]
Abstract
Although more than one type of visual opsin is present in the retina of most vertebrates, it was thought that each type of photoreceptor expresses only one opsin. However, evidence has accumulated that some photoreceptors contain more than one opsin, in many cases as a result of a developmental transition from the expression of one opsin to another. The salamander UV-sensitive (UV) cone is particularly notable because it contains three opsins (Makino and Dodd [1996] J Gen Physiol 108:27-34). Two opsin types are expressed at levels more than 100 times lower than the level of the primary opsin. Here, immunohistochemical experiments identified the primary component as a UV cone opsin and the two minor components as the short wavelength-sensitive (S) and long wavelength-sensitive (L) cone opsins. Based on single-cell recordings of 156 photoreceptors, the presence of three components in UV cones of hatchlings and terrestrial adults ruled out a developmental transition. There was no evidence for multiple opsin types within rods or S cones, but immunohistochemistry and partial bleaching in conjunction with single-cell recording revealed that both single and double L cones contained low levels of short wavelength-sensitive pigments in addition to the main L visual pigment. These results raise the possibility that coexpression of multiple opsins in other vertebrates was overlooked because a minor component absorbing at short wavelengths was masked by the main visual pigment or because the expression level of a component absorbing at long wavelengths was exceedingly low.
Collapse
Affiliation(s)
- Tomoki Isayama
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary and Harvard Medical School, Boston, Massachusetts, 02114
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mannu GS. Retinal phototransduction. NEUROSCIENCES (RIYADH, SAUDI ARABIA) 2014; 19:275-80. [PMID: 25274585 PMCID: PMC4727664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Vision is perhaps the most important of all our senses, and gives us an immense amount of information regarding the outside world. The initial format in which this information reaches the retina are photons; particles of energy radiation of a given wavelength emitted or reflected from our surroundings. The brain itself however, perceives information in electrical signals via action potentials and changes in electrochemical gradients. The processes involved in the transduction of photons into electrical potentials will be the focus of this article. This review article summarizes the recent advances in understanding these complex pathways and provides an overview of the main molecules involved in the neurobiology of vision.
Collapse
Affiliation(s)
- Gurdeep S. Mannu
- From the Academic Medicine Department, Norfolk and Norwich University Hospital, Norfolk, United Kingdom,Address correspondence and reprint request to: Dr. Gurdeep S. Mannu, Academic Medicine Department, Norfolk and Norwich University Hospital, Colney Lane, Norwich, Norfolk NR4 7UY, United Kingdom. Tel. +44 (1603) 286286. Fax. +44 (1603) 287211. E-mail:
| |
Collapse
|
33
|
Sundermeier TR, Zhang N, Vinberg F, Mustafi D, Kohno H, Golczak M, Bai X, Maeda A, Kefalov VJ, Palczewski K. DICER1 is essential for survival of postmitotic rod photoreceptor cells in mice. FASEB J 2014; 28:3780-91. [PMID: 24812086 DOI: 10.1096/fj.14-254292] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Photoreceptor cell death is the proximal cause of blindness in many retinal degenerative disorders; hence, understanding the gene regulatory networks that promote photoreceptor survival is at the forefront of efforts to combat blindness. Down-regulation of the microRNA (miRNA)-processing enzyme DICER1 in the retinal pigmented epithelium has been implicated in geographic atrophy, an advanced form of age-related macular degeneration (AMD). However, little is known about the function of DICER1 in mature rod photoreceptor cells, another retinal cell type that is severely affected in AMD. Using a conditional-knockout (cKO) mouse model, we report that loss of DICER1 in mature postmitotic rods leads to robust retinal degeneration accompanied by loss of visual function. At 14 wk of age, cKO mice exhibit a 90% reduction in photoreceptor nuclei and a 97% reduction in visual chromophore compared with those in control littermates. Before degeneration, cKO mice do not exhibit significant defects in either phototransduction or the visual cycle, suggesting that miRNAs play a primary role in rod photoreceptor survival. Using comparative small RNA sequencing analysis, we identified rod photoreceptor miRNAs of the miR-22, miR-26, miR-30, miR-92, miR-124, and let-7 families as potential factors involved in regulating the survival of rods.
