1
|
Ashok S, Ramachandra Rao S. Updates on protein-prenylation and associated inherited retinopathies. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1410874. [PMID: 39026984 PMCID: PMC11254824 DOI: 10.3389/fopht.2024.1410874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Membrane-anchored proteins play critical roles in cell signaling, cellular architecture, and membrane biology. Hydrophilic proteins are post-translationally modified by a diverse range of lipid molecules such as phospholipids, glycosylphosphatidylinositol, and isoprenes, which allows their partition and anchorage to the cell membrane. In this review article, we discuss the biochemical basis of isoprenoid synthesis, the mechanisms of isoprene conjugation to proteins, and the functions of prenylated proteins in the neural retina. Recent discovery of novel prenyltransferases, prenylated protein chaperones, non-canonical prenylation-target motifs, and reversible prenylation is expected to increase the number of inherited systemic and blinding diseases with aberrant protein prenylation. Recent important investigations have also demonstrated the role of several unexpected regulators (such as protein charge, sequence/protein-chaperone interaction, light exposure history) in the photoreceptor trafficking of prenylated proteins. Technical advances in the investigation of the prenylated proteome and its application in vision research are discussed. Clinical updates and technical insights into known and putative prenylation-associated retinopathies are provided herein. Characterization of non-canonical prenylation mechanisms in the retina and retina-specific prenylated proteome is fundamental to the understanding of the pathogenesis of protein prenylation-associated inherited blinding disorders.
Collapse
Affiliation(s)
- Sudhat Ashok
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
| | - Sriganesh Ramachandra Rao
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
- Research Service, VA Western New York Healthcare System, Buffalo, NY, United States
| |
Collapse
|
2
|
Kankanamge D, Tennakoon M, Karunarathne A, Gautam N. G protein gamma subunit, a hidden master regulator of GPCR signaling. J Biol Chem 2022; 298:102618. [PMID: 36272647 PMCID: PMC9678972 DOI: 10.1016/j.jbc.2022.102618] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022] Open
Abstract
Heterotrimeric G proteins (αβγ subunits) that are activated by G protein-coupled receptors (GPCRs) mediate the biological responses of eukaryotic cells to extracellular signals. The α subunits and the tightly bound βγ subunit complex of G proteins have been extensively studied and shown to control the activity of effector molecules. In contrast, the potential roles of the large family of γ subunits have been less studied. In this review, we focus on present knowledge about these proteins. Induced loss of individual γ subunit types in animal and plant models result in strikingly distinct phenotypes indicating that γ subtypes play important and specific roles. Consistent with these findings, downregulation or upregulation of particular γ subunit types result in various types of cancers. Clues about the mechanistic basis of γ subunit function have emerged from imaging the dynamic behavior of G protein subunits in living cells. This shows that in the basal state, G proteins are not constrained to the plasma membrane but shuttle between membranes and on receptor activation βγ complexes translocate reversibly to internal membranes. The translocation kinetics of βγ complexes varies widely and is determined by the membrane affinity of the associated γ subtype. On translocating, some βγ complexes act on effectors in internal membranes. The variation in translocation kinetics determines differential sensitivity and adaptation of cells to external signals. Membrane affinity of γ subunits is thus a parsimonious and elegant mechanism that controls information flow to internal cell membranes while modulating signaling responses.
Collapse
Affiliation(s)
- Dinesh Kankanamge
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Mithila Tennakoon
- Department of Chemistry, St Louis University, St Louis, Missouri, USA
| | | | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; Department of Genetics, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
3
|
Kolesnikov AV, Lobysheva E, Gnana-Prakasam JP, Kefalov VJ, Kisselev OG. Regulation of rod photoreceptor function by farnesylated G-protein γ-subunits. PLoS One 2022; 17:e0272506. [PMID: 35939447 PMCID: PMC9359561 DOI: 10.1371/journal.pone.0272506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022] Open
Abstract
Heterotrimeric G-protein transducin, Gt, is a key signal transducer and amplifier in retinal rod and cone photoreceptor cells. Despite similar subunit composition, close amino acid identity, and identical posttranslational farnesylation of their Gγ subunits, rods and cones rely on unique Gγ1 (Gngt1) and Gγc (Gngt2) isoforms, respectively. The only other farnesylated G-protein γ-subunit, Gγ11 (Gng11), is expressed in multiple tissues but not retina. To determine whether Gγ1 regulates uniquely rod phototransduction, we generated transgenic rods expressing Gγ1, Gγc, or Gγ11 in Gγ1-deficient mice and analyzed their properties. Immunohistochemistry and Western blotting demonstrated the robust expression of each transgenic Gγ in rod cells and restoration of Gαt1 expression, which is greatly reduced in Gγ1-deficient rods. Electroretinography showed restoration of visual function in all three transgenic Gγ1-deficient lines. Recordings from individual transgenic rods showed that photosensitivity impaired in Gγ1-deficient rods was also fully restored. In all dark-adapted transgenic lines, Gαt1 was targeted to the outer segments, reversing its diffuse localization found in Gγ1-deficient rods. Bright illumination triggered Gαt1 translocation from the rod outer to inner segments in all three transgenic strains. However, Gαt1 translocation in Gγ11 transgenic mice occurred at significantly dimmer background light. Consistent with this, transretinal ERG recordings revealed gradual response recovery in moderate background illumination in Gγ11 transgenic mice but not in Gγ1 controls. Thus, while farnesylated Gγ subunits are functionally active and largely interchangeable in supporting rod phototransduction, replacement of retina-specific Gγ isoforms by the ubiquitous Gγ11 affects the ability of rods to adapt to background light.
Collapse
Affiliation(s)
- Alexander V. Kolesnikov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA, United States of America
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elena Lobysheva
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jaya P. Gnana-Prakasam
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Vladimir J. Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA, United States of America
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Physiology and Biophysics, University of California, Irvine, CA, United States of America
| | - Oleg G. Kisselev
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
4
|
Diversity of the Gβγ complexes defines spatial and temporal bias of GPCR signaling. Cell Syst 2021; 12:324-337.e5. [PMID: 33667409 DOI: 10.1016/j.cels.2021.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 12/09/2020] [Accepted: 02/04/2021] [Indexed: 01/04/2023]
Abstract
The signal transduction by G-protein-coupled receptors (GPCRs) is mediated by heterotrimeric G proteins composed from one of the 16 Gα subunits and the inseparable Gβγ complex assembled from a repertoire of 5 Gβ and 12 Gγ subunits. However, the functional role of compositional diversity in Gβγ complexes has been elusive. Using optical biosensors, we examined the function of all Gβγ combinations in living cells and uncovered two major roles of Gβγ diversity. First, we demonstrate that the identity of Gβγ subunits greatly influences the kinetics and efficacy of GPCR responses at the plasma membrane. Second, we show that different Gβγ combinations are selectively dispatched from the plasma membrane to various cellular organelles on a timescale from milliseconds to minutes. We describe the mechanisms regulating these processes and document their implications for GPCR signaling via various Gα subunits, thereby illustrating a role for the compositional diversity of G protein heterotrimers.
