Phosphorylation of G Protein-Coupled Receptors: from the Barcode Hypothesis to the Flute Model

Total Page:16

File Type:pdf, Size:1020Kb

Phosphorylation of G Protein-Coupled Receptors: from the Barcode Hypothesis to the Flute Model 1521-0111/92/3/201–210$25.00 https://doi.org/10.1124/mol.116.107839 MOLECULAR PHARMACOLOGY Mol Pharmacol 92:201–210, September 2017 Copyright ª 2017 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. MINIREVIEW—MOLECULAR PHARMACOLOGY IN CHINA Phosphorylation of G Protein-Coupled Receptors: From the Barcode Hypothesis to the Flute Model Zhao Yang, Fan Yang, Daolai Zhang, Zhixin Liu, Amy Lin, Chuan Liu, Peng Xiao, Xiao Yu, and Jin-Peng Sun Downloaded from Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People’s Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People’s Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.) Received December 10, 2016; accepted February 23, 2017 molpharm.aspetjournals.org ABSTRACT Seven transmembrane G protein-coupled receptors (GPCRs) distinct functional outcomes. Our recent work using unnatural are often phosphorylated at the C terminus and on intracellular amino acid incorporation and fluorine-19 nuclear magnetic loops in response to various extracellular stimuli. Phosphoryla- resonance (19F-NMR) spectroscopy led to the flute model, tion of GPCRs by GPCR kinases and certain other kinases which provides preliminary insight into the receptor phospho- can promote the recruitment of arrestin molecules. The arrestins coding mechanism, by which receptor phosphorylation pat- critically regulate GPCR functions not only by mediating terns are recognized by an array of phosphate-binding pockets receptor desensitization and internalization, but also by redi- on arrestin and are translated into distinct conformations. rectingsignalingtoGprotein-independent pathways via These selective conformations are recognized by various at ASPET Journals on September 24, 2021 interactions with numerous downstream effector molecules. effector molecules downstream of arrestin. The phospho- Accumulating evidence over the past decade has given rise barcoding mechanism enables arrestin to recognize a wide to the phospho-barcode hypothesis, which states that ligand- range of phosphorylation patterns of GPCRs, contributing to specific phosphorylation patterns of a receptor direct its their diverse functions. Introduction activation, most GPCRs “floating” on the plasma membrane are phosphorylated at sites located on intracellular loops or Seven transmembrane-spanning G protein-coupled recep- C-terminal tails (Table 1). tors (GPCRs) comprise the largest known membrane protein Many different phosphorylation sites in different GPCRs family encoded by the human genome, and GPCRs regulate have been identified, mostly by mass spectrometry or almost all the known physiologic processes in humans by phospho-specific antibodies. By contrast, the functions of converting a broad range of extracellular stimuli (ranging receptor phosphorylation are often established by muta- from light to hormones and neurotransmitters) to intracellu- genesis both in vitro and in vivo (Budd et al., 2000; Jones lar signals (Ritter and Hall, 2009; Manglik and Kobilka, 2014; et al., 2007; Busillo et al., 2010; Bradley et al., 2016). In cells, Wisler et al., 2014; Dohlman, 2015). Upon ligand binding and the phosphorylation process is mediated by at least two classes of serine/threonine kinases, including the second This work was supported by the National Key Basic Research Program of messenger-dependent (e.g., PKA and protein kinase C China [Grant 2013CB967700]; the Fundamental Research Funds of Shandong [PKC]) and -independent kinases (i.e., GPCR kinases [GRKs]) University [Grants 2014JC029, 2016HW005]; the National Natural Science (Lefkowitz, 1998). As