Collapse
Affiliation(s)
| | | | - Frans Vinberg
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | | | | | | | | - Akiko Maeda
- Department of Pharmacology, Department of Ophthalmology and Visual Sciences, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA; and
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | |
Collapse
|
34
|
Abstract
Arrestin-1 is the second most abundant protein in rod photoreceptors and is nearly equimolar to rhodopsin. Its well-recognized role is to "arrest" signaling from light-activated, phosphorylated rhodopsin, a prototypical G protein-coupled receptor. In doing so, arrestin-1 plays a key role in the rapid recovery of the light response. Arrestin-1 exists in a basal conformation that is stabilized by two independent sets of intramolecular interactions. The intramolecular constraints are disrupted by encountering (1) active conformation of the receptor (R*) and (2) receptor-attached phosphates. Requirement for these two events ensures its highly specific high-affinity binding to phosphorylated, light-activated rhodopsin (P-R*). In the dark-adapted state, the basal form is further organized into dimers and tetramers. Emerging data suggest pleiotropic roles of arrestin-1 beyond the functional range of rod cells. These include light-induced arrestin-1 translocation from the inner segment to the outer segment, a process that may be protective against cellular damage incurred by constitutive signaling. Its expanding list of binding partners also hints at additional, yet to be characterized functions. Uncovering these novel roles of arrestin-1 is a subject of future studies.
Collapse
|
35
|
Kojima K, Imamoto Y, Maeda R, Yamashita T, Shichida Y. Rod visual pigment optimizes active state to achieve efficient G protein activation as compared with cone visual pigments. J Biol Chem 2013; 289:5061-73. [PMID: 24375403 DOI: 10.1074/jbc.m113.508507] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Most vertebrate retinas contain two types of photoreceptor cells, rods and cones, which show different photoresponses to mediate scotopic and photopic vision, respectively. These cells contain different types of visual pigments, rhodopsin and cone visual pigments, respectively, but little is known about the molecular properties of cone visual pigments under physiological conditions, making it difficult to link the molecular properties of rhodopsin and cone visual pigments with the differences in photoresponse between rods and cones. Here we prepared bovine and mouse rhodopsin (bvRh and mRh) and chicken and mouse green-sensitive cone visual pigments (cG and mG) embedded in nanodiscs and applied time-resolved fluorescence spectroscopy to compare their Gt activation efficiencies. Rhodopsin exhibited greater Gt activation efficiencies than cone visual pigments. Especially, the Gt activation efficiency of mRh was about 2.5-fold greater than that of mG at 37 °C, which is consistent with our previous electrophysiological data of knock-in mice. Although the active state (Meta-II) was in equilibrium with inactive states (Meta-I and Meta-III), quantitative determination of Meta-II in the equilibrium showed that the Gt activation efficiency per Meta-II of bvRh was also greater than those of cG and mG. These results indicated that efficient Gt activation by rhodopsin, resulting from an optimized active state of rhodopsin, is one of the causes of the high amplification efficiency of rods.