Collapse
|
5
|
Chaya T, Furukawa T. Post-translational modification enzymes as key regulators of ciliary protein trafficking. J Biochem 2021; 169:633-642. [PMID: 33681987 PMCID: PMC8423421 DOI: 10.1093/jb/mvab024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
Primary cilia are evolutionarily conserved microtubule-based organelles that protrude from the surface of almost all cell types and decode a variety of extracellular stimuli. Ciliary dysfunction causes human diseases named ciliopathies, which span a wide range of symptoms, such as developmental and sensory abnormalities. The assembly, disassembly, maintenance and function of cilia rely on protein transport systems including intraflagellar transport (IFT) and lipidated protein intraflagellar targeting (LIFT). IFT is coordinated by three multisubunit protein complexes with molecular motors along the ciliary axoneme, while LIFT is mediated by specific chaperones that directly recognize lipid chains. Recently, it has become clear that several post-translational modification enzymes play crucial roles in the regulation of IFT and LIFT. Here, we review our current understanding of the roles of these post-translational modification enzymes in the regulation of ciliary protein trafficking as well as their regulatory mechanisms, physiological significance and involvement in human diseases.
Collapse
Affiliation(s)
- Taro Chaya
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
6
|
Chaya T, Tsutsumi R, Varner LR, Maeda Y, Yoshida S, Furukawa T. Cul3-Klhl18 ubiquitin ligase modulates rod transducin translocation during light-dark adaptation. EMBO J 2019; 38:e101409. [PMID: 31696965 DOI: 10.15252/embj.2018101409] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 01/02/2023] Open
Abstract
Adaptation is a general feature of sensory systems. In rod photoreceptors, light-dependent transducin translocation and Ca2+ homeostasis are involved in light/dark adaptation and prevention of cell damage by light. However, the underlying regulatory mechanisms remain unclear. Here, we identify mammalian Cul3-Klhl18 ubiquitin ligase as a transducin translocation modulator during light/dark adaptation. Under dark conditions, Klhl18-/- mice exhibited decreased rod light responses and subcellular localization of the transducin α-subunit (Tα), similar to that observed in light-adapted Klhl18+/+ mice. Cul3-Klhl18 promoted ubiquitination and degradation of Unc119, a rod Tα-interacting protein. Unc119 overexpression phenocopied Tα mislocalization observed in Klhl18-/- mice. Klhl18 weakly recognized casein kinase-2-phosphorylated Unc119 protein, which is dephosphorylated by Ca2+ -dependent phosphatase calcineurin. Calcineurin inhibition increased Unc119 expression and Tα mislocalization in rods. These results suggest that Cul3-Klhl18 modulates rod Tα translocation during light/dark adaptation through Unc119 ubiquitination, which is affected by phosphorylation. Notably, inactivation of the Cul3-Klhl18 ligase and calcineurin inhibitors FK506 and cyclosporine A that are known immunosuppressant drugs repressed light-induced photoreceptor damage, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- Taro Chaya
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ryotaro Tsutsumi
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Leah Rie Varner
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yamato Maeda
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Satoyo Yoshida
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
7
|
Brooks C, Murphy J, Belcastro M, Heller D, Kolandaivelu S, Kisselev O, Sokolov M. Farnesylation of the Transducin G Protein Gamma Subunit Is a Prerequisite for Its Ciliary Targeting in Rod Photoreceptors. Front Mol Neurosci 2018; 11:16. [PMID: 29410614 PMCID: PMC5787109 DOI: 10.3389/fnmol.2018.00016] [Citation(s) in RCA: 12] [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: 11/22/2017] [Accepted: 01/10/2018] [Indexed: 12/26/2022] Open
Abstract
Primary cilia are microtubule-based organelles, which protrude from the plasma membrane and receive a wide range of extracellular signals. Various cilia use G protein-coupled receptors (GPCRs) for the detection of these signals. For instance, vertebrate rod photoreceptors use their cilia (also called outer segments) as antennae detecting photons by GPCR rhodopsin. Rhodopsin recognizes incoming light and activates its G protein, transducin, which is composed of three subunits α, β, and γ. Similar to all G protein γ subunits, the transducin Gγ1 subunit undergoes C-terminal prenylation resulting in the addition of an isoprenoid farnesyl; however, the significance of this posttranslational modification is unclear. To study the role of the farnesyl group, we genetically introduced a mutant Gγ1 that lacked the prenylation site into the retinal photoreceptors of mice. The biochemical and physiological analyses of these mice revealed that mutant Gγ1 dimerizes with the endogenous transducin Gβ1 subunit and that the resulting Gβγ dimers display reduced hydrophobicity. Although mutant Gβγ dimers could form a heterotrimeric G protein, they could not mediate phototransduction. This deficiency was due to a strong exclusion of non-farnesylated Gβγ complexes from the cilia (rod outer segments). Our results provide the first evidence that farnesylation is required for trafficking of G-protein βγ subunits to the cilium of rod photoreceptors.
Collapse
Affiliation(s)
- Celine Brooks
- Department of Ophthalmology, West Virginia University, Morgantown, WV, United States
| | - Joseph Murphy
- Department of Ophthalmology, West Virginia University, Morgantown, WV, United States
| | | | - Daniel Heller
- Department of Ophthalmology, West Virginia University, Morgantown, WV, United States
| | | | - Oleg Kisselev
- Department of Ophthalmology, Saint Louis University, St. Louis, MO, United States
| | - Maxim Sokolov
- Department of Ophthalmology, West Virginia University, Morgantown, WV, United States
- Department of Biochemistry, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
8
|
Temple KJ, Wright EN, Fierke CA, Gibbs RA. Exploration of GGTase-I substrate requirements. Part 1: Synthesis and biochemical evaluation of novel aryl-modified geranylgeranyl diphosphate analogs. Bioorg Med Chem Lett 2016; 26:3499-502. [PMID: 27342750 DOI: 10.1016/j.bmcl.2016.06.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/11/2016] [Accepted: 06/13/2016] [Indexed: 10/21/2022]
Abstract
Protein geranylgeranylation is a type of post-translational modification that aids in the localization of proteins to the plasma member where they elicit cellular signals. To better understand the isoprenoid requirements of GGTase-I, a series of aryl-modified geranylgeranyl diphosphate analogs were synthesized and screened against mammalian GGTase-I. Of our seven-member library of compounds, six analogs proved to be substrates of GGTase-I, with 6d having a krel=1.93 when compared to GGPP (krel=1.0).