a classic paradigm, phosphorylation of Foundation of China [Grants 31470789, 31611540337]; the Shandong Natural Science Fund for Distinguished Young Scholars [Grants JQ201320, JQ201517]; receptors by the former type of kinases is independent of and the Program for Changjiang Scholars and Innovative Research Team in ligand binding and directly uncouples the receptors from their University [Grant IRT13028]. Z.Y. and F.Y. contributed equally to this work. cognate G proteins, leading to heterologous desensitization https://doi.org/10.1124/mol.116.107839. (Hausdorff et al., 1990). In contrast, receptor phosphorylation ABBREVIATIONS: Akt, protein kinase B; b2AR, b2-adrenergic receptor; AT1aR, angiotensin II receptor type I; BRET, bioluminescence resonance energy transfer; ERK, extracellular signal–regulated kinase; 19F-NMR, fluorine-19 nuclear magnetic resonance; GPCR, G protein-coupled receptor; GPR120, G-protein coupled receptor 120; GRK, G protein-coupled receptor kinase; GRK2, G protein-coupled receptor kinase 2, or b-adrenergic receptor kinase 1; GRK2pp, GRK2-phosphopeptides; ICL3, third intracellular loop; M3-mAChR, M3-muscarinic acetylcholine receptor; PKA, protein kinase A; PKC, protein kinase C; Thr, threonine; V2R, V2 vasopressin receptor. 201 TABLE 1. 202 Examples of receptor phosphorylation at C-terminus and corresponding functions GPCR Phosphorylation Sites Kinase Method Cell Type Function References Angiotensin II receptor T332, S335, T336, PKC/GRK Mut CHO Internalization (Thomas et al., 1998) Yang et al. type I (AT1aR) S338 S331, S338, S348 PKC Mut CHO ND (Qian et al., 1999) b2 adrenergic receptor (b2AR) S345, S346 PKA Mut Mouse L cells Desensitization (Clark et al., 1989) S355, S356, S364 GRK Mut HEK293 Desensitization (Seibold et al., 2000) S355, S356 GRK6 MS and P-SA HEK293 Desensitization and b-arrestin-mediated (Nobles et al., 2011) ERK1/2 activation In vitro b-arrestin-1 mediated SRC activation (Yang et al., 2015) T360, S364, S396, GRK2 MS and P-SA HEK293 Desensitization and internalization (Nobles et al., 2011) S401, S407, S411 In vitro b-arrestin-1 mediated clathrin recruitment (Yang et al., 2015) and internalization T384, S396, S401, GRK2 PAA In vitro ND (Fredericks et al., 1996) and S407 T384, T393, S396, GRK5 PAA In vitro ND (Fredericks et al., 1996) S401, S407, S411 C-X-C motif chemokine S324, S325 PKC/GRK6 MS and P-SA HEK293 Receptor degradation (Busillo et al., 2010) receptor type 4 (CXCR-4) S330/339, cluster GRK2/6 MS and P-SA HEK293 b-arrestin-2 recruitment (Busillo et al., 2010) S346-S352 and calcium mobilization S330/339, cluster GRK3/6 MS and P-SA HEK293 b-arrestin-1 mediated ERK1/2 activation (Busillo et al., 2010) S346-S352 Dopamine receptor D1 (DRD1) T347 ND Mut HEK293 Promote secondary phosphorylation (Kim et al., 2004) of residues within the ICL3 T360 GRK2 Mut CHO Desensitization (Lamey et al., 2002) T428, S431 GRK4 Mut HEK293T b-arrestin-1 mediated internalization (Rankin et al., 2006) S431, T439, T446 ND Mut CHO Internalization (Lamey et al., 2002) Follicle-stimulating Cluster T638-T644 GRK2 Mut HEK293 b-arrestin-mediated desensitization (Kara et al., 2006) hormone receptor (FSHR) and internalization GABAB receptor (GABABR) S783 AMPK MS and EDA Rat hippocampal Receptor recycling (Terunuma et al., 2010) neurons S867 CaMKII MS Rat hippocampal Internalization (Guetg et al., 2010) neurons S892 PKA Mut HEK293 Internalization (Couve et al., 2002) Ghrelin receptor type S349, T350 ND MS HEK293 Stabilization of the receptor-b-arrestin1/2 (Bouzo-Lorenzo et al., 2016) 1a (GHSR1a) complex S362, S363, T366 ND MS HEK293 b-arrestin1/2 binding and internalization (Bouzo-Lorenzo et al., 2016) G protein-coupled T347, S350, S357 PKC/GRK6 Mut HEK293 b-arrestin-2 recruitment and calcium (Burns et al., 2014) receptor 120 (GPR120) mobilization