Collapse
Affiliation(s)
- Keiichi Kojima
- From the Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | | | |
Collapse
|
36
|
Cone phosphodiesterase-6α' restores rod function and confers distinct physiological properties in the rod phosphodiesterase-6β-deficient rd10 mouse. J Neurosci 2013; 33:11745-53. [PMID: 23864662 DOI: 10.1523/jneurosci.1536-13.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Phosphodiesterase-6 (PDE6) is the key effector enzyme of the vertebrate phototransduction pathway in rods and cones. Rod PDE6 catalytic core is composed of two distinct subunits, PDE6α and PDE6β, whereas two identical PDE6α' subunits form the cone PDE6 catalytic core. It is not known whether this difference in PDE6 catalytic subunit identity contributes to the functional differences between rods and cones. To address this question, we expressed cone PDE6α' in the photoreceptor cells of the retinal degeneration 10 (rd10) mouse that carries a mutation in rod PDEβ subunit. We show that adeno-associated virus-mediated subretinal delivery of PDE6α' rescues rod electroretinogram responses and preserves retinal structure, indicating that cone PDE6α' can couple effectively to the rod phototransduction pathway. We also show that restoration of light sensitivity in rd10 rods is attributable to assembly of PDE6α' with rod PDE6γ. Single-cell recordings revealed that, surprisingly, rods expressing cone PDE6α' are twofold more sensitive to light than wild-type rods, most likely because of the slower shutoff of their light responses. Unlike in wild-type rods, the response kinetics in PDE6α'-treated rd10 rods accelerated with increasing flash intensity, indicating a possible direct feedback modulation of cone PDE6α' activity. Together, these results demonstrate that cone PDE6α' can functionally substitute for rod PDEαβ in vivo, conferring treated rods with distinct physiological properties.
Collapse
|
37
|
Imamoto Y, Shichida Y. Cone visual pigments. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1837:664-73. [PMID: 24021171 DOI: 10.1016/j.bbabio.2013.08.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/07/2013] [Accepted: 08/30/2013] [Indexed: 12/16/2022]
Abstract
Cone visual pigments are visual opsins that are present in vertebrate cone photoreceptor cells and act as photoreceptor molecules responsible for photopic vision. Like the rod visual pigment rhodopsin, which is responsible for scotopic vision, cone visual pigments contain the chromophore 11-cis-retinal, which undergoes cis-trans isomerization resulting in the induction of conformational changes of the protein moiety to form a G protein-activating state. There are multiple types of cone visual pigments with different absorption maxima, which are the molecular basis of color discrimination in animals. Cone visual pigments form a phylogenetic sister group with non-visual opsin groups such as pinopsin, VA opsin, parapinopsin and parietopsin groups. Cone visual pigments diverged into four groups with different absorption maxima, and the rhodopsin group diverged from one of the four groups of cone visual pigments. The photochemical behavior of cone visual pigments is similar to that of pinopsin but considerably different from those of other non-visual opsins. G protein activation efficiency of cone visual pigments is also comparable to that of pinopsin but higher than that of the other non-visual opsins. Recent measurements with sufficient time-resolution demonstrated that G protein activation efficiency of cone visual pigments is lower than that of rhodopsin, which is one of the molecular bases for the lower amplification of cones compared to rods. In this review, the uniqueness of cone visual pigments is shown by comparison of their molecular properties with those of non-visual opsins and rhodopsin. This article is part of a Special Issue entitled: Retinal Proteins - You can teach an old dog new tricks.
