1
|
Ferng D, Sun W, Shieh BH. Differential activation of rhodopsin triggers distinct endocytic trafficking and recycling in vivo via differential phosphorylation. PLoS One 2024; 19:e0303882. [PMID: 38848405 PMCID: PMC11161057 DOI: 10.1371/journal.pone.0303882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
Activated GPCRs are phosphorylated and internalized mostly via clathrin-mediated endocytosis (CME), which are then sorted for recycling or degradation. We investigated how differential activation of the same GPCR affects its endocytic trafficking in vivo using rhodopsin as a model in pupal photoreceptors of flies expressing mCherry-tagged rhodopsin 1 (Rh1-mC) or GFP-tagged arrestin 1 (Arr1-GFP). Upon blue light stimulation, activated Rh1 recruited Arr1-GFP to the rhabdomere, which became co-internalized and accumulated in cytoplasmic vesicles of photoreceptors. This internalization was eliminated in shits1 mutants affecting dynamin. Moreover, it was blocked by either rdgA or rdgB mutations affecting the PIP2 biosynthesis. Together, the blue light-initiated internalization of Rh1 and Arr1 belongs to CME. Green light stimulation also triggered the internalization and accumulation of activated Rh1-mC in the cytoplasm but with faster kinetics. Importantly, Arr1-GFP was also recruited to the rhabdomere but not co-internalized with Rh1-mC. This endocytosis was not affected in shits1 nor rdgA mutants, indicating it is not CME. We explored the fate of internalized Rh1-mC following CME and observed it remained in cytoplasmic vesicles following 30 min of dark adaptation. In contrast, in the non-CME Rh1-mC appeared readily recycled back to the rhabdomere within five min of dark treatment. This faster recycling may be regulated by rhodopsin phosphatase, RdgC. Together, we demonstrate two distinct endocytic and recycling mechanisms of Rh1 via two light stimulations. It appears that each stimulation triggers a distinct conformation leading to different phosphorylation patterns of Rh1 capable of recruiting Arr1 to rhabdomeres. However, a more stable interaction leads to the co-internalization of Arr1 that orchestrates CME. A stronger Arr1 association appears to impede the recycling of the phosphorylated Rh1 by preventing the recruitment of RdgC. We conclude that conformations of activated rhodopsin determine the downstream outputs upon phosphorylation that confers differential protein-protein interactions.
Collapse
Affiliation(s)
- Darwin Ferng
- Department of Pharmacology, Center for Molecular Neuroscience and Vision Research Center, Vanderbilt University, Nashville, TN, United States of America
| | - Wesley Sun
- Department of Pharmacology, Center for Molecular Neuroscience and Vision Research Center, Vanderbilt University, Nashville, TN, United States of America
| | - Bih-Hwa Shieh
- Department of Pharmacology, Center for Molecular Neuroscience and Vision Research Center, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
2
|
Morshedian A, Jiang Z, Radu RA, Fain GL, Sampath AP. Genetic manipulation of rod-cone differences in mouse retina. PLoS One 2024; 19:e0300584. [PMID: 38709779 PMCID: PMC11073714 DOI: 10.1371/journal.pone.0300584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/01/2024] [Indexed: 05/08/2024] Open
Abstract
Though rod and cone photoreceptors use similar phototransduction mechanisms, previous model calculations have indicated that the most important differences in their light responses are likely to be differences in amplification of the G-protein cascade, different decay rates of phosphodiesterase (PDE) and pigment phosphorylation, and different rates of turnover of cGMP in darkness. To test this hypothesis, we constructed TrUx;GapOx rods by crossing mice with decreased transduction gain from decreased transducin expression, with mice displaying an increased rate of PDE decay from increased expression of GTPase-activating proteins (GAPs). These two manipulations brought the sensitivity of TrUx;GapOx rods to within a factor of 2 of WT cone sensitivity, after correcting for outer-segment dimensions. These alterations did not, however, change photoreceptor adaptation: rods continued to show increment saturation though at a higher background intensity. These experiments confirm model calculations that rod responses can mimic some (though not all) of the features of cone responses after only a few changes in the properties of transduction proteins.
Collapse
Affiliation(s)
- Ala Morshedian
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Zhichun Jiang
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Roxana A. Radu
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Gordon L. Fain
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Alapakkam P. Sampath
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| |
Collapse
|
3
|
FitzHugh ZT, Schiller MR. Systematic Assessment of Protein C-Termini Mutated in Human Disorders. Biomolecules 2023; 13:biom13020355. [PMID: 36830724 PMCID: PMC9953674 DOI: 10.3390/biom13020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
All proteins have a carboxyl terminus, and we previously summarized eight mutations in binding and trafficking sequence determinants in the C-terminus that, when disrupted, cause human diseases. These sequence elements for binding and trafficking sites, as well as post-translational modifications (PTMs), are called minimotifs or short linear motifs. We wanted to determine how frequently mutations in minimotifs in the C-terminus cause disease. We searched specifically for PTMs because mutation of a modified amino acid almost always changes the chemistry of the side chain and can be interpreted as loss-of-function. We analyzed data from ClinVar for disease variants, Minimotif Miner and the C-terminome for PTMs, and RefSeq for protein sequences, yielding 20 such potential disease-causing variants. After additional screening, they include six with a previously reported PTM disruption mechanism and nine with new hypotheses for mutated minimotifs in C-termini that may cause disease. These mutations were generally for different genes, with four different PTM types and several different diseases. Our study helps to identify new molecular mechanisms for nine separate variants that cause disease, and this type of analysis could be extended as databases grow and to binding and trafficking motifs. We conclude that mutated motifs in C-termini are an infrequent cause of disease.
Collapse
Affiliation(s)
- Zachary T. FitzHugh
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, 4505 S. Maryland Pkwy, Las Vegas, NV 89154, USA
- School of Life Sciences, University of Nevada, 4505 S. Maryland Parkway, Las Vegas, NV 89154, USA
| | - Martin R. Schiller
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, 4505 S. Maryland Pkwy, Las Vegas, NV 89154, USA
- School of Life Sciences, University of Nevada, 4505 S. Maryland Parkway, Las Vegas, NV 89154, USA
- Heligenics Inc., 833 Las Vegas Blvd. North, Suite B, Las Vegas, NV 89101, USA
- Correspondence: ; Tel.: +1-702-895-5546; Fax: +1-702-895-5728
| |
Collapse
|
4
|
Berkowitz BA, Podolsky RH, Childers KL, Roberts R, Katz R, Waseem R, Robbings BM, Hass DT, Hurley JB, Sweet IR, Goodman C, Qian H, Alvisio B, Heaps S. Transducin-Deficient Rod Photoreceptors Evaluated With Optical Coherence Tomography and Oxygen Consumption Rate Energy Biomarkers. Invest Ophthalmol Vis Sci 2022; 63:22. [PMID: 36576748 PMCID: PMC9804021 DOI: 10.1167/iovs.63.13.22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose To test the hypothesis that rod energy biomarkers in light and dark are similar in mice without functional rod transducin (Gnat1rd17). Methods Gnat1rd17 and wildtype (WT) mice were studied in canonically low energy demand (light) and high energy demand (dark) conditions. We measured rod inner segment ellipsoid zone (ISez) profile shape, external limiting membrane-retinal pigment epithelium (ELM-RPE) thickness, and magnitude of a hyporeflective band (HB) intensity dip located between photoreceptor tips and apical RPE; antioxidants were given in a subset of mice. Oxygen consumption rate (OCR) and visual performance indexes were also measured. Results The lower energy demand expected in light-adapted wildtype retinas was associated with an elongated ISez, thicker ELM-RPE, and higher HB magnitude, and lower OCR compared to high energy demand conditions in the dark. Gnat1rd17 mice showed a wildtype-like ISez profile shape at 20 minutes of light that became rounder at 60 minutes; at both times, ELM-RPE was smaller than wildtype values, and the HB magnitude was unmeasurable. OCR was higher than in the dark. Light-adapted Gnat1rd17 mice biomarkers were unaffected by anti-oxidants. Gnat1rd17 mice showed modest outer nuclear layer thinning and no reduction in visual performance indexes. Conclusions Light-stimulated changes in all biomarkers in WT mice are consistent with the established light-induced decrease in net energy demand. In contrast, biomarker changes in Gnat1rd17 mice raise the possibility that light increases net energy demand in the absence of rod phototransduction.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Robert H Podolsky
- Biostatistics and Study Methodology, Children's National Hospital, Silver Spring, Maryland, United States
| | - Karen Lins Childers
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, United States
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Ryan Katz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Rida Waseem
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Brian M Robbings
- Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington, United States.,Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Daniel T Hass
- Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - James B Hurley
- Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - Ian R Sweet
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Cole Goodman
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Bruno Alvisio
- OSIO Bioinformatics Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Sam Heaps
- OSIO Bioinformatics Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
5
|
Hsieh CL, Yao Y, Gurevich VV, Chen J. Arrestin Facilitates Rhodopsin Dephosphorylation in Vivo. J Neurosci 2022; 42:3537-3545. [PMID: 35332081 PMCID: PMC9053844 DOI: 10.1523/jneurosci.0141-22.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/08/2022] [Accepted: 03/17/2022] [Indexed: 01/14/2023] Open
Abstract
Deactivation of G-protein-coupled receptors (GPCRs) involves multiple phosphorylations followed by arrestin binding, which uncouples the GPCR from G-protein activation. Some GPCRs, such as rhodopsin, are reused many times. Arrestin dissociation and GPCR dephosphorylation are key steps in the recycling process. In vitro evidence suggests that visual arrestin (ARR1) binding to light-activated, phosphorylated rhodopsin hinders dephosphorylation. Whether ARR1 binding also affects rhodopsin dephosphorylation in vivo is not known. We investigated this using both male and female mice lacking ARR1. Mice were exposed to bright light and placed in darkness for different periods of time, and differently phosphorylated species of rhodopsin were assayed by isoelectric focusing. For WT mice, rhodopsin dephosphorylation was nearly complete by 1 h in darkness. Surprisingly, we observed that, in the Arr1 KO rods, rhodopsin remained phosphorylated even after 3 h. Delayed dephosphorylation in Arr1 KO rods cannot be explained by cell stress induced by persistent signaling, since it is not prevented by the removal of transducin, the visual G-protein, nor can it be explained by downregulation of protein phosphatase 2A, the putative rhodopsin phosphatase. We further show that cone arrestin (ARR4), which binds light-activated, phosphorylated rhodopsin poorly, had little effect in enhancing rhodopsin dephosphorylation, whereas mice expressing binding-competent mutant ARR1-3A showed a similar time course of rhodopsin dephosphorylation as WT. Together, these results reveal a novel role of ARR1 in facilitating rhodopsin dephosphorylation in vivoSIGNIFICANCE STATEMENT G-protein-coupled receptors (GPCRs) are transmembrane proteins used by cells to receive and respond to a broad range of extracellular signals that include neurotransmitters, hormones, odorants, and light (photons). GPCR signaling is terminated by two sequential steps: phosphorylation and arrestin binding. Both steps must be reversed when GPCRs are recycled and reused. Dephosphorylation, which is required for recycling, is an understudied process. Using rhodopsin as a prototypical GPCR, we discovered that arrestin facilitated rhodopsin dephosphorylation in living mice.