T347, T349, S350 ND MS CHO Flp-In cells b-arrestin-2 recruitment and Akt activation (Prihandoko et al., 2016) S357, S361 ND MS CHO Flp-In cells b-arrestin-2 recruitment and internalization (Prihandoko et al., 2016) m-opioid receptor (MOR) S356, S357, S363 PKC MS HEK293 Desensitization (Wang et al., 2002; Chen et al., 2013) T370 CaMKII MS HEK293 ND (Chen et al., 2013) S375 GRK2 MS HEK293 b-arrestin-2 recruitment and desensitization (Chen et al., 2013; Lowe et al., 2015) Thyrotropin-releasing S355, S360, S364, GRK2 Mut and P-SA CHO b-arrestin-2-mediated desensitization (Jones et al., 2007) hormone receptor (TRHR) T365 and internalization T365, T371, S383 CK2 Mut COS-1 b-arrestin-1-dependent internalization (Hanyaloglu et al., 2001) Orexin-2 receptor (OX2R) Cluster T399-S403 GRK Mut HEK293FT ND (Jaeger et al., 2014) Cluster S406-T409, GRK Mut HEK293FT Receptor-b-arrestin1/2-ubiquitin (Jaeger et al., 2014) Cluster T427-T431 complex formation AMPK, 5ʹ-AMP-activated protein kinase; CaMKII, Ca2+/calmodulin-dependent protein kinase II; CHO, Chinese hamster ovary cells; CK2, casein kinase II; COS-1, CV-1 in origin with SV40 genes monkey kidney cells; EDA, Edman degradation analysis; HEK293, human embryonic kidney 293 cells; Mut, mutagenesis; MS, mass spectrometry; ND, not determined; P-SA, phospho-specific antibody; PAA, phospho-amino acid analysis. Downloaded from from Downloaded molpharm.aspetjournals.org at ASPET Journals on September 24, 2021 24, September on Journals ASPET at Phosphorylation Barcoding of the GPCR 203 by GRKs, a kinase family consisting of seven members, is so the mechanism by which the phospho-barcode is recognized ligand-stimulation dependent and is followed by the recruit- and then converted to specific signaling remains largely ment of arrestin molecules to the receptor that sterically unknown,
Recommended publications
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • Rhodopsin Kinase Activity Modulates the Amplitude of the Visual Response in Drosophila
    Rhodopsin kinase activity modulates the amplitude of the visual response in Drosophila Seung-Jae Lee*, Hong Xu, and Craig Montell† Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 Edited by Robert J. Lefkowitz, Duke University Medical Center, Durham, NC, and approved June 30, 2004 (received for review March 29, 2004) A feature shared between Drosophila rhodopsin and nearly all tractability of fly genetics (14). As in mammals, light-activated other G protein-coupled receptors is agonist-dependent protein rhodopsin is phosphorylated and interacts with a protein, arres- phosphorylation. Despite extensive analyses of Drosophila photo- tin, which facilitates deactivation of the receptor. However, transduction, the identity and function of the rhodopsin kinase unlike mammalian phototransduction, light activation in Dro- (RK) have been elusive. Here, we provide evidence that G protein- sophila is coupled to stimulation of phospholipase C rather than coupled receptor kinase 1 (GPRK1), which is most similar to the a cGMP-phosphodiesterase. ␤-adrenergic receptor kinases, G protein-coupled receptor kinase 2 Two Drosophila genes encoding putative GRKs (GPRK1 and (GRK2) and GRK3, is the fly RK. We show that GPRK1 is enriched in GPRK2) were isolated more than a decade ago (15), but were photoreceptor cells, associates with the major Drosophila rhodop- dismissed as candidate RKs due in part to their much greater sin, Rh1, and phosphorylates the receptor. As is the case with similarity to mammalian nonvisual GRKs than to mammalian mammalian GRK2 and GRK3, Drosophila GPRK1 includes a C- RKs. Subsequent analysis of GPRK2 demonstrated that it is terminal pleckstrin homology domain, which binds to phosphoi- expressed in the ovaries and required for egg morphogenesis nositides and the G␤␥ subunit.