Collapse
Affiliation(s)
- Yasushi Imamoto
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Yoshinori Shichida
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
38
|
Zhang T, Fu Y. A Phe-rich region in short-wavelength sensitive opsins is responsible for their aggregation in the absence of 11-cis-retinal. FEBS Lett 2013; 587:2430-4. [PMID: 23792161 DOI: 10.1016/j.febslet.2013.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 06/04/2013] [Accepted: 06/07/2013] [Indexed: 01/05/2023]
Abstract
Human blue and mouse S-opsin are prone to aggregation in the absence of 11-cis-retinal, which underlie the rapid cone degeneration in human patients and animal models of Leber congenital amaurosis (LCA). By in silico analysis and domain swapping experiments, we show that a Phe-rich region in short-wavelength sensitive (SWS) opsins, but not in medium/long-wavelength sensitive opsins, is responsible for SWS opsin aggregation. Mutagenesis studies suggest that Phe residues in this region are critical in mediating protein aggregation. Fusing the Phe-rich region of SWS opsins to GFP causes the latter to aggregate. Our findings suggest that new therapeutics can be designed to disrupt the Phe-rich region in preventing cone degeneration due to S-opsin aggregation in LCA.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Ophthalmology and Visual Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
39
|
Imamoto Y, Seki I, Yamashita T, Shichida Y. Efficiencies of activation of transducin by cone and rod visual pigments. Biochemistry 2013; 52:3010-8. [PMID: 23570417 DOI: 10.1021/bi3015967] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
How the light-induced transducin (Gt) activation process differs biochemically between cone visual pigments and rod visual pigment (rhodopsin) has remained unclear, because the Gt-activating state (Meta-II) of cone visual pigment decays too fast to precisely measure the activation efficiency by conventional biochemical methods such as the GTPγS binding assay. Here we measured the activation efficiencies of chicken green-sensitive cone visual pigment (cG) and bovine rhodopsin (bRh) in real time by monitoring the intrinsic fluorescence of tryptophan residues in the pigments and Gt. Michaelis-Menten analysis of Gt activation showed that the initial velocity for cG was approximately half that for bRh, while their Michaelis constants were comparable. Gt activation by cG was immediately slowed because of the fast hydrolysis of the retinal Schiff base in Meta-II, but this hydrolysis was suppressed by forming the complex with Gt. Using mutants of cG and bRh for positions 122 and 189, which exhibit altered rates of chromophore hydrolysis in Meta-II, we found that the initial velocity of Gt activation is negatively correlated with the rate of chromophore hydrolysis. These results suggest that the amino acid residues at positions 122 and 189 account for not only the resistance to the chromophore hydrolysis in Meta-II but also the conformation of Meta-II for efficient Gt activation. The substantially longer lifetime of the Gt activating state of Rh would be necessary to suppress the spontaneous quenching by the stochastic decay of the Gt-activating state when a rod responds to a single photon.
Collapse
Affiliation(s)
- Yasushi Imamoto
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | |
Collapse
|
40
|
Mao W, Miyagishima KJ, Yao Y, Soreghan B, Sampath AP, Chen J. Functional comparison of rod and cone Gα(t) on the regulation of light sensitivity. J Biol Chem 2013; 288:5257-67. [PMID: 23288843 DOI: 10.1074/jbc.m112.430058] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The signaling cascades mediated by G protein-coupled receptors (GPCRs) exhibit a wide spectrum of spatial and temporal response properties to fulfill diverse physiological demands. However, the mechanisms that shape the signaling response of the GPCR are not well understood. In this study, we replaced cone transducin α (cTα) for rod transducin α (rTα) in rod photoreceptors of transgenic mice, which also express S opsin, to evaluate the role of Gα subtype on signal amplification from different GPCRs in the same cell; such analysis may explain functional differences between retinal rod and cone photoreceptors. We showed that ectopically expressed cTα 1) forms a heterotrimeric complex with rod Gβ(1)γ(1), 2) substitutes equally for rTα in generating photoresponses initiated by either rhodopsin or S-cone opsin, and 3) exhibited similar light-activated translocation as endogenous rTα in rods and endogenous cTα in cones. Thus, rTα and cTα appear functionally interchangeable. Interestingly, light sensitivity appeared to correlate with the concentration of cTα when expression is reduced below 35% of normal. However, quantification of endogenous cTα concentration in cones showed a higher level to rTα in rods. Thus, reduced sensitivity in cones cannot be explained by reduced coupling efficiency between the GPCR and G protein or a lower concentration of G protein in cones versus rods.