Collapse
Affiliation(s)
- Kayla J Temple
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; The Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47909, USA.
| | - Elia N Wright
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carol A Fierke
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard A Gibbs
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; The Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47909, USA
| |
Collapse
|
9
|
Dinculescu A, Stupay RM, Deng WT, Dyka FM, Min SH, Boye SL, Chiodo VA, Abrahan CE, Zhu P, Li Q, Strettoi E, Novelli E, Nagel-Wolfrum K, Wolfrum U, Smith WC, Hauswirth WW. AAV-Mediated Clarin-1 Expression in the Mouse Retina: Implications for USH3A Gene Therapy. PLoS One 2016; 11:e0148874. [PMID: 26881841 PMCID: PMC4755610 DOI: 10.1371/journal.pone.0148874] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 01/23/2016] [Indexed: 02/07/2023] Open
Abstract
Usher syndrome type III (USH3A) is an autosomal recessive disorder caused by mutations in clarin-1 (CLRN1) gene, leading to progressive retinal degeneration and sensorineural deafness. Efforts to develop therapies for preventing photoreceptor cell loss are hampered by the lack of a retinal phenotype in the existing USH3 mouse models and by conflicting reports regarding the endogenous retinal localization of clarin-1, a transmembrane protein of unknown function. In this study, we used an AAV-based approach to express CLRN1 in the mouse retina in order to determine the pattern of its subcellular localization in different cell types. We found that all major classes of retinal cells express AAV-delivered CLRN1 driven by the ubiquitous, constitutive small chicken β-actin promoter, which has important implications for the design of future USH3 gene therapy studies. Within photoreceptor cells, AAV-expressed CLRN1 is mainly localized at the inner segment region and outer plexiform layer, similar to the endogenous expression of other usher proteins. Subretinal delivery using a full strength viral titer led to significant loss of retinal function as evidenced by ERG analysis, suggesting that there is a critical limit for CLRN1 expression in photoreceptor cells. Taken together, these results suggest that CLRN1 expression is potentially supported by a variety of retinal cells, and the right combination of AAV vector dose, promoter, and delivery method needs to be selected to develop safe therapies for USH3 disorder.
Collapse
Affiliation(s)
- Astra Dinculescu
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| | - Rachel M. Stupay
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Wen-Tao Deng
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Frank M. Dyka
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Seok-Hong Min
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Sanford L. Boye
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Vince A. Chiodo
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Carolina E. Abrahan
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Ping Zhu
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Qiuhong Li
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | | | | | - Kerstin Nagel-Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - W. Clay Smith
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | | |
Collapse
|
10
|
Pearring JN, Salinas RY, Baker SA, Arshavsky VY. Protein sorting, targeting and trafficking in photoreceptor cells. Prog Retin Eye Res 2013; 36:24-51. [PMID: 23562855 DOI: 10.1016/j.preteyeres.2013.03.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/22/2013] [Accepted: 03/26/2013] [Indexed: 01/24/2023]
Abstract
Vision is the most fundamental of our senses initiated when photons are absorbed by the rod and cone photoreceptor neurons of the retina. At the distal end of each photoreceptor resides a light-sensing organelle, called the outer segment, which is a modified primary cilium highly enriched with proteins involved in visual signal transduction. At the proximal end, each photoreceptor has a synaptic terminal, which connects this cell to the downstream neurons for further processing of the visual information. Understanding the mechanisms involved in creating and maintaining functional compartmentalization of photoreceptor cells remains among the most fascinating topics in ocular cell biology. This review will discuss how photoreceptor compartmentalization is supported by protein sorting, targeting and trafficking, with an emphasis on the best-studied cases of outer segment-resident proteins.
Collapse
Affiliation(s)
- Jillian N Pearring
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
11
|
Thung L, Chakravorty D, Trusov Y, Jones AM, Botella JR. Signaling specificity provided by the Arabidopsis thaliana heterotrimeric G-protein γ subunits AGG1 and AGG2 is partially but not exclusively provided through transcriptional regulation. PLoS One 2013; 8:e58503. [PMID: 23520518 PMCID: PMC3592790 DOI: 10.1371/journal.pone.0058503] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 02/05/2013] [Indexed: 11/18/2022] Open
Abstract
The heterotrimeric G-protein complex in Arabidopsis thaliana consists of one α, one ß and three γ subunits. While two of the γ subunits, AGG1 and AGG2 have been shown to provide functional selectivity to the Gßγ dimer in Arabidopsis, it is unclear if such selectivity is embedded in their molecular structures or conferred by the different expression patterns observed in both subunits. In order to study the molecular basis for such selectivity we tested genetic complementation of AGG1- and AGG2 driven by the respectively swapped gene promoters. When expressed in the same tissues as AGG1, AGG2 rescues some agg1 mutant phenotypes such as the hypersensitivity to Fusarium oxysporum and D-mannitol as well as the altered levels of lateral roots, but does not rescue the early flowering phenotype. Similarly, AGG1 when expressed in the same tissues as AGG2 rescues the osmotic stress and lateral-root phenotypes observed in agg2 mutants but failed to rescue the heat-stress induction of flowering. The fact that AGG1 and AGG2 are functionally interchangeable in some pathways implies that, at least for those pathways, signaling specificity resides in the distinctive spatiotemporal expression patterns exhibited by each γ subunit. On the other hand, the lack of complementation for some phenotypes indicates that there are pathways in which signaling specificity is provided by differences in the primary AGG1 and AGG2 amino acid sequences.
Collapse
Affiliation(s)
- Leena Thung
- Plant Genetic Engineering Laboratory, School of Agriculture and Food Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - David Chakravorty
- Plant Genetic Engineering Laboratory, School of Agriculture and Food Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Yuri Trusov
- Plant Genetic Engineering Laboratory, School of Agriculture and Food Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Alan M. Jones
- Departments of Biology and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - José Ramón Botella
- Plant Genetic Engineering Laboratory, School of Agriculture and Food Sciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Mao W, Miyagishima KJ, Yao Y, Soreghan B, Sampath AP, Chen J. Functional comparison of rod and cone Gα(t) on the regulation of light sensitivity. J Biol Chem 2013; 288:5257-67. [PMID: 23288843 DOI: 10.1074/jbc.m112.430058] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The signaling cascades mediated by G protein-coupled receptors (GPCRs) exhibit a wide spectrum of spatial and temporal response properties to fulfill diverse physiological demands. However, the mechanisms that shape the signaling response of the GPCR are not well understood. In this study, we replaced cone transducin α (cTα) for rod transducin α (rTα) in rod photoreceptors of transgenic mice, which also express S opsin, to evaluate the role of Gα subtype on signal amplification from different GPCRs in the same cell; such analysis may explain functional differences between retinal rod and cone photoreceptors. We showed that ectopically expressed cTα 1) forms a heterotrimeric complex with rod Gβ(1)γ(1), 2) substitutes equally for rTα in generating photoresponses initiated by either rhodopsin or S-cone opsin, and 3) exhibited similar light-activated translocation as endogenous rTα in rods and endogenous cTα in cones. Thus, rTα and cTα appear functionally interchangeable. Interestingly, light sensitivity appeared to correlate with the concentration of cTα when expression is reduced below 35% of normal. However, quantification of endogenous cTα concentration in cones showed a higher level to rTα in rods. Thus, reduced sensitivity in cones cannot be explained by reduced coupling efficiency between the GPCR and G protein or a lower concentration of G protein in cones versus rods.