Collapse
Affiliation(s)
- Chia-Ling Hsieh
- Ziliha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California 90089
| | - Yun Yao
- Ziliha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California 90089
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Jeannie Chen
- Ziliha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
6
|
Kawamura S, Tachibanaki S. Molecular basis of rod and cone differences. Prog Retin Eye Res 2021; 90:101040. [PMID: 34974196 DOI: 10.1016/j.preteyeres.2021.101040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022]
Abstract
In the vertebrate retina, rods and cones both detect light, but they are different in functional aspects such as light sensitivity and time resolution, for example, and in some of cell biological aspects. For functional aspects, both photoreceptors are known to share a common mechanism, phototransduction cascade, consisting of a series of enzyme reactions to convert a photon-capture signal to an electrical signal. To understand the mechanisms of the functional differences between rods and cones at the molecular level, we compared biochemically each of the reactions in the phototransduction cascade between rods and cones using the cells isolated and purified from carp retina. Although proteins in the cascade are functionally similar between rods and cones, their activities together with their expression levels are mostly different between these photoreceptors. In general, reactions to generate a response are slightly less effective, as a total, in cones than in rods, but each of the reactions for termination and recovery of a response are much more effective in cones. These findings explain lower light sensitivity and briefer light responses in cones than in rods. In addition, our considerations suggest that a Ca2+-binding protein, S-modulin or recoverin, has a currently unnoticed role in shaping light responses. With comparison of the expression levels of proteins and/or mRNAs using purified cells, several proteins were found to be specifically or predominantly expressed in cones. These proteins would be of interest for future studies on the difference between rods and cones.
Collapse
Affiliation(s)
- Satoru Kawamura
- Graduate School of Frontier Biosciences, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan; Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan.
| | - Shuji Tachibanaki
- Graduate School of Frontier Biosciences, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan; Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
7
|
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: 8] [Impact Index Per Article: 2.0] [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
|
8
|
Chaudhary PK, Kim S. The GRKs Reactome: Role in Cell Biology and Pathology. Int J Mol Sci 2021; 22:ijms22073375. [PMID: 33806057 PMCID: PMC8036551 DOI: 10.3390/ijms22073375] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptor kinases (GRKs) are protein kinases that function in concert with arrestins in the regulation of a diverse class of G protein-coupled receptors (GPCRs) signaling. Although GRKs and arrestins are key participants in the regulation of GPCR cascades, the complex regulatory mechanisms of GRK expression, its alternation, and their function are not thoroughly understood. Several studies together with the work from our lab in recent years have revealed the critical role of these kinases in various physiological and pathophysiological processes, including cardiovascular biology, inflammation and immunity, neurodegeneration, thrombosis, and hemostasis. A comprehensive understanding of the mechanisms underlying functional interactions with multiple receptor proteins and how these interactions take part in the development of various pathobiological processes may give rise to novel diagnostic and therapeutic strategies. In this review, we summarize the current research linking the role of GRKs to various aspects of cell biology, pathology, and therapeutics, with a particular focus on thrombosis and hemostasis.
Collapse
|
9
|
Gurevich EV, Gurevich VV. GRKs as Modulators of Neurotransmitter Receptors. Cells 2020; 10:cells10010052. [PMID: 33396400 PMCID: PMC7823573 DOI: 10.3390/cells10010052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Many receptors for neurotransmitters, such as dopamine, norepinephrine, acetylcholine, and neuropeptides, belong to the superfamily of G protein-coupled receptors (GPCRs). A general model posits that GPCRs undergo two-step homologous desensitization: the active receptor is phosphorylated by kinases of the G protein-coupled receptor kinase (GRK) family, whereupon arrestin proteins specifically bind active phosphorylated receptors, shutting down G protein-mediated signaling, facilitating receptor internalization, and initiating distinct signaling pathways via arrestin-based scaffolding. Here, we review the mechanisms of GRK-dependent regulation of neurotransmitter receptors, focusing on the diverse modes of GRK-mediated phosphorylation of receptor subtypes. The immediate signaling consequences of GRK-mediated receptor phosphorylation, such as arrestin recruitment, desensitization, and internalization/resensitization, are equally diverse, depending not only on the receptor subtype but also on phosphorylation by GRKs of select receptor residues. We discuss the signaling outcome as well as the biological and behavioral consequences of the GRK-dependent phosphorylation of neurotransmitter receptors where known.
Collapse
|
10
|
Suwanmajo T, Ramesh V, Krishnan J. Exploring cyclic networks of multisite modification reveals origins of information processing characteristics. Sci Rep 2020; 10:16542. [PMID: 33024185 PMCID: PMC7539153 DOI: 10.1038/s41598-020-73045-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022] Open
Abstract
Multisite phosphorylation (and generally multisite modification) is a basic way of encoding substrate function and circuits/networks of post-translational modifications (PTM) are ubiquitous in cell signalling. The information processing characteristics of PTM systems are a focal point of broad interest. The ordering of modifications is a key aspect of multisite modification, and a broad synthesis of the impact of ordering of modifications is still missing. We focus on a basic class of multisite modification circuits: the cyclic mechanism, which corresponds to the same ordering of phosphorylation and dephosphorylation, and examine multiple variants involving common/separate kinases and common/separate phosphatases. This is of interest both because it is encountered in concrete cellular contexts, and because it serves as a bridge between ordered (sequential) mechanisms (representing one type of ordering) and random mechanisms (which have no ordering). We show that bistability and biphasic dose response curves of the maximally modified phosphoform are ruled out for basic structural reasons independent of parameters, while oscillations can result with even just one shared enzyme. We then examine the effect of relaxing some basic assumptions about the ordering of modification. We show computationally and analytically how bistability, biphasic responses and oscillations can be generated by minimal augmentations to the cyclic mechanism even when these augmentations involved reactions operating in the unsaturated limit. All in all, using this approach we demonstrate (1) how the cyclic mechanism (with single augmentations) represents a modification circuit using minimal ingredients (in terms of shared enzymes and sequestration of enzymes) to generate bistability and oscillations, when compared to other mechanisms, (2) new design principles for rationally designing PTM systems for a variety of behaviour, (3) a basis and a necessary step for understanding the origins and robustness of behaviour observed in basic multisite modification systems.
Collapse
Affiliation(s)
- Thapanar Suwanmajo
- Center of Excellence in Materials Science and Technology, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Vaidhiswaran Ramesh
- Department of Chemical Engineering, Centre for Process Systems Engineering, Imperial College London, London, SW7 2AZ, UK
| | - J Krishnan
- Department of Chemical Engineering, Centre for Process Systems Engineering, Imperial College London, London, SW7 2AZ, UK.
- Institute for Systems and Synthetic Biology, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
11
|
Abstract
We have used recent measurements of mammalian cone light responses and voltage-gated currents to calculate cone ATP utilization and compare it to that of rods. The largest expenditure of ATP results from ion transport, particularly from removal of Na+ entering outer segment light-dependent channels and inner segment hyperpolarization-activated cyclic nucleotide-gated channels, and from ATP-dependent pumping of Ca2+ entering voltage-gated channels at the synaptic terminal. Single cones expend nearly twice as much energy as single rods in darkness, largely because they make more synapses with second-order retinal cells and thus must extrude more Ca2+ In daylight, cone ATP utilization per cell remains high because cones never remain saturated and must continue to export Na+ and synaptic Ca2+ even in bright illumination. In mouse and human retina, rods greatly outnumber cones and consume more energy overall even in background light. In primates, however, the high density of cones in the fovea produces a pronounced peak of ATP utilization, which becomes particularly prominent in daylight and may make this part of the retina especially sensitive to changes in energy availability.
Collapse
|
12
|
Valdez-Lopez JC, Gulati S, Ortiz EA, Palczewski K, Robinson PR. Melanopsin Carboxy-terminus phosphorylation plasticity and bulk negative charge, not strict site specificity, achieves phototransduction deactivation. PLoS One 2020; 15:e0228121. [PMID: 32236094 PMCID: PMC7112210 DOI: 10.1371/journal.pone.0228121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/06/2020] [Indexed: 11/19/2022] Open
Abstract
Melanopsin is a visual pigment expressed in a small subset of ganglion cells in the mammalian retina known as intrinsically photosensitive retinal ganglion cells (ipRGCs) and is implicated in regulating non-image forming functions such as circadian photoentrainment and pupil constriction and contrast sensitivity in image formation. Mouse melanopsin's Carboxy-terminus (C-terminus) possesses 38 serine and threonine residues, which can potentially serve as phosphorylation sites for a G-protein Receptor Kinase (GRK) and be involved in the deactivation of signal transduction. Previous studies suggest that S388, T389, S391, S392, S394, S395 on the proximal region of the C-terminus of mouse melanopsin are necessary for melanopsin deactivation. We expressed a series of mouse melanopsin C-terminal mutants in HEK293 cells and using calcium imaging, and we found that the necessary cluster of six serine and threonine residues, while being critical, are insufficient for proper melanopsin deactivation. Interestingly, the additional six serine and threonine residues adjacent to the required six sites, in either proximal or distal direction, are capable of restoring wild-type deactivation of melanopsin. These findings suggest an element of plasticity in the molecular basis of melanopsin phosphorylation and deactivation. In addition, C-terminal chimeric mutants and molecular modeling studies support the idea that the initial steps of deactivation and β-arrestin binding are centered around these critical phosphorylation sites (S388-S395). The degree of functional versatility described in this study, along with ipRGC biophysical heterogeneity and the possible use of multiple signal transduction cascades, might contribute to the diverse ipRGC light responses for use in non-image and image forming behaviors, even though all six sub types of ipRGCs express the same melanopsin gene OPN4.