    [Show full text]
  • Balancing the Photoreceptor Proteome: Proteostasis Network Therapeutics for Inherited Retinal Disease
    G C A T T A C G G C A T genes Review Balancing the Photoreceptor Proteome: Proteostasis Network Therapeutics for Inherited Retinal Disease Siebren Faber and Ronald Roepman * Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands * Correspondence: [email protected] Received: 10 June 2019; Accepted: 16 July 2019; Published: 24 July 2019 Abstract: The light sensing outer segments of photoreceptors (PRs) are renewed every ten days due to their high photoactivity, especially of the cones during daytime vision. This demands a tremendous amount of energy, as well as a high turnover of their main biosynthetic compounds, membranes, and proteins. Therefore, a refined proteostasis network (PN), regulating the protein balance, is crucial for PR viability. In many inherited retinal diseases (IRDs) this balance is disrupted leading to protein accumulation in the inner segment and eventually the death of PRs. Various studies have been focusing on therapeutically targeting the different branches of the PR PN to restore the protein balance and ultimately to treat inherited blindness. This review first describes the different branches of the PN in detail. Subsequently, insights are provided on how therapeutic compounds directed against the different PN branches might slow down or even arrest the appalling, progressive blinding conditions. These insights are supported by findings of PN modulators in other research disciplines. Keywords: protein trafficking; protein folding; protein degradation; chaperones; chaperonins; heat shock response; unfolded protein response; autophagy; therapy 1. Introduction The rod and cone photoreceptor (PR) cells are the most abundant cell types in the human retina, with ~6.4 million cones and up to 125 million rods per adult retina [1].
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2010/0158968 A1 Panitch Et Al
    US 20100158968A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2010/0158968 A1 Panitch et al. (43) Pub. Date: Jun. 24, 2010 (54) CELL-PERMEANT PEPTIDE-BASED Publication Classification INHIBITOR OF KINASES (51) Int. Cl. (76) Inventors: Alyssa Panitch, West Lafayette, IN st e8 CR (US); Brandon Seal, West ( .01) Lafayette, IN (US) A638/10 (2006.01) s A638/16 (2006.01) Correspondence Address: A6IP 43/00 (2006.01) GREENBERG TRAURIG, LLP (52) U.S. Cl. ................ 424/422:514/15: 514/13: 514/14 200 PARKAVE., P.O. BOX 677 FLORHAMPARK, NJ 07932 (US) (57) ABSTRACT The described invention provides kinase inhibiting composi (21) Appl. No.: 12/634,476 tions containing a therapeutic amount of a therapeutic inhibi (22) Filed: Dec. 9, 2009 torpeptide that inhibits at least one kinase enzyme, methods e 19 for treating an inflammatory disorder whose pathophysiology comprises inflammatory cytokine expression, and methods Related U.S. Application Data for treating an inflammatory disorder whose pathophysiology (60) Provisional application No. 61/121,396, filed on Dec. comprises inflammatory cytokine expression using the kinase 10, 2008. inhibiting compositions. 20 { ki> | 0: & c s - --- 33- x: SE PEPELE, ics 1.-- E- X K. AAA 22.9 --- KKK. Y.A., 3.2; C. -r { AAEASA. A. E. i : A X AAAAAAA; ; ; ; :-n. 4:-: is SEEKESAN.ARESA, 3523 -- -- Yili.A.R.AKA: 5,342 3. {{RCE: Rix i: Patent Application Publication US 2010/0158968A1 & ******** NO s ***** · Patent Application Publication Jun. 24, 2010 Sheet 2 of 11 US 2010/0158968A1 it, O Peptide: Cso: g E 100 WRRKAWRRKANRO, GWAA.