Collapse
Affiliation(s)
- Wen Mao
- Department of Cell and Neurobiology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | |
Collapse
|
13
|
Najafi M, Calvert PD. Transport and localization of signaling proteins in ciliated cells. Vision Res 2012; 75:11-8. [PMID: 22922002 DOI: 10.1016/j.visres.2012.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/05/2012] [Accepted: 08/08/2012] [Indexed: 11/16/2022]
Abstract
Most cells in the human body elaborate cilia which serve a wide variety of functions, including cell and tissue differentiation during development, sensing physical and chemical properties of the extracellular milieu and mechanical force generation. Common among cilia is the transduction of external stimuli into signals that regulate the activities of the cilia and the cells that possess them. These functions require the transport and localization of specialized proteins to the cilium, a process that many recent studies have shown to be vital for normal cell function and, ultimately, the health of the organism. Here we discuss several mechanisms proposed for the transport and localization of soluble and peripheral membrane proteins to, or their exclusion from the ciliary compartment with a focus on how the structure of the cytoplasm and the size and shape of proteins influence these processes. Additionally, we examine the impact of cell and protein structure on our ability to accurately measure the relative concentrations of fluorescently tagged proteins amongst various cellular domains, which is integral to our understanding of the molecular mechanisms underlying protein localization and transport.
Collapse
Affiliation(s)
- Mehdi Najafi
- Department of Ophthalmology and the Center for Vision Research, SUNY Upstate Medical University, United States
| | | |
Collapse
|
14
|
Belcastro M, Song H, Sinha S, Song C, Mathers PH, Sokolov M. Phosphorylation of phosducin accelerates rod recovery from transducin translocation. Invest Ophthalmol Vis Sci 2012; 53:3084-91. [PMID: 22491418 DOI: 10.1167/iovs.11-8798] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE In rods saturated by light, the G protein transducin undergoes translocation from the outer segment compartment, which results in the uncoupling of transducin from its innate receptor, rhodopsin. We measured the kinetics of recovery from this adaptive cellular response, while also investigating the role of phosducin, a phosphoprotein binding transducin βγ subunits in its de-phosphorylated state, in regulating this process. METHODS Mice were exposed to a moderate rod-saturating light triggering transducin translocation, and then allowed to recover in the dark while free running. The kinetics of the return of the transducin subunits to the outer segments were compared in transgenic mouse models expressing full-length phosducin, and phosducin lacking phosphorylation sites serine 54 and 71, using Western blot analysis of serial tangential sections of the retina. RESULTS In mice expressing normal phosducin, transducin α and βγ subunits returned to the outer segments with a half-time (t(1/2)) of ∼24 and 29 minutes, respectively. In the phosducin phosphorylation mutants, the transducin α subunit moved four times slower, with t(1/2) ∼95 minutes, while the movement of transducin βγ was less affected. CONCLUSIONS We demonstrate that the recovery of rod photoreceptors from the ambient saturating levels of illumination, in terms of the return of the light-dispersed transducin subunits to the rod outer segments, occurs six times faster than reported previously. Our data also support the notion that the accumulation of transducin α subunit in the outer segment is driven by its re-binding to the transducin βγ dimer, because this process is accelerated significantly by phosducin phosphorylation.
Collapse
|
15
|
Arshavsky VY, Burns ME. Photoreceptor signaling: supporting vision across a wide range of light intensities. J Biol Chem 2011; 287:1620-6. [PMID: 22074925 DOI: 10.1074/jbc.r111.305243] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
For decades, photoreceptors have been an outstanding model system for elucidating basic principles in sensory transduction and biochemistry and for understanding many facets of neuronal cell biology. In recent years, new knowledge of the kinetics of signaling and the large-scale movements of proteins underlying signaling has led to a deeper appreciation of the photoreceptor's unique challenge in mediating the first steps in vision over a wide range of light intensities.
Collapse
Affiliation(s)
- Vadim Y Arshavsky
- Departments of Ophthalmology and Pharmacology, Duke University, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
16
|
Sobierajska K, Joachimiak E, Bregier C, Fabczak S, Fabczak H. Effect of phosducin silencing on the photokinetic motile response of Blepharisma japonicum. Photochem Photobiol Sci 2010; 10:19-24. [PMID: 20976371 DOI: 10.1039/c0pp00221f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The coloured ciliate Blepharisma japonicum changes swimming velocity (positive photokinesis) and elongates its body in response to a prolonged illumination. We have recently proposed that alterations in the phosphorylation level of the ciliate phosducin (Pdc) may be involved in light-induced cell elongation, which in turn affects the interaction of βγ-dimer of G-proteins (Gβγ) with β-tubulin and subsequent cytoskeletal remodelling. The cellular mechanism that governs the photokinetic effect in this ciliate has not been elucidated. In the present study, we utilise real-time PCR to demonstrate that the levels of ciliate Pdc mRNA are significantly reduced in Pdc-RNAi-treated cells compared to cells fed with bacteria carrying the empty vector (control cells). Using western immunoblotting, we confirmed that these cells treated with Pdc-RNAi expressed a substantially lower level of the Pdc protein. The assay also revealed that in ciliates treated with Pdc-RNAi and exposed to light, the cytosolic level of Gβ (~36 kDa) was reduced, whereas the level of Gβ localized to the membrane (~32 kDa) was increased compared to control cells. In addition, behavioural analysis of the cells indicated a substantial reduction of photokinesis. The findings in this study provide additional characterization of the functional properties of the ciliate Pdc protein and we discuss a likely role for this phosphoprotein in the photokinetic phenomenon of the ciliate protist Blepharisma.
Collapse
Affiliation(s)
- Katarzyna Sobierajska
- Department of Cell Biology, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093, Warsaw, Poland
| | | | | | | | | |
Collapse
|
17
|
Chisari M, Saini DK, Cho JH, Kalyanaraman V, Gautam N. G protein subunit dissociation and translocation regulate cellular response to receptor stimulation. PLoS One 2009; 4:e7797. [PMID: 19936219 PMCID: PMC2777387 DOI: 10.1371/journal.pone.0007797] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 10/16/2009] [Indexed: 01/23/2023] Open
Abstract
We examined the role of G proteins in modulating the response of living cells to receptor activation. The response of an effector, phospholipase C-β to M3 muscarinic receptor activation was measured using sensors that detect the generation of inositol triphosphate or diacylglycerol. The recently discovered translocation of Gβγ from plasma membrane to endomembranes on receptor activation attenuated this response. A FRET based G protein sensor suggested that in contrast to translocating Gβγ, non-translocating Gβγ subunits do not dissociate from the αq subunit on receptor activation leading to prolonged retention of the heterotrimer state and an accentuated response. M3 receptors with tethered αq induced differential responses to receptor activation in cells with or without an endogenous translocation capable γ subunit. G protein heterotrimer dissociation and βγ translocation are thus unanticipated modulators of the intensity of a cell's response to an extracellular signal.