Collapse
MESH Headings
- HEK293 Cells
- Humans
- Light Signal Transduction/physiology
- Models, Molecular
- Mutagenesis, Site-Directed
- Mutation
- Phosphorylation/physiology
- Protein Binding
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Rod Opsins/chemistry
- Rod Opsins/genetics
- Rod Opsins/metabolism
- Serine/genetics
- Serine/metabolism
- Threonine/genetics
- Threonine/metabolism
- beta-Arrestin 1/chemistry
- beta-Arrestin 1/metabolism
Collapse
Affiliation(s)
- Juan C. Valdez-Lopez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Sahil Gulati
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California, United States of America
- Gatan Inc, Pleasanton, California, United States of America
| | - Elelbin A. Ortiz
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Krzysztof Palczewski
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California, United States of America
| | - Phyllis R. Robinson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| |
Collapse
|
13
|
Lokappa SB, Cornwall MC, Chen J. Isoelectric Focusing to Quantify Rhodopsin Phosphorylation in Mouse Retina. Bio Protoc 2019; 9:e3300. [PMID: 33654813 PMCID: PMC7854240 DOI: 10.21769/bioprotoc.3300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 11/02/2022] Open
Abstract
Rhodopsin is a G-protein coupled receptor (GPCR) that mediates vision under dim light. Upon light exposure, rhodopsin is phosphorylated at multiple serine and threonine sites at its carboxyl-terminus by rhodopsin kinase (GRK1). This, in turn, reduces its ability to activate the visual G-protein transducin. Binding of light-activated, phosphorylated rhodopsin by arrestin (ARR1) fully terminates the catalytic activity of rhodopsin. Quantification of the levels of the differentially phosphorylated rhodopsin species provides definitive information about the role of phosphorylated rhodopsin in visual functions. Isoelectric Focusing (IEF) is a technique which is used to separate ampholytic components, such as proteins, based on their isoelectric point (pI). It is a useful technique used to distinguish protein isoforms and post-translational modifications such as phosphorylation, glycosylation, deamination, and acetylation, due to their effects on the protein's pI. Isoelectric Focusing can provide high resolution of differentially phosphorylated forms of a protein. Though other techniques such as kinase activity assays, phospho-specific antibodies, western blot, enzyme-linked immunosorbent assays (ELISA), radiolabeling and mass spectrometry are used to detect and quantify protein phosphorylation, IEF is a simple and cost-effective method to quantify rhodopsin phosphorylation, as it can readily detect individual phosphorylated forms. Here we provide a detailed protocol for determining phosphorylated rhodopsin species using the Isoelectric Focusing technique.
Collapse
Affiliation(s)
- Sowmya Bekshe Lokappa
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - M. Carter Cornwall
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jeannie Chen
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
14
|
Mayer D, Damberger FF, Samarasimhareddy M, Feldmueller M, Vuckovic Z, Flock T, Bauer B, Mutt E, Zosel F, Allain FHT, Standfuss J, Schertler GFX, Deupi X, Sommer ME, Hurevich M, Friedler A, Veprintsev DB. Distinct G protein-coupled receptor phosphorylation motifs modulate arrestin affinity and activation and global conformation. Nat Commun 2019; 10:1261. [PMID: 30890705 PMCID: PMC6424980 DOI: 10.1038/s41467-019-09204-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular functions of arrestins are determined in part by the pattern of phosphorylation on the G protein-coupled receptors (GPCRs) to which arrestins bind. Despite high-resolution structural data of arrestins bound to phosphorylated receptor C-termini, the functional role of each phosphorylation site remains obscure. Here, we employ a library of synthetic phosphopeptide analogues of the GPCR rhodopsin C-terminus and determine the ability of these peptides to bind and activate arrestins using a variety of biochemical and biophysical methods. We further characterize how these peptides modulate the conformation of arrestin-1 by nuclear magnetic resonance (NMR). Our results indicate different functional classes of phosphorylation sites: 'key sites' required for arrestin binding and activation, an 'inhibitory site' that abrogates arrestin binding, and 'modulator sites' that influence the global conformation of arrestin. These functional motifs allow a better understanding of how different GPCR phosphorylation patterns might control how arrestin functions in the cell.
Collapse
Affiliation(s)
- Daniel Mayer
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland.
- Department of Biology, ETH Zürich, 8093, Zürich, Switzerland.
- Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, 92093-0636, California, USA.
| | | | | | - Miki Feldmueller
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
- Department of Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Ziva Vuckovic
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
- Department of Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Tilman Flock
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
- Department of Biology, ETH Zürich, 8093, Zürich, Switzerland
- Fitzwilliam College, Cambridge, CB3 0DG, UK
| | - Brian Bauer
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Eshita Mutt
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
| | | | | | - Jörg Standfuss
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
| | - Gebhard F X Schertler
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
- Department of Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Xavier Deupi
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland
- Condensed Matter Theory, Paul Scherrer Institute, 5232, Villigen, Switzerland
| | - Martha E Sommer
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Mattan Hurevich
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Assaf Friedler
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dmitry B Veprintsev
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232, Villigen, Switzerland.
- Department of Biology, ETH Zürich, 8093, Zürich, Switzerland.
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, NG7 2RD, UK.
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
15
|
Knockout of zebrafish interleukin 7 receptor (IL7R) by the CRISPR/Cas9 system delays retinal neurodevelopment. Cell Death Dis 2018; 9:273. [PMID: 29449560 PMCID: PMC5833684 DOI: 10.1038/s41419-018-0337-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/04/2018] [Accepted: 01/22/2018] [Indexed: 11/24/2022]
Abstract
Interleukin 7 receptor (il7r), a transmembrane receptor, belongs to the type I cytokine receptor family. Il7r is involved in the pathogenesis of neurodegenerative disorders, such as multiple sclerosis. Targeted knockdown of il7r leads to delayed myelination, highlighting the potential role of il7r in the development of the nervous system. Zebrafish is an ideal model for the study of neurogenesis; moreover, the il7r gene is highly conserved between zebrafish and human. The aim of the present study was to investigate the novel function of il7r in neurogenesis. First, an il7r−/− homozygous mutant line was generated by clustered regularly interspaced short palindromic repeats (CRISPR)-associated 9 (CRISPR/Cas9) technology. Second, the gross development of il7r−/− mutants revealed remarkably smaller eyes and delayed retinal neurodifferentiation. Third, microarray analysis revealed that genes associated with the phototransduction signalling pathway were strongly down-regulated in il7r−/− mutants. Finally, the results from behavioural tests indicated that visual function was impaired in il7r−/− mutant larvae. Overall, our data demonstrate that a lack of il7r retards the development of the retina. Thus, il7r is an essential molecule for maintaining normal retinal development in zebrafish.
Collapse
|
16
|
Dephosphorylation by protein phosphatase 2A regulates visual pigment regeneration and the dark adaptation of mammalian photoreceptors. Proc Natl Acad Sci U S A 2017; 114:E9675-E9684. [PMID: 29078372 DOI: 10.1073/pnas.1712405114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Resetting of G-protein-coupled receptors (GPCRs) from their active state back to their biologically inert ground state is an integral part of GPCR signaling. This "on-off" GPCR cycle is regulated by reversible phosphorylation. Retinal rod and cone photoreceptors arguably represent the best-understood example of such GPCR signaling. Their visual pigments (opsins) are activated by light, transduce the signal, and are then inactivated by a GPCR kinase and arrestin. Although pigment inactivation by phosphorylation is well understood, the enzyme(s) responsible for pigment dephosphorylation and the functional significance of this reaction remain unknown. Here, we show that protein phosphatase 2A (PP2A) acts as opsin phosphatase in both rods and cones. Elimination of PP2A substantially slows pigment dephosphorylation, visual chromophore recycling, and ultimately photoreceptor dark adaptation. These findings demonstrate that visual pigment dephosphorylation regulates the dark adaptation of photoreceptors and provide insights into the role of this reaction in GPCR signaling.
Collapse
|
17
|
Effect of Rhodopsin Phosphorylation on Dark Adaptation in Mouse Rods. J Neurosci 2017; 36:6973-87. [PMID: 27358455 DOI: 10.1523/jneurosci.3544-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 05/17/2016] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Rhodopsin is a prototypical G-protein-coupled receptor (GPCR) that is activated when its 11-cis-retinal moiety is photoisomerized to all-trans retinal. This step initiates a cascade of reactions by which rods signal changes in light intensity. Like other GPCRs, rhodopsin is deactivated through receptor phosphorylation and arrestin binding. Full recovery of receptor sensitivity is then achieved when rhodopsin is regenerated through a series of steps that return the receptor to its ground state. Here, we show that dephosphorylation of the opsin moiety of rhodopsin is an extremely slow but requisite step in the restoration of the visual pigment to its ground state. We make use of a novel observation: isolated mouse retinae kept in standard media for routine physiologic recordings display blunted dephosphorylation of rhodopsin. Isoelectric focusing followed by Western blot analysis of bleached isolated retinae showed little dephosphorylation of rhodopsin for up to 4 h in darkness, even under conditions when rhodopsin was completely regenerated. Microspectrophotometeric determinations of rhodopsin spectra show that regenerated phospho-rhodopsin has the same molecular photosensitivity as unphosphorylated rhodopsin and that flash responses measured by trans-retinal electroretinogram or single-cell suction electrode recording displayed dark-adapted kinetics. Single quantal responses displayed normal dark-adapted kinetics, but rods were only half as sensitive as those containing exclusively unphosphorylated rhodopsin. We propose a model in which light-exposed retinae contain a mixed population of phosphorylated and unphosphorylated rhodopsin. Moreover, complete dark adaptation can only occur when all rhodopsin has been dephosphorylated, a process that requires >3 h in complete darkness. SIGNIFICANCE STATEMENT G-protein-coupled receptors (GPCRs) constitute the largest superfamily of proteins that compose ∼4% of the mammalian genome whose members share a common membrane topology. Signaling by GPCRs regulate a wide variety of physiological processes, including taste, smell, hearing, vision, and cardiovascular, endocrine, and reproductive homeostasis. An important feature of GPCR signaling is its timely termination. This normally occurs when, after their activation, GPCRs are rapidly phosphorylated by specific receptor kinases and subsequently bound by cognate arrestins. Recovery of receptor sensitivity to the ground state then requires dephosphorylation of the receptor and unbinding of arrestin, processes that are poorly understood. Here we investigate in mouse rod photoreceptors the relationship between rhodopsin dephosphorylation and recovery of visual sensitivity.