    [Show full text]
  • This Thesis Has Been Submitted in Fulfilment of the Requirements for a Postgraduate Degree (E.G
    This thesis has been submitted in fulfilment of the requirements for a postgraduate degree (e.g. PhD, MPhil, DClinPsychol) at the University of Edinburgh. Please note the following terms and conditions of use: This work is protected by copyright and other intellectual property rights, which are retained by the thesis author, unless otherwise stated. A copy can be downloaded for personal non-commercial research or study, without prior permission or charge. This thesis cannot be reproduced or quoted extensively from without first obtaining permission in writing from the author. The content must not be changed in any way or sold commercially in any format or medium without the formal permission of the author. When referring to this work, full bibliographic details including the author, title, awarding institution and date of the thesis must be given. Molecular mechanisms underlying Retinitis pigmentosa type 2 Rodanthi Lyraki Thesis submitted for the degree of Doctor of Philosophy University of Edinburgh 2017 Declaration I declare that this thesis is my own work, and that the experiments described here were conducted by me except where explicitly stated. This work has not been submitted for any other degree or professional qualification. Rodanthi Lyraki, August 2017 ii Preface “Photoreceptors sit on a knife edge separating function and survival from dysfunction and death, and almost any defect seems capable of tipping them towards cell death.” - Alan F. Wright et al., “Photoreceptor degeneration: genetic and mechanistic dissection of a complex trait” iii Acknowledgements I feel very fortunate to have carried out my PhD in the Institute of Genetics and Molecular Medicine in Edinburgh, where I had the opportunity to interact with first- class scientists on a daily basis.
    [Show full text]
  • Abnormal Photoresponses and Light-Induced Apoptosis in Rods Lacking Rhodopsin Kinase
    Proc. Natl. Acad. Sci. USA Vol. 96, pp. 3718–3722, March 1999 Cell Biology Abnormal photoresponses and light-induced apoptosis in rods lacking rhodopsin kinase CHING-KANG CHEN*†,MARIE E. BURNS†‡,MARIBETH SPENCER§,GREGORY A. NIEMI§,JEANNIE CHEN¶, i JAMES B. HURLEY§,DENIS A. BAYLOR‡, AND MELVIN I. SIMON* *Division of Biology, 147-75, California Institute of Technology, Pasadena, CA 91125; ‡Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305; §Howard Hughes Medical Institute and Department of Biochemistry, Box 357370, University of Washington, Seattle, WA 98195; and ¶Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA 90033 Contributed by Melvin Simon, January 12, 1999 ABSTRACT Phosphorylation is thought to be an essential 10) and phosphorylate rhodopsin’s C-terminal residues equally first step in the prompt deactivation of photoexcited rhodop- well in vitro (11), it is unclear which kinase is mainly responsible sin. In vitro, the phosphorylation can be catalyzed either by for rhodopsin deactivation in vivo. We determined the role of rhodopsin kinase (RK) or by protein kinase C (PKC). To RK in rhodopsin deactivation in intact rods by deactivating investigate the specific role of RK, we inactivated both alleles both alleles of the RK gene. We found that RK is required for of the RK gene in mice. This eliminated the light-dependent the normal deactivation of rhodopsin and that in its absence, phosphorylation of rhodopsin and caused the single-photon dramatic functional and structural changes occurred. response to become larger and longer lasting than normal. These results demonstrate that RK is required for normal rhodopsin deactivation.
    [Show full text]
  • Control of Rhodopsin Activity in Vision
    Control of rhodopsin DENIS A. BAYLOR, MARIE E. BURNS activity in vision Abstract high concentration in the cytoplasm in darkness and that binds to cationic channels in the Although rhodopsin's role in activating the surface membrane, holding them open. phototransduction cascade is well known, the Hydrolysis of cGMP allows the channels to processes that deactivate rhodopsin, and thus close, interrupting an inward current of sodium, the rest of the cascade, are less well calcium and magnesium ions and producing a understood. At least three proteins appear to hyperpolarisation of the membrane. The play a role: rhodopsin kinase, arrestin and hyperpolarisation reduces the rate at which recoverin. Here we review recent physiological neurotransmitter is released from the synaptic studies of the molecular mechanisms of terminal of the rod. rhodopsin deactivation. The approach was to The purpose of this paper is to review recent monitor the light responses of individual work on the important but still poorly mouse rods in which rhodopsin was altered or understood mechanisms that terminate the arrestin was deleted by transgenic techniques. light-evoked catalytic activity of rhodopsin. Removal of rhodopsin's carboxy-terminal These mechanisms, which fix the intensity and residues which contain phosphorylation sites duration of the activation of the transduction implicated in deactivation, prolonged the flash cascade, need to satisfy strong functional response 20-fold and caused it to become constraints. Rhodopsin activity must be highly variable. In rods that did not express arrestin the flash response recovered partially, terminated rapidly so that an absorbed photon but final recovery was slowed over lOO-fold.