Collapse
Affiliation(s)
- Mariangela Chisari
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Deepak Kumar Saini
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joon-Ho Cho
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Vani Kalyanaraman
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - N. Gautam
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
18
|
Giusto NM, Pasquaré SJ, Salvador GA, Ilincheta de Boschero MG. Lipid second messengers and related enzymes in vertebrate rod outer segments. J Lipid Res 2009; 51:685-700. [PMID: 19828910 DOI: 10.1194/jlr.r001891] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Rod outer segments (ROSs) are specialized light-sensitive organelles in vertebrate photoreceptor cells. Lipids in ROS are of considerable importance, not only in providing an adequate environment for efficient phototransduction, but also in originating the second messengers involved in signal transduction. ROSs have the ability to adapt the sensitivity and speed of their responses to ever-changing conditions of ambient illumination. A major contributor to this adaptation is the light-driven translocation of key signaling proteins into and out of ROS. The present review shows how generation of the second lipid messengers from phosphatidylcholine, phosphatidic acid, and diacylglycerol is modulated by the different illumination states in the vertebrate retina. Findings suggest that the light-induced translocation of phototransduction proteins influences the enzymatic activities of phospholipase D, lipid phosphate phosphatase, diacylglyceride lipase, and diacylglyceride kinase, all of which are responsible for the generation of the second messenger molecules.
Collapse
Affiliation(s)
- Norma M Giusto
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina.
| | | | | | | |
Collapse
|
19
|
Saini DK, Chisari M, Gautam N. Shuttling and translocation of heterotrimeric G proteins and Ras. Trends Pharmacol Sci 2009; 30:278-86. [PMID: 19427041 DOI: 10.1016/j.tips.2009.04.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 04/02/2009] [Accepted: 04/02/2009] [Indexed: 12/21/2022]
Abstract
Heterotrimeric G proteins (alphabetagamma) and Ras proteins are activated by cell-surface receptors that sense extracellular signals. Both sets of proteins were traditionally thought to be constrained to the plasma membrane and some intracellular membranes. Live-cell-imaging experiments have now shown that these proteins are mobile inside a cell, shuttling continually between the plasma membrane and intracellular membranes in the basal state, maintaining these proteins in dynamic equilibrium in different membrane compartments. Furthermore, on receptor activation, a family of G protein betagamma subunits translocates rapidly and reversibly to the Golgi and endoplasmic reticulum enabling direct communication between the extracellular signal and intracellular membranes. A member of the Ras family has similarly been shown to translocate on activation. Although the impact of this unexpected intracellular movement of signaling proteins on cell physiology is likely to be distinct, there are striking similarities in the properties of these two families of signal-transducing proteins.
Collapse
Affiliation(s)
- Deepak K Saini
- Department of Anesthesiology, Box 8054, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
20
|
Interaction of retinal guanylate cyclase with the alpha subunit of transducin: potential role in transducin localization. Biochem J 2009; 417:803-12. [PMID: 18840097 DOI: 10.1042/bj20081513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vertebrate phototransduction is mediated by cGMP, which is generated by retGC (retinal guanylate cyclase) and degraded by cGMP phosphodiesterase. Light stimulates cGMP hydrolysis via the G-protein transducin, which directly binds to and activates phosphodiesterase. Bright light also causes relocalization of transducin from the OS (outer segments) of the rod cells to the inner compartments. In the present study, we show experimental evidence for a previously unknown interaction between G(alphat) (the transducin alpha subunit) and retGC. G(alphat) co-immunoprecipitates with retGC from the retina or from co-transfected COS-7 cells. The retGC-G(alphat) complex is also present in cones. The interaction also occurs in mice lacking RGS9 (regulator of G-protein signalling 9), a protein previously shown to associate with both G(alphat) and retGC. The G(alphat)-retGC interaction is mediated primarily by the kinase homology domain of retGC, which binds GDP-bound G(alphat) stronger than the GTP[S] (GTPgammaS; guanosine 5'-[gamma-thio]triphosphate) form. Neither G(alphat) nor G(betagamma) affect retGC-mediated cGMP synthesis, regardless of the presence of GCAP (guanylate cyclase activating protein) and Ca2+. The rate of light-dependent transducin redistribution from the OS to the inner segments is markedly accelerated in the retGC-1-knockout mice, while the migration of transducin to the OS after the onset of darkness is delayed. Supplementation of permeabilized photoreceptors with cGMP does not affect transducin translocation. Taken together, these results suggest that the protein-protein interaction between G(alphat) and retGC represents a novel mechanism regulating light-dependent translocation of transducin in rod photoreceptors.
Collapse
|
21
|
Kosloff M, Alexov E, Arshavsky VY, Honig B. Electrostatic and lipid anchor contributions to the interaction of transducin with membranes: mechanistic implications for activation and translocation. J Biol Chem 2008; 283:31197-207. [PMID: 18782760 PMCID: PMC2576562 DOI: 10.1074/jbc.m803799200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The heterotrimeric G protein transducin is a key component of the
vertebrate phototransduction cascade. Transducin is peripherally attached to
membranes of the rod outer segment, where it interacts with other proteins at
the membrane-cytosol interface. However, upon sustained activation by light,
the dissociated Gtα and
Gβ1γ1 subunits of transducin translocate from
the outer segment to other parts of the rod cell. Here we used a computational
approach to analyze the interaction strength of transducin and its subunits
with acidic lipid bilayers, as well as the range of orientations that they are
allowed to occupy on the membrane surface. Our results suggest that the
combined constraints of electrostatics and lipid anchors substantially limit
the rotational degrees of freedom of the membrane-bound transducin
heterotrimer. This may contribute to a faster transducin activation rate by
accelerating transducin-rhodopsin complex formation. Notably, the membrane
interactions of the dissociated transducin subunits are very different from
those of the heterotrimer. As shown previously,
Gβ1γ1 experiences significant attractive
interactions with negatively charged membranes, whereas our new results
suggest that Gtα is electrostatically repelled by such
membranes. We suggest that this repulsion could facilitate the membrane
dissociation and intracellular translocation of Gtα.
Moreover, based on similarities in sequence and electrostatic properties, we
propose that the properties described for transducin are common to its
homologs within the Gi subfamily. In a broader view, this work
exemplifies how the activity-dependent association and dissociation of a G
protein can change both the affinity for membranes and the range of allowed
orientations, thereby modulating G protein function.
Collapse
Affiliation(s)
- Mickey Kosloff
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Heterotrimeric G proteins dissociate into their component Galpha and Gbetagamma subunits when these proteins are activated in solution. Until recently, it has not been known if subunit dissociation also occurs in cells. The development of optical methods to study G protein activation in live cells has made it possible to demonstrate heterotrimer dissociation at the plasma membrane. However, subunit dissociation is far from complete, and many active [guanosine triphosphate (GTP)-bound] heterotrimers are intact in a steady state. This unexpectedly reluctant dissociation calls for inclusion of a GTP-bound heterotrimeric state in models of the G protein cycle and places renewed emphasis on the relation between subunit dissociation and effector activation.