Collapse
|
18
|
Van Cauwenbergh C, Coppieters F, Roels D, De Jaegere S, Flipts H, De Zaeytijd J, Walraedt S, Claes C, Fransen E, Van Camp G, Depasse F, Casteels I, de Ravel T, Leroy BP, De Baere E. Mutations in Splicing Factor Genes Are a Major Cause of Autosomal Dominant Retinitis Pigmentosa in Belgian Families. PLoS One 2017; 12:e0170038. [PMID: 28076437 PMCID: PMC5226823 DOI: 10.1371/journal.pone.0170038] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 12/27/2016] [Indexed: 12/14/2022] Open
Abstract
Purpose Autosomal dominant retinitis pigmentosa (adRP) is characterized by an extensive genetic heterogeneity, implicating 27 genes, which account for 50 to 70% of cases. Here 86 Belgian probands with possible adRP underwent genetic testing to unravel the molecular basis and to assess the contribution of the genes underlying their condition. Methods Mutation detection methods evolved over the past ten years, including mutation specific methods (APEX chip analysis), linkage analysis, gene panel analysis (Sanger sequencing, targeted next-generation sequencing or whole exome sequencing), high-resolution copy number screening (customized microarray-based comparative genomic hybridization). Identified variants were classified following American College of Medical Genetics and Genomics (ACMG) recommendations. Results Molecular genetic screening revealed mutations in 48/86 cases (56%). In total, 17 novel pathogenic mutations were identified: four missense mutations in RHO, five frameshift mutations in RP1, six mutations in genes encoding spliceosome components (SNRNP200, PRPF8, and PRPF31), one frameshift mutation in PRPH2, and one frameshift mutation in TOPORS. The proportion of RHO mutations in our cohort (14%) is higher than reported in a French adRP population (10.3%), but lower than reported elsewhere (16.5–30%). The prevalence of RP1 mutations (10.5%) is comparable to other populations (3.5%-10%). The mutation frequency in genes encoding splicing factors is unexpectedly high (altogether 19.8%), with PRPF31 the second most prevalent mutated gene (10.5%). PRPH2 mutations were found in 4.7% of the Belgian cohort. Two families (2.3%) have the recurrent NR2E3 mutation p.(Gly56Arg). The prevalence of the recurrent PROM1 mutation p.(Arg373Cys) was higher than anticipated (3.5%). Conclusions Overall, we identified mutations in 48 of 86 Belgian adRP cases (56%), with the highest prevalence in RHO (14%), RP1 (10.5%) and PRPF31 (10.5%). Finally, we expanded the molecular spectrum of PRPH2, PRPF8, RHO, RP1, SNRNP200, and TOPORS-associated adRP by the identification of 17 novel mutations.
Collapse
Affiliation(s)
- Caroline Van Cauwenbergh
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Frauke Coppieters
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Dimitri Roels
- Department of Ophthalmology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Sarah De Jaegere
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Helena Flipts
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
- Center for Human Genetics, University Hospitals Leuven, Louvain, Belgium
| | - Julie De Zaeytijd
- Department of Ophthalmology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Sophie Walraedt
- Department of Ophthalmology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Charlotte Claes
- Center for Medical Genetics Antwerp, Antwerp University, Antwerp, Belgium
| | - Erik Fransen
- Center for Medical Genetics Antwerp, Antwerp University, Antwerp, Belgium
| | - Guy Van Camp
- Center for Medical Genetics Antwerp, Antwerp University, Antwerp, Belgium
| | - Fanny Depasse
- Department of Ophthalmology, Hôpital Erasme-ULB, Brussels, Belgium
| | - Ingele Casteels
- Department of Ophthalmology, University Hospitals Leuven, Louvain, Belgium
| | - Thomy de Ravel
- Center for Human Genetics, University Hospitals Leuven, Louvain, Belgium
| | - Bart P. Leroy
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
- Department of Ophthalmology, Ghent University and Ghent University Hospital, Ghent, Belgium
- Division of Ophthalmology & Center for Cellular & Molecular Therapy, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Elfride De Baere
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
- * E-mail:
| |
Collapse
|
19
|
|
20
|
Iwamoto N, D'Alessandro LA, Depner S, Hahn B, Kramer BA, Lucarelli P, Vlasov A, Stepath M, Böhm ME, Deharde D, Damm G, Seehofer D, Lehmann WD, Klingmüller U, Schilling M. Context-specific flow through the MEK/ERK module produces cell- and ligand-specific patterns of ERK single and double phosphorylation. Sci Signal 2016; 9:ra13. [PMID: 26838549 DOI: 10.1126/scisignal.aab1967] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The same pathway, such as the mitogen-activated protein kinase (MAPK) pathway, can produce different cellular responses, depending on stimulus or cell type. We examined the phosphorylation dynamics of the MAPK kinase MEK and its targets extracellular signal-regulated kinase 1 and 2 (ERK1/2) in primary hepatocytes and the transformed keratinocyte cell line HaCaT A5 exposed to either hepatocyte growth factor or interleukin-6. By combining quantitative mass spectrometry with dynamic modeling, we elucidated network structures for the reversible threonine and tyrosine phosphorylation of ERK in both cell types. In addition to differences in the phosphorylation and dephosphorylation reactions, the HaCaT network model required two feedback mechanisms, which, as the experimental data suggested, involved the induction of the dual-specificity phosphatase DUSP6 and the scaffold paxillin. We assayed and modeled the accumulation of the double-phosphorylated and active form of ERK1/2, as well as the dynamics of the changes in the monophosphorylated forms of ERK1/2. Modeling the differences in the dynamics of the changes in the distributions of the phosphorylated forms of ERK1/2 suggested that different amounts of MEK activity triggered context-specific responses, with primary hepatocytes favoring the formation of double-phosphorylated ERK1/2 and HaCaT A5 cells that produce both the threonine-phosphorylated and the double-phosphorylated form. These differences in phosphorylation distributions explained the threshold, sensitivity, and saturation of the ERK response. We extended the findings of differential ERK phosphorylation profiles to five additional cultured cell systems and matched liver tumor and normal tissue, which revealed context-specific patterns of the various forms of phosphorylated ERK.
Collapse
Affiliation(s)
- Nao Iwamoto
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Lorenza A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sofia Depner
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Bettina Hahn
- Molecular Structure Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Bernhard A Kramer
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Philippe Lucarelli
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Artyom Vlasov
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Markus Stepath
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Martin E Böhm
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Daniela Deharde
- Department of General, Visceral and Transplantation Surgery, Campus Virchow Clinic, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg Damm
- Department of General, Visceral and Transplantation Surgery, Campus Virchow Clinic, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Daniel Seehofer
- Department of General, Visceral and Transplantation Surgery, Campus Virchow Clinic, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Wolf D Lehmann
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Molecular Structure Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Marcel Schilling
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
21
|
Reingruber J, Holcman D, Fain GL. How rods respond to single photons: Key adaptations of a G-protein cascade that enable vision at the physical limit of perception. Bioessays 2015; 37:1243-52. [PMID: 26354340 DOI: 10.1002/bies.201500081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Rod photoreceptors are among the most sensitive light detectors in nature. They achieve their remarkable sensitivity across a wide variety of species through a number of essential adaptations: a specialized cellular geometry, a G-protein cascade with an unusually stable receptor molecule, a low-noise transduction mechanism, a nearly perfect effector enzyme, and highly evolved mechanisms of feedback control and receptor deactivation. Practically any change in protein expression, enzyme activity, or feedback control can be shown to impair photon detection, either by decreasing sensitivity or signal-to-noise ratio, or by reducing temporal resolution. Comparison of mammals to amphibians suggests that rod outer-segment morphology and the molecules and mechanism of transduction may have evolved together to optimize light sensitivity in darkness, which culminates in the extraordinary ability of these cells to respond to single photons at the ultimate limit of visual perception.
Collapse
Affiliation(s)
- Jürgen Reingruber
- IBENS, Group of Computational Biology and Applied Mathematics, École Normale Supérieure, Paris, France.,INSERM U1024, Paris, France
| | - David Holcman
- IBENS, Group of Computational Biology and Applied Mathematics, École Normale Supérieure, Paris, France.,Department of Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
| | - Gordon L Fain
- Department of Integrative Biology and Physiology, Terasaki Life Sciences, University of California, Los Angeles, CA, USA.,Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Yamaoka H, Tachibanaki S, Kawamura S. Dephosphorylation during bleach and regeneration of visual pigment in carp rod and cone membranes. J Biol Chem 2015; 290:24381-90. [PMID: 26286749 DOI: 10.1074/jbc.m115.674101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
On absorption of light by vertebrate visual pigment, the chromophore, 11-cis retinal, is isomerized to all-trans retinal to activate the phototransduction cascade, which leads to a hyperpolarizing light response. Activated pigment is inactivated by phosphorylation on the protein moiety, opsin. Isomerized all-trans retinal is ultimately released from opsin, and the pigment is regenerated by binding to 11-cis retinal. In this pigment regeneration cycle, the phosphates incorporated should be removed in order that the pigment regains the capability of activating the phototransduction cascade. However, it is not clear yet how pigment dephosphorylation takes place in the regeneration cycle. First in this study, we tried to estimate the dephosphorylation activity in living carp rods and cones and found that the activity, which is present mainly in the cytoplasm in both rods and cones, is three times higher in cones than in rods. Second, we examined at which stage the dephosphorylation takes place; before or after the release of all-trans retinal, during pigment regeneration, or after pigment regeneration. For this purpose we prepared three types of phosphorylated substrates in purified carp rod and cone membranes: phosphorylated bleaching intermediate, phosphorylated opsin, and phosphorylated and regenerated pigment. We also examined the effect of pigment regeneration on the dephosphorylation. The results showed that the dephosphorylation does not show substrate preference in the regeneration cycle and suggested that the dephosphorylation takes place constantly. The results also suggest that, under bright light, some of the regenerated visual pigment remains phosphorylated to reduce the light sensitivity in cones.