Collapse
Affiliation(s)
- Wen Mao
- Department of Cell and Neurobiology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | |
Collapse
|
41
|
Chen J, Sampath AP. Structure and Function of Rod and Cone Photoreceptors. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
42
|
Zhang T, Baehr W, Fu Y. Chemical chaperone TUDCA preserves cone photoreceptors in a mouse model of Leber congenital amaurosis. Invest Ophthalmol Vis Sci 2012; 53:3349-56. [PMID: 22531707 DOI: 10.1167/iovs.12-9851] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Mutations in either retinoid isomerase (RPE65) or lecithin-retinol acyltransferase (LRAT) lead to Leber congenital amaurosis (LCA). By using the Lrat(-/-) mouse model, previous studies have shown that the rapid cone degeneration in LCA was caused by endoplasmic reticulum (ER) stress induced by S-opsin aggregation. The purpose of this study is to examine the efficacy of an ER chemical chaperone, tauroursodeoxycholic acid (TUDCA), in preserving cones in the Lrat(-/-) model. METHODS Lrat(-/-) mice were systemically administered with TUDCA and vehicle (0.15 M NaHCO(3)) every 3 days from P9 to P28. Cone cell survival was determined by counting cone cells on flat-mounted retinas. The expression and subcellular localization of cone-specific proteins were analyzed by western blotting and immunohistochemistry, respectively. RESULTS TUDCA treatment reduced ER stress and apoptosis in Lrat(-/-) retina. It significantly slowed down cone degeneration in Lrat(-/-) mice, resulting in a ∼3-fold increase in cone density in the ventral and central retina as compared with the vehicle-treated mice at P28. Furthermore, TUDCA promoted the degradation of cone membrane-associated proteins by enhancing the ER-associated protein degradation pathway. CONCLUSIONS Systemic injection of TUDCA is effective in reducing ER stress, preventing apoptosis, and preserving cones in Lrat(-/-) mice. TUDCA has the potential to lead to the development of a new class of therapeutic drugs for treating LCA.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Ophthalmology & Visual Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah 84132, USA
| | | | | |
Collapse
|
43
|
Kefalov VJ. Rod and cone visual pigments and phototransduction through pharmacological, genetic, and physiological approaches. J Biol Chem 2011; 287:1635-41. [PMID: 22074928 DOI: 10.1074/jbc.r111.303008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Activation of the visual pigment by light in rod and cone photoreceptors initiates our visual perception. As a result, the signaling properties of visual pigments, consisting of a protein, opsin, and a chromophore, 11-cis-retinal, play a key role in shaping the light responses of photoreceptors. The combination of pharmacological, physiological, and genetic tools has been a powerful approach advancing our understanding of the interactions between opsin and chromophore and how they affect the function of visual pigments. The signaling properties of the visual pigments modulate many aspects of the function of rods and cones, producing their unique physiological properties.
Collapse
Affiliation(s)
- Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| |
Collapse
|
44
|
Arshavsky VY, Burns ME. Photoreceptor signaling: supporting vision across a wide range of light intensities. J Biol Chem 2011; 287:1620-6. [PMID: 22074925 DOI: 10.1074/jbc.r111.305243] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
For decades, photoreceptors have been an outstanding model system for elucidating basic principles in sensory transduction and biochemistry and for understanding many facets of neuronal cell biology. In recent years, new knowledge of the kinetics of signaling and the large-scale movements of proteins underlying signaling has led to a deeper appreciation of the photoreceptor's unique challenge in mediating the first steps in vision over a wide range of light intensities.
Collapse
Affiliation(s)
- Vadim Y Arshavsky
- Departments of Ophthalmology and Pharmacology, Duke University, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
45
|
Abstract
A negative phototransduction feedback in rods and cones is critical for the timely termination of their light responses and for extending their function to a wide range of light intensities. The calcium feedback mechanisms that modulate phototransduction in rods have been studied extensively. However, the corresponding modulation mechanisms that enable cones to terminate rapidly their light responses and to adapt in bright light, properties critical for our daytime vision, are still not understood. In cones, calcium feedback to guanylyl cyclase is potentially a key step in phototransduction modulation. The guanylyl cyclase activity is modulated by the calcium-binding guanylyl cyclase activating proteins (GCAP1 and GCAP2). Here, we used single-cell and transretinal recordings from mouse to determine how GCAPs modulate dark-adapted responses as well as light adaptation in mammalian cones. Deletion of GCAPs increased threefold the amplitude and dramatically prolonged the light responses in dark-adapted mouse cones. It also reduced the operating range of mouse cones in background illumination and severely impaired their light adaptation. Thus, GCAPs exert powerful modulation on the mammalian cone phototransduction cascade and play an important role in setting the functional properties of cones in darkness and during light adaptation. Surprisingly, despite their better adaptation capacity and wider calcium dynamic range, mammalian cones were modulated by GCAPs to a lesser extent than mammalian rods. We conclude that a disparity in the strength of GCAP modulation cannot explain the differences in the dark-adapted properties or in the operating ranges of mammalian rods and cones.