Collapse
Affiliation(s)
- Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912-2300, USA.
| |
Collapse
|
23
|
Abstract
Signalling by heterotrimeric G proteins is often isoform-specific, meaning certain effectors are regulated exclusively by one family of heterotrimers. For example, in excitable cells inwardly rectifying potassium (GIRK) channels are activated by G betagamma dimers derived specifically from G(i/o) heterotrimers. Since all active heterotrimers are thought to dissociate and release free G betagamma dimers, it is unclear why these channels respond primarily to dimers released by G(i/o) heterotrimers. We reconstituted GIRK channel activation in cells where we could quantify heterotrimer expression at the plasma membrane, GIRK channel activation, and heterotrimer dissociation. We find that G(oA) heterotrimers are more effective activators of GIRK channels than G(s) heterotrimers when comparable amounts of each are available. We also find that active G(oA) heterotrimers dissociate more readily than active G(s) heterotrimers. Differential dissociation may thus provide a simple explanation for G alpha-specific activation of GIRK channels and other G betagamma-sensitive effectors.
Collapse
Affiliation(s)
- Gregory J Digby
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30809, USA
| | | | | |
Collapse
|
24
|
Slepak VZ, Hurley JB. Mechanism of light-induced translocation of arrestin and transducin in photoreceptors: interaction-restricted diffusion. IUBMB Life 2008; 60:2-9. [PMID: 18379987 DOI: 10.1002/iub.7] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Many signaling proteins change their location within cells in response to external stimuli. In photoreceptors, this phenomenon is remarkably robust. The G protein of rod photoreceptors and rod transducin concentrates in the outer segments (OS) of these neurons in darkness. Within approximately 30 minutes after illumination, rod transducin redistributes throughout all of the outer and inner compartments of the cell. Visual arrestin concurrently relocalises from the inner compartments to become sequestered primarily within the OS. In the past several years, the question of whether these proteins are actively moved by molecular motors or whether they are redistributed by simple diffusion has been extensively debated. This review focuses on the most essential works in the area and concludes that the basic principle driving this protein movement is diffusion. The directionality and light dependence of this movement is achieved by the interactions of arrestin and transducin with their spatially restricted binding partners.
Collapse
Affiliation(s)
- Vladlen Z Slepak
- Department of Molecular and Cellular Pharmacology and Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA.
| | | |
Collapse
|
25
|
Light-dependent compartmentalization of transducin in rod photoreceptors. Mol Neurobiol 2008; 37:44-51. [PMID: 18425604 DOI: 10.1007/s12035-008-8015-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 03/17/2008] [Indexed: 10/22/2022]
Abstract
Three major visual signaling proteins, transducin, arrestin, and recoverin undergo bidirectional translocations between the outer segment and inner compartments of rod photoreceptors in a light-dependent manner. The light-dependent translocation of proteins is believed to contribute to adaptation and neuroprotection of photoreceptor cells. The potential physiological significance and mechanisms of light-controlled protein translocations are at the center of current discussion. In this paper, I outline the latest advances in understanding the mechanisms of bidirectional translocation of transducin and determinants of its steady-state distribution in dark- and light-adapted photoreceptor cells.
Collapse
|
26
|
Chen J, Wu M, Sezate SA, Matsumoto H, Ramsey M, McGinnis JF. Interaction of glyceraldehyde-3-phosphate dehydrogenase in the light-induced rod alpha-transducin translocation. J Neurochem 2007; 104:1280-92. [PMID: 18028335 DOI: 10.1111/j.1471-4159.2007.05081.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The light-dependent subcellular translocation of rod alpha-transducin (GNAT-1, or rod Talpha) has been well documented. In dark-adapted animals, rod Talpha (rTalpha) is predominantly located in the rod outer segment (ROS) and translocates into the rod inner segment (RIS) upon exposure to the light. Neither the molecular participants nor the mechanism(s) involved in this protein trafficking are known. We hypothesized that other proteins must interact with rTalpha to affect the translocations. Using the MBP-rTalpha fusion pulldown assay, the yeast two-hybrid assay and the co-immunoprecipitation assay, we identified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and rTalpha as interacting proteins. Immunoprecipitation also showed beta-actin associates with rTalpha in the dark but not in the light. To further investigate the involvement of GAPDH in light-induced rod Talpha translocation, GAPDH mRNA was knocked down in vivo by transient expression of siRNAs in rat photoreceptor cells. Under completely dark- and light-adapted conditions, the translocation of rTalpha was not significantly different within the 'GAPDH knock-down photoreceptor cells' compared to the non-transfected control cells. However, under partial dark-adaptation, rTalpha translocated more slowly in the 'GAPDH knock-down cells' supporting the conclusion that GAPDH is involved in rTalpha translocation from the RIS to the ROS during dark adaptation.
Collapse
Affiliation(s)
- Junping Chen
- Oklahoma Center for Neuroscience (OCNS), The University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| | | | | | | | | | | |
Collapse
|
27
|
Kerov V, Rubin WW, Natochin M, Melling NA, Burns ME, Artemyev NO. N-terminal fatty acylation of transducin profoundly influences its localization and the kinetics of photoresponse in rods. J Neurosci 2007; 27:10270-7. [PMID: 17881533 PMCID: PMC6672661 DOI: 10.1523/jneurosci.2494-07.2007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
N-terminal acylation of the alpha-subunits of heterotrimeric G-proteins is believed to play a major role in regulating the cellular localization and signaling of G-proteins, but physiological evidence has been lacking. To examine the functional significance of N-acylation of a well understood G-protein alpha-subunit, transducin (G alpha(t)), we generated transgenic mice that expressed a mutant G alpha(t) lacking N-terminal acylation sequence (G alpha(t)G2A). Rods expressing G alpha(t)G2A showed a severe defect in transducin cellular localization. In contrast to native G alpha(t), which resides in the outer segments of dark-adapted rods, G alpha(t)G2A was found predominantly in the inner compartments of the photoreceptor cells. Remarkably, transgenic rods with the outer segments containing G alpha(t)G2A at 5-6% of the G alpha(t) levels in wild-type rods showed only a sixfold reduction in sensitivity and a threefold decrease in the amplification constant. The much smaller than predicted reduction may reflect an increase in the lateral diffusion of transducin and an increased activation rate by photoexcited rhodopsin or more efficient activation of cGMP phosphodiesterase 6 by G alpha(t)G2A; alternatively, nonlinear relationships between concentration and the activation rate of transducin also potentially contribute to the mismatch between the amplification constant and quantitative expression analysis of G alpha(t)G2A rods. Furthermore, the G2A mutation reduced the GTPase activity of transducin and resulted in two to three times slower than normal recovery of flash responses of transgenic rods, indicating the role of G alpha(t) membrane tethering for its efficient inactivation by the regulator of G-protein signaling 9 GTPase-activating protein complex. Thus, N-acylation is critical for correct compartmentalization of transducin and controls the rate of its deactivation.