Collapse
Affiliation(s)
| | - Shuji Tachibanaki
- From the Graduate School of Frontier Biosciences and the Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka 565-0871, Japan
| | - Satoru Kawamura
- From the Graduate School of Frontier Biosciences and the Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka 565-0871, Japan
| |
Collapse
|
23
|
Azevedo AW, Doan T, Moaven H, Sokal I, Baameur F, Vishnivetskiy SA, Homan KT, Tesmer JJG, Gurevich VV, Chen J, Rieke F. C-terminal threonines and serines play distinct roles in the desensitization of rhodopsin, a G protein-coupled receptor. eLife 2015; 4. [PMID: 25910054 PMCID: PMC4438306 DOI: 10.7554/elife.05981] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/23/2015] [Indexed: 12/31/2022] Open
Abstract
Rod photoreceptors generate measurable responses to single-photon activation of individual molecules of the G protein-coupled receptor (GPCR), rhodopsin. Timely rhodopsin desensitization depends on phosphorylation and arrestin binding, which quenches G protein activation. Rhodopsin phosphorylation has been measured biochemically at C-terminal serine residues, suggesting that these residues are critical for producing fast, low-noise responses. The role of native threonine residues is unclear. We compared single-photon responses from rhodopsin lacking native serine or threonine phosphorylation sites. Contrary to expectation, serine-only rhodopsin generated prolonged step-like single-photon responses that terminated abruptly and randomly, whereas threonine-only rhodopsin generated responses that were only modestly slower than normal. We show that the step-like responses of serine-only rhodopsin reflect slow and stochastic arrestin binding. Thus, threonine sites play a privileged role in promoting timely arrestin binding and rhodopsin desensitization. Similar coordination of phosphorylation and arrestin binding may more generally permit tight control of the duration of GPCR activity. DOI:http://dx.doi.org/10.7554/eLife.05981.001 ‘Rod’ cells in the eye enable us to see in starlight. Inside these cells, a protein called rhodopsin is activated by light, which leads to an electrical signal being produced that travels to the brain. The duration of the electrical signal depends on the time it takes for the rhodopsin to be deactivated. Rhodopsin is a member of a large class of receptor proteins known as G protein-coupled receptors that regulate many processes throughout the body. Previous studies have shown that rhodopsin is deactivated by the attachment of phosphate groups to the protein. This allows another protein called arrestin to bind to rhodopsin. The phosphates can be attached to particular amino acids—the building blocks of proteins—at one end of rhodopsin. Three of these are a type of amino acid called serine. Previous work has shown that light increases the speed at which phosphate groups are added to these serines, suggesting that they are important for producing rapid electrical signals. The other three amino acids are of a different type—called threonine—but it is less clear what role they play in deactivating rhodopsin. Here, Azevedo et al. studied mutant forms of rhodopsin that were missing the serines or threonines in mice. The experiments show that loss of the serines only slightly slowed the electrical signals. However, loss of the threonines resulted in much slower electrical signals that ended at random times. This was due to rhodopsin being less able to bind to arrestin. Azevedo et al. propose a new model for how rhodopsin is deactivated. Once light activates the protein, phosphate groups are rapidly added to the serines, which begins to lower the activity of rhodopsin. However, it is the slower addition of phosphates to the threonines that is essential to promote arrestin binding and fully deactivate the protein. Other proteins belonging to the G protein-coupled receptor family also have these serines and threonines, and thus, may be regulated in a similar way. DOI:http://dx.doi.org/10.7554/eLife.05981.002
Collapse
Affiliation(s)
- Anthony W Azevedo
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Thuy Doan
- Department of Ophthalmology, University of Washington, Seattle, United States
| | - Hormoz Moaven
- Departments of Cell & Neurobiology and Ophthalmology, Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, United States
| | - Iza Sokal
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Faiza Baameur
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, United States
| | - Sergey A Vishnivetskiy
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, United States
| | - Kristoff T Homan
- Life Sciences Institute, Departments of Pharmacology and Biological Chemistry, University of Michigan, Ann Arbor, United States
| | - John J G Tesmer
- Life Sciences Institute, Departments of Pharmacology and Biological Chemistry, University of Michigan, Ann Arbor, United States
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, United States
| | - Jeannie Chen
- Departments of Cell & Neurobiology and Ophthalmology, Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, United States
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| |
Collapse
|
24
|
Tomizuka J, Tachibanaki S, Kawamura S. Phosphorylation-independent suppression of light-activated visual pigment by arrestin in carp rods and cones. J Biol Chem 2015; 290:9399-411. [PMID: 25713141 DOI: 10.1074/jbc.m114.634543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Indexed: 01/25/2023] Open
Abstract
Visual pigment in photoreceptors is activated by light. Activated visual pigment (R*) is believed to be inactivated by phosphorylation of R* with subsequent binding of arrestin. There are two types of photoreceptors, rods and cones, in the vertebrate retina, and they express different subtypes of arrestin, rod and cone type. To understand the difference in the function between rod- and cone-type arrestin, we first identified the subtype of arrestins expressed in rods and cones in carp retina. We found that two rod-type arrestins, rArr1 and rArr2, are co-expressed in a rod and that a cone-type arrestin, cArr1, is expressed in blue- and UV-sensitive cones; the other cone-type arrestin, cArr2, is expressed in red- and green-sensitive cones. We quantified each arrestin subtype and estimated its concentration in the outer segment of a rod or a cone in the dark; they were ∼0.25 mm (rArr1 plus rArr2) in a rod and 0.6-0.8 mm (cArr1 or cArr2) in a cone. The effect of each arrestin was examined. In contrast to previous studies, both rod and cone arrestins suppressed the activation of transducin in the absence of visual pigment phosphorylation, and all of the arrestins examined (rArr1, rArr2, and cArr2) bound transiently to most probably nonphosphorylated R*. One rod arrestin, rArr2, bound firmly to phosphorylated pigment, and the other two, rArr1 and cArr2, once bound to phosphorylated R* but dissociated from it during incubation. Our results suggested a novel mechanism of arrestin effect on the suppression of the R* activity in both rods and cones.
Collapse
Affiliation(s)
| | - Shuji Tachibanaki
- From the Graduate School of Frontier Biosciences, Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka 565-0871, Japan
| | - Satoru Kawamura
- From the Graduate School of Frontier Biosciences, Department of Biological Sciences, Graduate School of Science, Osaka University, Yamada-oka 1-3, Suita, Osaka 565-0871, Japan
| |
Collapse
|
25
|
Abstract
Rhodopsin is a key light-sensitive protein expressed exclusively in rod photoreceptor cells of the retina. Failure to express this transmembrane protein causes a lack of rod outer segment formation and progressive retinal degeneration, including the loss of cone photoreceptor cells. Molecular studies of rhodopsin have paved the way to understanding a large family of cell-surface membrane proteins called G protein-coupled receptors (GPCRs). Work started on rhodopsin over 100 years ago still continues today with substantial progress made every year. These activities underscore the importance of rhodopsin as a prototypical GPCR and receptor required for visual perception-the fundamental process of translating light energy into a biochemical cascade of events culminating in vision.
Collapse
Affiliation(s)
- Lukas Hofmann
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | | |
Collapse
|
26
|
Palczewski K. Chemistry and biology of the initial steps in vision: the Friedenwald lecture. Invest Ophthalmol Vis Sci 2014; 55:6651-72. [PMID: 25338686 DOI: 10.1167/iovs.14-15502] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Visual transduction is the process in the eye whereby absorption of light in the retina is translated into electrical signals that ultimately reach the brain. The first challenge presented by visual transduction is to understand its molecular basis. We know that maintenance of vision is a continuous process requiring the activation and subsequent restoration of a vitamin A-derived chromophore through a series of chemical reactions catalyzed by enzymes in the retina and retinal pigment epithelium (RPE). Diverse biochemical approaches that identified key proteins and reactions were essential to achieve a mechanistic understanding of these visual processes. The three-dimensional arrangements of these enzymes' polypeptide chains provide invaluable insights into their mechanisms of action. A wealth of information has already been obtained by solving high-resolution crystal structures of both rhodopsin and the retinoid isomerase from pigment RPE (RPE65). Rhodopsin, which is activated by photoisomerization of its 11-cis-retinylidene chromophore, is a prototypical member of a large family of membrane-bound proteins called G protein-coupled receptors (GPCRs). RPE65 is a retinoid isomerase critical for regeneration of the chromophore. Electron microscopy (EM) and atomic force microscopy have provided insights into how certain proteins are assembled to form much larger structures such as rod photoreceptor cell outer segment membranes. A second challenge of visual transduction is to use this knowledge to devise therapeutic approaches that can prevent or reverse conditions leading to blindness. Imaging modalities like optical coherence tomography (OCT) and scanning laser ophthalmoscopy (SLO) applied to appropriate animal models as well as human retinal imaging have been employed to characterize blinding diseases, monitor their progression, and evaluate the success of therapeutic agents. Lately two-photon (2-PO) imaging, together with biochemical assays, are revealing functional aspects of vision at a new molecular level. These multidisciplinary approaches combined with suitable animal models and inbred mutant species can be especially helpful in translating provocative cell and tissue culture findings into therapeutic options for further development in animals and eventually in humans. A host of different approaches and techniques is required for substantial progress in understanding fundamental properties of the visual system.
Collapse
Affiliation(s)
- Krzysztof Palczewski
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
27
|
Blasic JR, Matos-Cruz V, Ujla D, Cameron EG, Hattar S, Halpern ME, Robinson PR. Identification of critical phosphorylation sites on the carboxy tail of melanopsin. Biochemistry 2014; 53:2644-9. [PMID: 24678795 PMCID: PMC4010260 DOI: 10.1021/bi401724r] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
![]()
Light-activated
opsins undergo carboxy-terminal phosphorylation,
which contributes to the deactivation of their photoresponse. The
photopigment melanopsin possesses an unusually long carboxy tail containing
37 serine and threonine sites that are potential sites for phosphorylation
by a G-protein dependent kinase (GRK). Here, we show that a small
cluster of six to seven sites is sufficient for deactivation of light-activated
mouse melanopsin. Surprisingly, these sites are distinct from those
that regulate deactivation of rhodopsin. In zebrafish, there are five
different melanopsin genes that encode proteins with distinct carboxy-terminal
domains. Naturally occurring changes in the same cluster of phosphorylatable
amino acids provides diversity in the deactivation kinetics of the
zebrafish proteins. These results suggest that variation in phosphorylation
sites provides flexibility in the duration and kinetics of melanopsin-mediated
light responses.