Collapse
|
46
|
Gurevich VV, Hanson SM, Song X, Vishnivetskiy SA, Gurevich EV. The functional cycle of visual arrestins in photoreceptor cells. Prog Retin Eye Res 2011; 30:405-30. [PMID: 21824527 DOI: 10.1016/j.preteyeres.2011.07.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 01/14/2023]
Abstract
Visual arrestin-1 plays a key role in the rapid and reproducible shutoff of rhodopsin signaling. Its highly selective binding to light-activated phosphorylated rhodopsin is an integral part of the functional perfection of rod photoreceptors. Structure-function studies revealed key elements of the sophisticated molecular mechanism ensuring arrestin-1 selectivity and paved the way to the targeted manipulation of the arrestin-1 molecule to design mutants that can compensate for congenital defects in rhodopsin phosphorylation. Arrestin-1 self-association and light-dependent translocation in photoreceptor cells work together to keep a constant supply of active rhodopsin-binding arrestin-1 monomer in the outer segment. Recent discoveries of arrestin-1 interaction with other signaling proteins suggest that it is a much more versatile signaling regulator than previously thought, affecting the function of the synaptic terminals and rod survival. Elucidation of the fine molecular mechanisms of arrestin-1 interactions with rhodopsin and other binding partners is necessary for the comprehensive understanding of rod function and for devising novel molecular tools and therapeutic approaches to the treatment of visual disorders.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave, PRB, Rm 417D, Nashville, TN 37232, USA.
| | | | | | | | | |
Collapse
|
47
|
Abstract
Vision begins with photoisomerization of visual pigments. Thermal energy can complement photon energy to drive photoisomerization, but it also triggers spontaneous pigment activation as noise that interferes with light detection. For half a century, the mechanism underlying this dark noise has remained controversial. We report here a quantitative relation between a pigment's photoactivation energy and its peak-absorption wavelength, λ(max). Using this relation and assuming that pigment activations by light and heat go through the same ground-state isomerization energy barrier, we can predict the relative noise of diverse pigments with multi-vibrational-mode thermal statistics. The agreement between predictions and our measurements strongly suggests that pigment noise arises from canonical isomerization. The predicted high noise for pigments with λ(max) in the infrared presumably explains why they apparently do not exist in nature.
Collapse
Affiliation(s)
- Dong-Gen Luo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
48
|
Sun H. Membrane receptors and transporters involved in the function and transport of vitamin A and its derivatives. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:99-112. [PMID: 21704730 DOI: 10.1016/j.bbalip.2011.06.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 06/01/2011] [Accepted: 06/10/2011] [Indexed: 12/12/2022]
Abstract
The eye is the human organ most sensitive to vitamin A deficiency because of vision's absolute and heavy dependence on vitamin A for light perception. Studies of the molecular basis of vision have provided important insights into the intricate mechanistic details of the function, transport and recycling of vitamin A and its derivatives (retinoid). This review focuses on retinoid-related membrane receptors and transporters. Three kinds of mammalian membrane receptors and transporters are discussed: opsins, best known as vitamin A-based light sensors in vision; ABCA4, an ATP-dependent transporter specializes in the transport of vitamin A derivative; and STRA6, a recently identified membrane receptor that mediates cellular uptake of vitamin A. The evolutionary driving forces for their existence and the wide spectrum of human diseases associated with these proteins are discussed. Lessons learned from the study of the visual system might be useful for understanding retinoid biology and retinoid-related diseases in other organ systems as well. This article is part of a Special Issue entitled Retinoid and Lipid Metabolism.