Collapse
Affiliation(s)
- Vasily Kerov
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa 52242, and
| | - William W. Rubin
- Center for Neuroscience and Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California 95616
| | - Michael Natochin
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa 52242, and
| | - Nathan A. Melling
- Center for Neuroscience and Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California 95616
| | - Marie E. Burns
- Center for Neuroscience and Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California 95616
| | - Nikolai O. Artemyev
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa 52242, and
| |
Collapse
|
28
|
Saini DK, Kalyanaraman V, Chisari M, Gautam N. A family of G protein βγ subunits translocate reversibly from the plasma membrane to endomembranes on receptor activation. J Biol Chem 2007; 282:24099-108. [PMID: 17581822 PMCID: PMC2238721 DOI: 10.1074/jbc.m701191200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present model of G protein activation by G protein-coupled receptors exclusively localizes their activation and function to the plasma membrane (PM). Observation of the spatiotemporal response of G protein subunits in a living cell to receptor activation showed that 6 of the 12 members of the G protein gamma subunit family translocate specifically from the PM to endomembranes. The gamma subunits translocate as betagamma complexes, whereas the alpha subunit is retained on the PM. Depending on the gamma subunit, translocation occurs predominantly to the Golgi complex or the endoplasmic reticulum. The rate of translocation also varies with the gamma subunit type. Different gamma subunits, thus, confer distinct spatiotemporal properties to translocation. A striking relationship exists between the amino acid sequences of various gamma subunits and their translocation properties. gamma subunits with similar translocation properties are more closely related to each other. Consistent with this relationship, introducing residues conserved in translocating subunits into a non-translocating subunit results in a gain of function. Inhibitors of vesicle-mediated trafficking and palmitoylation suggest that translocation is diffusion-mediated and controlled by acylation similar to the shuttling of G protein subunits (Chisari, M., Saini, D. K., Kalyanaraman, V., and Gautam, N. (2007) J. Biol. Chem. 282, 24092-24098). These results suggest that the continual testing of cytosolic surfaces of cell membranes by G protein subunits facilitates an activated cell surface receptor to direct potentially active G protein betagamma subunits to intracellular membranes.
Collapse
Affiliation(s)
- Deepak Kumar Saini
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Vani Kalyanaraman
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Mariangela Chisari
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Narasimhan Gautam
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
- To whom correspondence should be addressed: Box 8054, Washington University School of Medicine, St. Louis, MO 63110. Tel.: 314-362 8568; Fax: 314-362-8571; E-mail:
| |
Collapse
|
29
|
Marrari Y, Crouthamel M, Irannejad R, Wedegaertner PB. Assembly and trafficking of heterotrimeric G proteins. Biochemistry 2007; 46:7665-77. [PMID: 17559193 PMCID: PMC2527407 DOI: 10.1021/bi700338m] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To be activated by cell surface G protein-coupled receptors, heterotrimeric G proteins must localize at the cytoplasmic surface of plasma membranes. Moreover, some G protein subunits are able to traffic reversibly from the plasma membrane to intracellular locations upon activation. This current topic will highlight new insights into how nascent G protein subunits are assembled and how they arrive at plasma membranes. In addition, recent reports have increased our knowledge of activation-induced trafficking of G proteins. Understanding G protein assembly and trafficking will lead to a greater understanding of novel ways that cells regulate G protein signaling.
Collapse
Affiliation(s)
| | | | | | - Philip B. Wedegaertner
- *address correspondence to: Philip B. Wedegaertner, Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 S. 10 St., 839 BLSB, Philadelphia, PA 19107, Tel: 215-503-3137, Fax: 215-923-2117, e-mail:
| |
Collapse
|
30
|
Rosenzweig DH, Nair KS, Wei J, Wang Q, Garwin G, Saari JC, Chen CK, Smrcka AV, Swaroop A, Lem J, Hurley JB, Slepak VZ. Subunit dissociation and diffusion determine the subcellular localization of rod and cone transducins. J Neurosci 2007; 27:5484-94. [PMID: 17507570 PMCID: PMC2655354 DOI: 10.1523/jneurosci.1421-07.2007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Activation of rod photoreceptors by light induces a massive redistribution of the heterotrimeric G-protein transducin. In darkness, transducin is sequestered within the membrane-enriched outer segments of the rod cell. In light, it disperses throughout the entire neuron. We show here that redistribution of rod transducin by light requires activation, but it does not require ATP. This observation rules out participation of molecular motors in the redistribution process. In contrast to the light-stimulated redistribution of rod transducin in rods, cone transducin in cones does not redistribute during activation. Remarkably, when cone transducin is expressed in rods, it does undergo light-stimulated redistribution. We show here that the difference in subcellular localization of activated rod and cone G-proteins correlates with their affinity for membranes. Activated rod transducin releases from membranes, whereas activated cone transducin remains bound to membranes. A synthetic peptide that dissociates G-protein complexes independently of activation facilitates dispersion of both rod and cone transducins within the cells. This peptide also facilitates detachment of both G-proteins from the membranes. Together, these results show that it is the dissociation state of transducin that determines its localization in photoreceptors. When rod transducin is stimulated, its subunits dissociate, leave outer segment membranes, and equilibrate throughout the cell. Cone transducin subunits do not dissociate during activation and remain sequestered within the outer segment. These findings indicate that the subunits of some heterotrimeric G-proteins remain associated during activation in their native environments.
Collapse
Affiliation(s)
- Derek H. Rosenzweig
- Department of Molecular and Cellular Pharmacology and Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - K. Saidas Nair
- Department of Molecular and Cellular Pharmacology and Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida 33136
| | | | - Qiang Wang
- Department of Molecular and Cellular Pharmacology and Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Greg Garwin
- Ophthalmology, University of Washington, Seattle, Washington 98195
| | - John C. Saari
- Ophthalmology, University of Washington, Seattle, Washington 98195
| | - Ching-Kang Chen
- Department of Biochemistry, Virginia Commonwealth University, Richmond, Virginia 23284
| | - Alan V. Smrcka
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14603
| | - Anand Swaroop
- Departments of Ophthalmology and Visual Sciences, and Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, and
| | - Janis Lem
- Molecular Cardiology Research Institute, Tufts–New England Medical Center, Boston, Massachusetts 02111
| | | | - Vladlen Z. Slepak
- Department of Molecular and Cellular Pharmacology and Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida 33136
| |
Collapse
|
31
|
Lobanova ES, Finkelstein S, Song H, Tsang SH, Chen CK, Sokolov M, Skiba NP, Arshavsky VY. Transducin translocation in rods is triggered by saturation of the GTPase-activating complex. J Neurosci 2007; 27:1151-60. [PMID: 17267570 PMCID: PMC6673185 DOI: 10.1523/jneurosci.5010-06.2007] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Light causes massive translocation of G-protein transducin from the light-sensitive outer segment compartment of the rod photoreceptor cell. Remarkably, significant translocation is observed only when the light intensity exceeds a critical threshold level. We addressed the nature of this threshold using a series of mutant mice and found that the threshold can be shifted to either a lower or higher light intensity, dependent on whether the ability of the GTPase-activating complex to inactivate GTP-bound transducin is decreased or increased. We also demonstrated that the threshold is not dependent on cellular signaling downstream from transducin. Finally, we showed that the extent of transducin alpha subunit translocation is affected by the hydrophobicity of its acyl modification. This implies that interactions with membranes impose a limitation on transducin translocation. Our data suggest that transducin translocation is triggered when the cell exhausts its capacity to activate transducin GTPase, and a portion of transducin remains active for a sufficient time to dissociate from membranes and to escape from the outer segment. Overall, the threshold marks the switch of the rod from the highly light-sensitive mode of operation required under limited lighting conditions to the less-sensitive energy-saving mode beneficial in bright light, when vision is dominated by cones.