Collapse
Affiliation(s)
- Joseph R Blasic
- Department of Biological Sciences, University of Maryland, Baltimore County , Baltimore, Maryland 21250, United States
| | | | | | | | | | | | | |
Collapse
|
28
|
Invergo BM, Dell'Orco D, Montanucci L, Koch KW, Bertranpetit J. A comprehensive model of the phototransduction cascade in mouse rod cells. MOLECULAR BIOSYSTEMS 2014; 10:1481-9. [PMID: 24675755 DOI: 10.1039/c3mb70584f] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Vertebrate visual phototransduction is perhaps the most well-studied G-protein signaling pathway. A wealth of available biochemical and electrophysiological data has resulted in a rich history of mathematical modeling of the system. However, while the most comprehensive models have relied upon amphibian biochemical and electrophysiological data, modern research typically employs mammalian species, particularly mice, which exhibit significantly faster signaling dynamics. In this work, we present an adaptation of a previously published, comprehensive model of amphibian phototransduction that can produce quantitatively accurate simulations of the murine photoresponse. We demonstrate the ability of the model to predict responses to a wide range of stimuli and under a variety of mutant conditions. Finally, we employ the model to highlight a likely unknown mechanism related to the interaction between rhodopsin and rhodopsin kinase.
Collapse
Affiliation(s)
- Brandon M Invergo
- IBE - Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), CEXS-UPF-PRBB, Barcelona, Catalonia, Spain
| | | | | | | | | |
Collapse
|
29
|
Sommer ME, Hofmann KP, Heck M. Not just signal shutoff: the protective role of arrestin-1 in rod cells. Handb Exp Pharmacol 2014; 219:101-16. [PMID: 24292826 DOI: 10.1007/978-3-642-41199-1_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The retinal rod cell is an exquisitely sensitive single-photon detector that primarily functions in dim light (e.g., moonlight). However, rod cells must routinely survive light intensities more than a billion times greater (e.g., bright daylight). One serious challenge to rod cell survival in daylight is the massive amount of all-trans-retinal that is released by Meta II, the light-activated form of the photoreceptor rhodopsin. All-trans-retinal is toxic, and its condensation products have been implicated in disease. Our recent work has developed the concept that rod arrestin (arrestin-1), which terminates Meta II signaling, has an additional role in protecting rod cells from the consequences of bright light by limiting free all-trans-retinal. In this chapter we will elaborate upon the molecular mechanisms by which arrestin-1 serves as both a single-photon response quencher as well as an instrument of rod cell survival in bright light. This discussion will take place within the framework of three distinct functional modules of vision: signal transduction, the retinoid cycle, and protein translocation.
Collapse
Affiliation(s)
- Martha E Sommer
- Institut für Medizinische Physik und Biophysik (CC2), Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany,
| | | | | |
Collapse
|
30
|
Abstract
G-protein-coupled receptors (GPCRs) are the primary interaction partners for arrestins. The visual arrestins, arrestin1 and arrestin4, physiologically bind to only very few receptors, i.e., rhodopsin and the color opsins, respectively. In contrast, the ubiquitously expressed nonvisual variants β-arrestin1 and 2 bind to a large number of receptors in a fairly nonspecific manner. This binding requires two triggers, agonist activation and receptor phosphorylation by a G-protein-coupled receptor kinase (GRK). These two triggers are mediated by two different regions of the arrestins, the "phosphorylation sensor" in the core of the protein and a less well-defined "activation sensor." Binding appears to occur mostly in a 1:1 stoichiometry, involving the N-terminal domain of GPCRs, but in addition a second GPCR may loosely bind to the C-terminal domain when active receptors are abundant.Arrestin binding initially uncouples GPCRs from their G-proteins. It stabilizes receptors in an active conformation and also induces a conformational change in the arrestins that involves a rotation of the two domains relative to each other plus changes in the polar core. This conformational change appears to permit the interaction with further downstream proteins. The latter interaction, demonstrated mostly for β-arrestins, triggers receptor internalization as well as a number of nonclassical signaling pathways.Open questions concern the exact stoichiometry of the interaction, possible specificity with regard to the type of agonist and of GRK involved, selective regulation of downstream signaling (=biased signaling), and the options to use these mechanisms as therapeutic targets.
Collapse
Affiliation(s)
- Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Straße 9, 97078, Würzburg, Germany,
| | | |
Collapse
|
31
|
Park PSH. Constitutively active rhodopsin and retinal disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 70:1-36. [PMID: 24931191 DOI: 10.1016/b978-0-12-417197-8.00001-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rhodopsin is the light receptor in rod photoreceptor cells of the retina that initiates scotopic vision. In the dark, rhodopsin is bound to the chromophore 11-cis retinal, which locks the receptor in an inactive state. The maintenance of an inactive rhodopsin in the dark is critical for rod photoreceptor cells to remain highly sensitive. Perturbations by mutation or the absence of 11-cis retinal can cause rhodopsin to become constitutively active, which leads to the desensitization of photoreceptor cells and, in some instances, retinal degeneration. Constitutive activity can arise in rhodopsin by various mechanisms and can cause a variety of inherited retinal diseases including Leber congenital amaurosis, congenital night blindness, and retinitis pigmentosa. In this review, the molecular and structural properties of different constitutively active forms of rhodopsin are overviewed, and the possibility that constitutive activity can arise from different active-state conformations is discussed.
Collapse
Affiliation(s)
- Paul Shin-Hyun Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
32
|
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
|
33
|
Saprunova VB, Lelekova MA, Kolosova NG, Bakeeva LE. SkQ1 slows development of age-dependent destructive processes in retina and vascular layer of eyes of wistar and OXYS rats. BIOCHEMISTRY (MOSCOW) 2012; 77:648-58. [PMID: 22817465 DOI: 10.1134/s0006297912060120] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We show the development of clearly pronounced age-related pathological changes in eye tissues of Wistar and OXYS rats. Photoreceptor cells were virtually absent in all OXYS rats in the age of 24 months. Massive accumulations of lipofuscin granules were detected in the pigmented epithelium cells. Flattening, overgrowing, and degradation of endothelial cells of choriocapillaries were also observed. Along with these changes, vessels without signs of degradation were detected in the pigmented epithelium. In 24-month-old Wistar rats these changes were local and were seen in only some of the animals. The mitochondria-targeted antioxidant SkQ1 (the rats were given SkQ1 daily with food at the dose of 250 nmol/kg for 5 months, starting from the age of 19 months) prevented the development of these pathological changes in both Wistar and OXYS rats. The data were subjected to mathematical processing and statistical analysis.
Collapse
Affiliation(s)
- V B Saprunova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia.
| | | | | | | |
Collapse
|
34
|
Distinct loops in arrestin differentially regulate ligand binding within the GPCR opsin. Nat Commun 2012; 3:995. [PMID: 22871814 PMCID: PMC3455371 DOI: 10.1038/ncomms2000] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 07/10/2012] [Indexed: 01/07/2023] Open
Abstract
G-protein-coupled receptors are universally regulated by arrestin binding. Here we show that rod arrestin induces uptake of the agonist all-trans-retinol in only half the population of phosphorylated opsin in the native membrane. Agonist uptake blocks subsequent entry of the inverse agonist 11-cis-retinal (that is, regeneration of rhodopsin), but regeneration is not blocked in the other half of aporeceptors. Environmentally sensitive fluorophores attached to arrestin reported that conformational changes in loopV−VI (N-domain) are coupled to the entry of agonist, while loopXVIII−XIX (C-domain) engages the aporeceptor even before agonist is added. The data are most consistent with a model in which each domain of arrestin engages its own aporeceptor, and the different binding preferences of the domains lead to asymmetric ligand binding by the aporeceptors. Such a mechanism would protect the rod cell in bright light by concurrently sequestering toxic all-trans-retinol and allowing regeneration with 11-cis-retinal. Following retinal cis/trans isomerisation, the active form of the G-protein-coupled receptor rhodopsin decays to opsin and all-trans-retinal. In this study, arrestin, a regulator of G-protein-coupled receptor activity, is shown to facilitate the concurrent sequestering of toxic all-trans-retinal and regeneration of 11-cis-retinal within the opsin population.
Collapse
|
35
|
Gross OP, Pugh EN, Burns ME. Calcium feedback to cGMP synthesis strongly attenuates single-photon responses driven by long rhodopsin lifetimes. Neuron 2012; 76:370-82. [PMID: 23083739 PMCID: PMC3594095 DOI: 10.1016/j.neuron.2012.07.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2012] [Indexed: 11/26/2022]
Abstract
Rod photoreceptors generate amplified, reproducible responses to single photons via a G protein signaling cascade. Surprisingly, genetic perturbations that dramatically alter the deactivation of the principal signal amplifier, the GPCR rhodopsin (R∗), do not much alter the amplitude of single-photon responses (SPRs). These same perturbations, when crossed into a line lacking calcium feedback regulation of cGMP synthesis, produced much larger alterations in SPR amplitudes. Analysis of SPRs from rods with and without feedback reveal that the consequences of trial-to-trial fluctuations in R∗ lifetime in normal rods are also dampened by feedback regulation of cGMP synthesis. Thus, calcium feedback trumps the mechanisms of R∗ deactivation in determining the SPR amplitude, attenuating responses arising from longer R∗ lifetimes to a greater extent than those arising from shorter ones. As a result, rod SPRs achieve a more stereotyped amplitude, a characteristic considered important for reliable transmission through the visual system.
Collapse
Affiliation(s)
- Owen P. Gross
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Edward N. Pugh
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95618, USA
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA 95618, USA
| | - Marie E. Burns
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA 95618, USA
- Center for Neuroscience and Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA 95618, USA
| |
Collapse
|
36
|
Yanamala N, Gardner E, Riciutti A, Klein-Seetharaman J. The cytoplasmic rhodopsin-protein interface: potential for drug discovery. Curr Drug Targets 2012; 13:3-14. [PMID: 21777183 PMCID: PMC3275648 DOI: 10.2174/138945012798868461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Revised: 02/08/2011] [Accepted: 02/10/2011] [Indexed: 01/20/2023]
Abstract
The mammalian dim-light photoreceptor rhodopsin is a prototypic G protein coupled receptor (GPCR), interacting with the G protein, transducin, rhodopsin kinase, and arrestin. All of these proteins interact with rhodopsin at its cytoplasmic surface. Structural and modeling studies have provided in-depth descriptions of the respective interfaces. Overlap and thus competition for binding surfaces is a major regulatory mechanism for signal processing. Recently, it was found that the same surface is also targeted by small molecules. These ligands can directly interfere with the binding and activation of the proteins of the signal transduction cascade, but they can also allosterically modulate the retinal ligand binding pocket. Because the pocket that is targeted contains residues that are highly conserved across Class A GPCRs, these findings imply that it may be possible to target multiple GPCRs with the same ligand(s). This is desirable for example in complex diseases such as cancer where multiple GPCRs participate in the disease networks.