Collapse
Affiliation(s)
- Hui Sun
- Department of Physiology, Jules Stein Eye Institute, Brian Research Institute, and Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Cone opsin determines the time course of cone photoreceptor degeneration in Leber congenital amaurosis. Proc Natl Acad Sci U S A 2011; 108:8879-84. [PMID: 21555576 DOI: 10.1073/pnas.1017127108] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mutations in RPE65 or lecithin-retinol acyltransferase (LRAT) disrupt 11-cis-retinal recycling and cause Leber congenital amaurosis (LCA), the most severe retinal dystrophy in early childhood. We used Lrat(-)(/-), a murine model for LCA, to investigate the mechanism of rapid cone degeneration. Although both M and S cone opsins mistrafficked as reported previously, mislocalized M-opsin was degraded whereas mislocalized S-opsin accumulated in Lrat(-)(/-) cones before the onset of massive ventral/central cone degeneration. As the ventral and central retina express higher levels of S-opsin than the dorsal retina in mice, our results may explain why ventral and central cones degenerate more rapidly than dorsal cones in Rpe65(-)(/-) and Lrat(-)(/-) LCA models. In addition, human blue opsin and mouse S-opsin, but not mouse M-opsin or human red/green opsins, aggregated to form cytoplasmic inclusions in transfected cells, which may explain why blue cone function is lost earlier than red/green-cone function in patients with LCA. The aggregation of short-wavelength opsins likely caused rapid cone degenerations through an endoplasmic reticulum stress pathway, as demonstrated in both the Lrat(-)(/-) retina and transfected cells. Replacing rhodopsin with S-opsin in Lrat(-)(/-) rods resulted in mislocalization and aggregation of S-opsin in the inner segment and the synaptic region of rods, ER stress, and dramatically accelerated rod degeneration. Our results demonstrate that cone opsins play a major role in determining the degeneration rate of photoreceptors in LCA.
Collapse
|
50
|
Renninger SL, Gesemann M, Neuhauss SCF. Cone arrestin confers cone vision of high temporal resolution in zebrafish larvae. Eur J Neurosci 2011; 33:658-67. [PMID: 21299656 DOI: 10.1111/j.1460-9568.2010.07574.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vision of high temporal resolution depends on careful regulation of photoresponse kinetics, beginning with the lifetime of activated photopigment. The activity of rhodopsin is quenched by high-affinity binding of arrestin to photoexcited phosphorylated photopigment, which effectively terminates the visual transduction cascade. This regulation mechanism is well established for rod photoreceptors, yet its role for cone vision is still controversial. In this study we therefore analyzed arrestin function in the cone-dominated vision of larval zebrafish. For both rod (arrS ) and cone (arr3 ) arrestin we isolated two paralogs, each expressed in the respective subset of photoreceptors. Labeling with paralog-specific antibodies revealed subfunctionalized expression of Arr3a in M- and L-cones, and Arr3b in S- and UV-cones. The inactivation of arr3a by morpholino knockdown technology resulted in a severe delay in photoresponse recovery which, under bright light conditions, was rate-limiting. Comparison to opsin phosphorylation-deficient animals confirmed the role of cone arrestin in late cone response recovery. Arr3a activity partially overlapped with the function of the cone-specific kinase Grk7a involved in initial response recovery. Behavioral measurements further revealed Arr3a deficiency to be sufficient to reduce temporal contrast sensitivity, providing evidence for the importance of arrestin in cone vision of high temporal resolution.
Collapse
Affiliation(s)
- Sabine L Renninger
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | | | |
Collapse
|