Collapse
Affiliation(s)
- Ekaterina S. Lobanova
- Albert Eye Research Institute, Duke University Medical Center, Durham, North Carolina 27710
| | - Stella Finkelstein
- Albert Eye Research Institute, Duke University Medical Center, Durham, North Carolina 27710
| | - Hongman Song
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Stephen H. Tsang
- Brown Glaucoma Laboratory, Edward Harkness Eye Institute, Columbia University, New York, New York 10032
| | - Ching-Kang Chen
- Department of Biochemistry, Virginia Commonwealth University, Richmond, Virginia 23298, and
| | - Maxim Sokolov
- Sensory Neuroscience Research Center and West Virginia University Eye Institute, Morgantown, West Virginia 26506
| | - Nikolai P. Skiba
- Albert Eye Research Institute, Duke University Medical Center, Durham, North Carolina 27710
| | - Vadim Y. Arshavsky
- Albert Eye Research Institute, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
32
|
Abstract
Phototransduction is the process by which light triggers an electrical signal in a photoreceptor cell. Image-forming vision in vertebrates is mediated by two types of photoreceptors: the rods and the cones. In this review, we provide a summary of the success in which the mouse has served as a vertebrate model for studying rod phototransduction, with respect to both the activation and termination steps. Cones are still not as well-understood as rods partly because it is difficult to work with mouse cones due to their scarcity and fragility. The situation may change, however.
Collapse
Affiliation(s)
- Yingbin Fu
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
33
|
Calvert PD, Strissel KJ, Schiesser WE, Pugh EN, Arshavsky VY. Light-driven translocation of signaling proteins in vertebrate photoreceptors. Trends Cell Biol 2006; 16:560-8. [PMID: 16996267 DOI: 10.1016/j.tcb.2006.09.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 08/07/2006] [Accepted: 09/07/2006] [Indexed: 11/16/2022]
Abstract
The dynamic localization of proteins within cells is often determined by environmental stimuli. In retinal photoreceptors, light exposure results in the massive translocation of three key signal transduction proteins, transducin, arrestin and recoverin, into and out of the outer segment compartment where phototransduction takes place. This phenomenon has rapidly taken the center stage of photoreceptor cell biology, thanks to the introduction of new quantitative and transgenic approaches. Here, we discuss evidence that intracellular protein translocation contributes to adaptation of photoreceptors to diurnal changes in ambient light intensity and summarize the current debate on whether it is driven by diffusion or molecular motors.
Collapse
Affiliation(s)
- Peter D Calvert
- Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | | | | |
Collapse
|
34
|
Wada Y, Sugiyama J, Okano T, Fukada Y. GRK1 and GRK7: unique cellular distribution and widely different activities of opsin phosphorylation in the zebrafish rods and cones. J Neurochem 2006; 98:824-37. [PMID: 16787417 DOI: 10.1111/j.1471-4159.2006.03920.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Retinal cone cells exhibit distinctive photoresponse with a more restrained sensitivity to light and a more rapid shutoff kinetics than those of rods. To understand the molecular basis for these characteristics of cone responses, we focused on the opsin deactivation process initiated by G protein-coupled receptor kinase (GRK) 1 and GRK7 in the zebrafish, an animal model suitable for studies on retinal physiology and biochemistry. Screening of the ocular cDNAs identified two homologs for each of GRK1 (1A and 1B) and GRK7 (7-1 and 7-2), and they were classified into three GRK subfamilies, 1 A, 1B and 7 by phylogenetic analysis. In situ hybridization and immunohistochemical studies localized both GRK1B and GRK7-1 in the cone outer segments and GRK1A in the rod outer segments. The opsin/GRKs molar ratio was estimated to be 569 in the rod and 153 in the cone. The recombinant GRKs phosphorylated light-activated rhodopsin, and the Vmax value of the major cone subtype, GRK7-1, was 32-fold higher than that of the rod kinase, GRK1A. The reinforced activity of the cone kinase should provide a strengthened shutoff mechanism of the light-signaling in the cone and contribute to the characteristics of the cone responses by reducing signal amplification efficiency.
Collapse
Affiliation(s)
- Yasutaka Wada
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
35
|
Ueda Y, Tammitsu N, Imai H, Honda Y, Shichida Y. Recovery of rod-mediated a-wave during light-adaptation in mGluR6-deficient mice. Vision Res 2006; 46:1655-64. [PMID: 16243375 DOI: 10.1016/j.visres.2005.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 09/07/2005] [Accepted: 09/07/2005] [Indexed: 11/30/2022]
Abstract
The purpose of this study was to compare the a-waves of mGluR6-deficient mice (KO) to that of wild-type mice (WT), and to determine whether the light-adapted electroretinogram of the KO mice originate exclusively from cones. Dark-adapted a-waves were recorded under the same conditions from both types of mice. With a 96-cd/m(2) background, the a-wave from both types of mice showed a rapid recovery over a 50-min period. The analysis of the a-waves in KO mice indicated that the recovery was determined mainly by the rod component. The light-adapted b-wave of WT mice showed no corresponding recovery. We conclude that rod contribution must be considered in the analyses of the light-adapted a-waves of KO mice.
Collapse
Affiliation(s)
- Yoshiki Ueda
- Department of Ophthalmology, Nagahama City Hospital, Oh-Inuicho, Nagahama, Shiga, Japan.
| | | | | | | | | |
Collapse
|
36
|
Strissel KJ, Sokolov M, Trieu LH, Arshavsky VY. Arrestin translocation is induced at a critical threshold of visual signaling and is superstoichiometric to bleached rhodopsin. J Neurosci 2006; 26:1146-53. [PMID: 16436601 PMCID: PMC6674573 DOI: 10.1523/jneurosci.4289-05.2006] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Light induces massive translocation of major signaling proteins between the subcellular compartments of photoreceptors. Among them is visual arrestin responsible for quenching photoactivated rhodopsin, which moves into photoreceptor outer segments during illumination. Here, for the first time, we determined the light dependency of arrestin translocation, which revealed two key features of this phenomenon. First, arrestin translocation is triggered when the light intensity approaches a critical threshold corresponding to the upper limits of the normal range of rod responsiveness. Second, the amount of arrestin entering rod outer segments under these conditions is superstoichiometric to the amount of photoactivated rhodopsin, exceeding it by at least 30-fold. We further showed that it is not the absolute amount of excited rhodopsin but rather the extent of downstream cascade activity that triggers translocation. Finally, we demonstrated that the total amount of arrestin in the rod cell is nearly 10-fold higher than previously thought and therefore sufficient to inactivate the entire pool of rhodopsin at any level of illumination. Thus, arrestin movement to the outer segment leads to an increase in the free arrestin concentration and thereby may serve as a powerful mechanism of light adaptation.
Collapse
Affiliation(s)
- Katherine J Strissel
- Department of Ophthalmology, Harvard Medical School, The Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|