Collapse
Affiliation(s)
- Naveena Yanamala
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Eric Gardner
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Alec Riciutti
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Judith Klein-Seetharaman
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| |
Collapse
|
37
|
Butcher AJ, Kong KC, Prihandoko R, Tobin AB. Physiological role of G-protein coupled receptor phosphorylation. Handb Exp Pharmacol 2012:79-94. [PMID: 22222696 DOI: 10.1007/978-3-642-23274-9_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is now well established that G-protein coupled receptors (GPCRs) are hyper-phosphorylated following agonist occupation usually at serine and threonine residues contained on the third intracellular loop and C-terminal tail. After some 2 decades of intensive research, the nature of protein kinases involved in this process together with the signalling consequences of receptor phosphorylation has been firmly established. The major challenge that the field currently faces is placing all this information within a physiological context and determining to what extent does phosphoregulation of GPCRs impact on whole animal responses. In this chapter, we address this issue by describing how GPCR phosphorylation might vary depending on the cell type in which the receptor is expressed and how this might be employed to drive selective regulation of physiological responses.
Collapse
Affiliation(s)
- Adrian J Butcher
- Department of Cell Physiology and Pharmacology, University of Leicester, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | | | | | | |
Collapse
|
38
|
Quaroni L, Zlateva T, Normand E. Detection of Weak Absorption Changes from Molecular Events in Time-Resolved FT-IR Spectromicroscopy Measurements of Single Functional Cells. Anal Chem 2011; 83:7371-80. [DOI: 10.1021/ac201318z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Luca Quaroni
- Swiss Light Source, Paul Scherrer Institut, 5232, Villigen-PSI, Switzerland
| | - Theodora Zlateva
- Department of Biochemistry and Cancer Research Center, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Elise Normand
- Canadian Light Source Inc., University of Saskatchewan, Saskatoon, SK, S7N 0X4, Canada
| |
Collapse
|
39
|
Ghosh M, Schonbrunn A. Differential temporal and spatial regulation of somatostatin receptor phosphorylation and dephosphorylation. J Biol Chem 2011; 286:13561-73. [PMID: 21343287 DOI: 10.1074/jbc.m110.215723] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The G(i)-coupled somatostatin 2A receptor (sst2A) mediates many of the neuromodulatory and neuroendocrine actions of somatostatin (SS) and is targeted by the SS analogs used to treat neuroendocrine tumors. As for other G protein-coupled receptors, agonists stimulate sst2A receptor phosphorylation on multiple residues, and phosphorylation at different sites has distinct effects on receptor internalization and uncoupling. To elucidate the spatial and temporal regulation of sst2A receptor phosphorylation, we examined agonist-stimulated phosphorylation of multiple receptor GPCR kinase sites using phospho-site-specific antibodies. SS increased receptor phosphorylation sequentially, first on Ser-341/343 and then on Thr-353/354, followed by receptor internalization. Reversal of receptor phosphorylation was determined by the duration of prior agonist exposure. In acutely stimulated cells, in which most receptors remained on the cell surface, dephosphorylation occurred only on Thr-353/354. In contrast, both Ser-341/343 and Thr-353/354 were rapidly dephosphorylated when cells were stimulated long enough to allow receptor internalization before agonist removal. Consistent with these observations, dephosphorylation of Thr-353/354 was not affected by either hypertonic sucrose or dynasore, which prevent receptor internalization, whereas dephosphorylation of Ser-341/343 was completely blocked. An okadaic acid- and fostriecin-sensitive phosphatase catalyzed the dephosphorylation of Thr-353/354 both intracellularly and at the cell surface. In contrast, dephosphorylation of Ser-341/343 was insensitive to these inhibitors. Our results show that the phosphorylation and dephosphorylation of neighboring GPCR kinase sites in the sst2A receptor are subject to differential spatial and temporal regulation. Thus, the pattern of receptor phosphorylation is determined by the duration of agonist stimulation and compartment-specific enzymatic activity.
Collapse
Affiliation(s)
- Madhumita Ghosh
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas 77225, USA
| | | |
Collapse
|
40
|
Caruso G, Bisegna P, Lenoci L, Andreucci D, Gurevich VV, Hamm HE, DiBenedetto E. Kinetics of rhodopsin deactivation and its role in regulating recovery and reproducibility of rod photoresponse. PLoS Comput Biol 2010; 6:e1001031. [PMID: 21200415 PMCID: PMC3002991 DOI: 10.1371/journal.pcbi.1001031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 11/11/2010] [Indexed: 12/31/2022] Open
Abstract
The single photon response (SPR) in vertebrate phototransduction is regulated by the dynamics of R* during its lifetime, including the random number of phosphorylations, the catalytic activity and the random sojourn time at each phosphorylation level. Because of this randomness the electrical responses are expected to be inherently variable. However the SPR is highly reproducible. The mechanisms that confer to the SPR such a low variability are not completely understood. The kinetics of rhodopsin deactivation is investigated by a Continuous Time Markov Chain (CTMC) based on the biochemistry of rhodopsin activation and deactivation, interfaced with a spatio-temporal model of phototransduction. The model parameters are extracted from the photoresponse data of both wild type and mutant mice, having variable numbers of phosphorylation sites and, with the same set of parameters, the model reproduces both WT and mutant responses. The sources of variability are dissected into its components, by asking whether a random number of turnoff steps, a random sojourn time between steps, or both, give rise to the known variability. The model shows that only the randomness of the sojourn times in each of the phosphorylated states contributes to the Coefficient of Variation (CV) of the response, whereas the randomness of the number of R* turnoff steps has a negligible effect. These results counter the view that the larger the number of decay steps of R*, the more stable the photoresponse is. Our results indicate that R* shutoff is responsible for the variability of the photoresponse, while the diffusion of the second messengers acts as a variability suppressor.
Collapse
Affiliation(s)
- Giovanni Caruso
- Construction Technologies Institute, National Research Council, Rome, Italy
| | - Paolo Bisegna
- Department of Civil Engineering, University of Rome Tor Vergata, Rome, Italy
| | - Leonardo Lenoci
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Daniele Andreucci
- Department of Mathematical Methods and Models, University of Rome La Sapienza, Rome, Italy
| | - Vsevolod V. Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Heidi E. Hamm
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Emmanuele DiBenedetto
- Department of Mathematics, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
The visual system is one of the most energetically demanding systems in the brain. The currency of energy is ATP, which is generated most efficiently from oxidative metabolism in the mitochondria. ATP supports multiple neuronal functions. Foremost is repolarization of the membrane potential after depolarization. Neuronal activity, ATP generation, blood flow, oxygen consumption, glucose utilization, and mitochondrial oxidative metabolism are all interrelated. In the retina, phototransduction, neurotransmitter utilization, and protein/organelle transport are energy-dependent, yet repolarization-after-depolarization consumes the bulk of the energy. Repolarization in photoreceptor inner segments maintains the dark current. Repolarization by all neurons along the visual pathway following depolarizing excitatory glutamatergic neurotransmission preserves cellular integrity and permits reactivation. The higher metabolic activity in the magno- versus the parvo-cellular pathway, the ON- versus the OFF-pathway in some (and the reverse in other) species, and in specialized functional representations in the visual cortex all reflect a greater emphasis on the processing of specific visual attributes. Neuronal activity and energy metabolism are tightly coupled processes at the cellular and even at the molecular levels. Deficiencies in energy metabolism, such as in diabetes, mitochondrial DNA mutation, mitochondrial protein malfunction, and oxidative stress can lead to retinopathy, visual deficits, neuronal degeneration, and eventual blindness.
Collapse
Affiliation(s)
- Margaret T T Wong-Riley
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
42
|
Burns ME, Pugh EN. Lessons from photoreceptors: turning off g-protein signaling in living cells. Physiology (Bethesda) 2010; 25:72-84. [PMID: 20430952 DOI: 10.1152/physiol.00001.2010] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phototransduction in retinal rods is one of the most extensively studied G-protein signaling systems. In recent years, our understanding of the biochemical steps that regulate the deactivation of the rod's response to light has greatly improved. Here, we summarize recent advances and highlight some of the remaining puzzles in this model signaling system.
Collapse
Affiliation(s)
- Marie E Burns
- Departments of Ophthalmology and Vision Science, University of California, Davis, California, USA.
| | | |
Collapse
|
43
|
Saprunova VB, Pilipenko DI, Alexeevsky AV, Fursova AZ, Kolosova NG, Bakeeva LE. Lipofuscin granule dynamics during development of age-related macular degeneration. BIOCHEMISTRY (MOSCOW) 2010; 75:130-8. [PMID: 20367599 DOI: 10.1134/s0006297910020021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The pigment epithelium cell structure and therapeutic effect of antioxidant SkQ1, selectively penetrating into mitochondria from eye drops, were studied upon development in OXYS rats of age-related retinopathy as a model of macular degeneration. The characteristic dynamics and ultrastructural peculiarities of the layer of electron-dense cytoplasmic structures of the pigment epithelium apex part and incorporated lipofuscin granules were revealed. The therapy of OXYS animals for 68 days using 250 nM SkQ1 drops decreased the extent of development of age-related macular degeneration. Electron-microscopic investigation showed that SkQ1 prevented development of ultrastructural changes in the pigment epithelium characteristic of macular degeneration, the condition of which after therapy with SkQ1 drops corresponded to ultrastructure of pigment epithelium in Wistar rats of the same age having no symptoms of retinal damage. It is supposed that ultrastructural changes in the electron-dense layer upon development of age-related macular degeneration are indicative of disturbances in the optical cycle functioning, especially of disturbances in functioning of photoreceptor membranes.
Collapse
Affiliation(s)
- V B Saprunova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | | | | | | | | | | |
Collapse
|
44
|
Mechanistic basis for the failure of cone transducin to translocate: why cones are never blinded by light. J Neurosci 2010; 30:6815-24. [PMID: 20484624 DOI: 10.1523/jneurosci.0613-10.2010] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The remarkable ability of our vision to function under ever-changing conditions of ambient illumination is mediated by multiple molecular mechanisms regulating the light sensitivity of rods and cones. One such mechanism involves massive translocation of signaling proteins, including the G-protein transducin, into and out of the light-sensitive photoreceptor outer segment compartment. Transducin translocation extends the operating range of rods, but in cones transducin never translocates, which is puzzling because cones typically function in much brighter light than rods. Using genetically manipulated mice in which the rates of transducin activation and inactivation were altered, we demonstrate that, like in rods, transducin translocation in cones can be triggered when transducin activation exceeds a critical level, essentially saturating the photoresponse. However, this level is never achieved in wild-type cones: their superior ability to tightly control the rates of transducin activation and inactivation, responsible for avoiding saturation by light, also accounts for the prevention of transducin translocation at any light intensity.
Collapse
|
45
|
Lee KA, Nawrot M, Garwin GG, Saari JC, Hurley JB. Relationships among visual cycle retinoids, rhodopsin phosphorylation, and phototransduction in mouse eyes during light and dark adaptation. Biochemistry 2010; 49:2454-63. [PMID: 20155952 DOI: 10.1021/bi1001085] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phosphorylation and regeneration of rhodopsin, the prototypical G-protein-coupled receptor, each can influence light and dark adaptation. To evaluate their relative contributions, we quantified rhodopsin, retinoids, phosphorylation, and photosensitivity in mice during a 90 min illumination followed by dark adaptation. During illumination, all-trans-retinyl esters and, to a lesser extent, all-trans-retinal accumulate and reach the steady state in <1 h. Each major phosphorylation site on rhodopsin reaches a steady state level of phosphorylation at a different time during illumination. The dominant factor that limits dark adaptation is isomerization of retinal. During dark adaptation, dephosphorylation of rhodopsin occurs in two phases. The faster phase corresponds to rapid dephosphorylation of regenerated rhodopsin present at the end of the illumination period. The slower phase corresponds to dephosphorylation of rhodopsin as it forms by regeneration. We conclude that rhodopsin phosphorylation has three physiological functions: it quenches phototransduction, reduces sensitivity during light adaptation, and suppresses bleached rhodopsin activity during dark adaptation.
Collapse
Affiliation(s)
- Kimberly A Lee
- Department of Biochemistry (Box 357350), University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | |
Collapse
|
46
|
Busillo JM, Armando S, Sengupta R, Meucci O, Bouvier M, Benovic JL. Site-specific phosphorylation of CXCR4 is dynamically regulated by multiple kinases and results in differential modulation of CXCR4 signaling. J Biol Chem 2010; 285:7805-17. [PMID: 20048153 DOI: 10.1074/jbc.m109.091173] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The chemokine receptor CXCR4 is a widely expressed G protein-coupled receptor that has been implicated in a number of diseases including human immunodeficiency virus, cancer, and WHIM syndrome, with the latter two involving dysregulation of CXCR4 signaling. To better understand the role of phosphorylation in regulating CXCR4 signaling, tandem mass spectrometry and phospho-specific antibodies were used to identify sites of agonist-promoted phosphorylation. These studies demonstrated that Ser-321, Ser-324, Ser-325, Ser-330, Ser-339, and two sites between Ser-346 and Ser-352 were phosphorylated in HEK293 cells. We show that Ser-324/5 was rapidly phosphorylated by protein kinase C and G protein-coupled receptor kinase 6 (GRK6) upon CXCL12 treatment, whereas Ser-339 was specifically and rapidly phosphorylated by GRK6. Ser-330 was also phosphorylated by GRK6, albeit with slower kinetics. Similar results were observed in human astroglia cells, where endogenous CXCR4 was rapidly phosphorylated on Ser-324/5 by protein kinase C after CXCL12 treatment, whereas Ser-330 was slowly phosphorylated. Analysis of CXCR4 signaling in HEK293 cells revealed that calcium mobilization was primarily negatively regulated by GRK2, GRK6, and arrestin3, whereas GRK3, GRK6, and arrestin2 played a primary role in positively regulating ERK1/2 activation. In contrast, GRK2 appeared to play a negative role in ERK1/2 activation. Finally, we show that arrestin association with CXCR4 is primarily driven by the phosphorylation of far C-terminal residues on the receptor. These studies reveal that site-specific phosphorylation of CXCR4 is dynamically regulated by multiple kinases resulting in both positive and negative modulation of CXCR4 signaling.
Collapse
Affiliation(s)
- John M Busillo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | |
Collapse
|
47
|
Arrestin competition influences the kinetics and variability of the single-photon responses of mammalian rod photoreceptors. J Neurosci 2009; 29:11867-79. [PMID: 19776273 DOI: 10.1523/jneurosci.0819-09.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Reliable signal transduction via G-protein-coupled receptors requires proper receptor inactivation. For example, signals originating from single rhodopsin molecules vary little from one to the next, requiring reproducible inactivation of rhodopsin by phosphorylation and arrestin binding. We determined how reduced concentrations of rhodopsin kinase (GRK1) and/or arrestin1 influenced the kinetics and variability of the single-photon responses of mouse rod photoreceptors. These experiments revealed that arrestin, in addition to its role in quenching the activity of rhodopsin, can tune the kinetics of rhodopsin phosphorylation by competing with GRK1. This competition influenced the variability of the active lifetime of rhodopsin. Biasing the competition in favor of GRK1 revealed that rhodopsin remained active through much of the single-photon response under the conditions of our experiments. This long-lasting rhodopsin activity can explain the characteristic time course of single-photon response variability. Indeed, explaining the late time-to-peak of the variance required an active lifetime of rhodopsin approximately twice that of the G-protein transducin. Competition between arrestins and kinases may be a general means of influencing signals mediated by G-protein-coupled receptors, particularly when activation of a few receptors produces signals of functional importance.
Collapse
|
48
|
Abstract
Multisite phosphorylation is an important mechanism for fine-tuned regulation of protein function. Mathematical models developed over recent years have contributed to elucidation of the functional consequences of a variety of molecular mechanisms involved in processing of the phosphorylation sites. Here we review the results of such models, together with salient experimental findings on multisite protein phosphorylation. We discuss how molecular mechanisms that can be distinguished with respect to the order and processivity of phosphorylation, as well as other factors, regulate changes in the sensitivity and kinetics of the response, the synchronization of molecular events, signalling specificity, and other functional implications.
Collapse
Affiliation(s)
- Carlos Salazar
- Research Group Modeling of Biological Systems (B086), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany.
| | | |
Collapse
|
49
|
Morgans CW, Kensel-Hammes P, Hurley JB, Burton K, Idzerda R, McKnight GS, Bajjalieh SM. Loss of the Synaptic Vesicle Protein SV2B results in reduced neurotransmission and altered synaptic vesicle protein expression in the retina. PLoS One 2009; 4:e5230. [PMID: 19381277 PMCID: PMC2667261 DOI: 10.1371/journal.pone.0005230] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 03/18/2009] [Indexed: 11/25/2022] Open
Abstract
The Synaptic Vesicle Protein 2 (SV2) family of transporter-like proteins is expressed exclusively in vesicles that undergo calcium-regulated exocytosis. Of the three isoforms expressed in mammals, SV2B is the most divergent. Here we report studies of SV2B location and function in the retina. Immunolabeling studies revealed that SV2B is detected in rod photoreceptor synaptic terminals where it is the primary isoform. In mice lacking SV2B, synaptic transmission at the synapse between photoreceptors and bipolar neurons was decreased, as evidenced by a significant reduction in the amplitude of the b-wave in electroretinogram recordings. Quantitative immunoblot analyses of whole eyes revealed that loss of SV2B was associated with reduced levels of synaptic vesicle proteins including synaptotagmin, VAMP, synaptophysin and the vesicular glutamate transporter V-GLUT1. Immunolabeling studies revealed that SV2B is detected in rod photoreceptor synaptic terminals where it is the primary isoform. Thus, SV2B contributes to the modulation of synaptic vesicle exocytosis and plays a significant role in regulating synaptic protein content.
Collapse
Affiliation(s)
- Catherine W. Morgans
- Casey Eye Institute, Oregon Health & Sciences University, Portland, Oregon, United States of America
| | - Patricia Kensel-Hammes
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - James B. Hurley
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Kimberly Burton
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Rejean Idzerda
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - G. Stanley McKnight
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Sandra M. Bajjalieh
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
50
|
Okawa H, Sampath AP, Laughlin SB, Fain GL. ATP consumption by mammalian rod photoreceptors in darkness and in light. Curr Biol 2008; 18:1917-21. [PMID: 19084410 DOI: 10.1016/j.cub.2008.10.029] [Citation(s) in RCA: 269] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 10/06/2008] [Accepted: 10/08/2008] [Indexed: 11/30/2022]
Abstract
Why do vertebrates use rods and cones that hyperpolarize, when in insect eyes a single depolarizing photoreceptor can function at all light levels? We answer this question at least in part with a comprehensive assessment of ATP consumption for mammalian rods from voltages and currents and recently published physiological and biochemical data. In darkness, rods consume 10(8) ATP s(-1), about the same as Drosophila photoreceptors. Ion fluxes associated with phototransduction and synaptic transmission dominate; as in CNS, the contribution of enzymes of the second-messenger cascade is surprisingly small. Suppression of rod responses in daylight closes light-gated channels and reduces total energy consumption by >75%, but in Drosophila light opens channels and increases consumption 5-fold. Rods therefore provide an energy-efficient mechanism not present in rhabdomeric photoreceptors. Rods are metabolically less "costly" than cones, because cones do not saturate in bright light and use more ATP s(-1) for transducin activation and rhodopsin phosphorylation. This helps to explain why the vertebrate retina is duplex, and why some diurnal animals like primates have a small number of cones, concentrated in a region of high acuity.
Collapse
Affiliation(s)
- Haruhisa Okawa
- Neuroscience Graduate Program, Zilkha Neurogenetic Institute, USC Keck School of Medicine, Los Angeles, CA 90089, USA
| | | | | | | |
Collapse
|