    [Show full text]
  • The Lar Protein Tyrosine Phospatase Enables DPGF B-Receptor Activation
    THE LAR PROTEIN TYROSINE PHOSPHATASE ENABLES PDGF β-RECEPTOR ACTIVATION AND SIGNAL TRANSDUCTION by WEI ZHENG A thesis submitted to The University of Birmingham for the degree of DOCTOR OF PHILOSOPHY School of Biosciences The University of Birmingham April 2013 University of Birmingham Research Archive e-theses repository This unpublished thesis/dissertation is copyright of the author and/or third parties. The intellectual property rights of the author or third parties in respect of this work are as defined by The Copyright Designs and Patents Act 1988 or as modified by any successor legislation. Any use made of information contained in this thesis/dissertation must be in accordance with that legislation and must be properly acknowledged. Further distribution or reproduction in any format is prohibited without the permission of the copyright holder. This work was published, please quote the reference. The published paper was written in collaboration with the supervisor and other authors. ABSTRACT Many cellular activities including cell survival, proliferation, migration and differentiation are controlled by growth factors and their corresponding tyrosine kinases receptor (RTKs). Growth factor receptor activation is strictly regulated by protein tyrosine phosphatases (PTPs). Here I investigated whether the receptor protein tyrosine phosphatase (RPTP) LAR, which is known to modify the activity of several RTKs, also regulates platelet derived growth factor (PDGF) receptor activity and signalling. Mouse embryonic fibroblasts (MEFs) expressing mutant LAR lacking its phosphatase domains (LARΔP) showed reduced phosphorylation of PDGFβ receptor (PDGFβR) compared with wild type (WT) cells. This was rescued by re-expression of WT LAR. The decreased phosphorylation of the PDGFβR was independent of ligand concentration and occurred on all tyrosine residues, suggesting that LAR is required for full PDGFβR kinase activation.
    [Show full text]
  • Reprogramming the Retina: Next Generation Strategies of Retinal Neuroprotection and Gene Therapy Vector Potency Assessment
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2018 Reprogramming The Retina: Next Generation Strategies Of Retinal Neuroprotection And Gene Therapy Vector Potency Assessment Devin Scott Mcdougald University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Genetics Commons, Molecular Biology Commons, and the Virology Commons Recommended Citation Mcdougald, Devin Scott, "Reprogramming The Retina: Next Generation Strategies Of Retinal Neuroprotection And Gene Therapy Vector Potency Assessment" (2018). Publicly Accessible Penn Dissertations. 3158. https://repository.upenn.edu/edissertations/3158 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/3158 For more information, please contact [email protected]. Reprogramming The Retina: Next Generation Strategies Of Retinal Neuroprotection And Gene Therapy Vector Potency Assessment Abstract Mutations within over 250 known genes are associated with inherited retinal degeneration. Clinical success following gene replacement therapy for Leber’s congenital amaurosis type 2 establishes a platform for the development of downstream treatments targeting other forms of inherited and acquired ocular disease. Unfortunately, several challenges relevant to complex disease pathology and limitations of current gene transfer technologies impede the development of gene replacement for each specific form of retinal degeneration. Here we describe gene augmentation strategies mediated by recombinant AAV vectors that impede retinal degeneration in pre-clinical models of acquired and inherited vision loss. We demonstrate distinct neuroprotective effects upon retinal ganglion cell survival and function in experimental optic neuritis following AAV-mediated gene augmentation. Gene transfer of the antioxidant transcription factor, NRF2, improves RGC survival while overexpression of the pro-survival and anti- inflammatory protein, SIRT1, promotes preservation of visual function.
    [Show full text]
  • KAM-1325-Service Inf
    VersionVersion 2018 September ANTIBODY MICROARRAY SERVICES INFORMATION PACKAGE Toll free: 1-866-KINEXUS Facsimile: 604-323-2548 E-mail: [email protected] www.kinexus.ca TABLE OF CONTENTS Overview of Kinex™ KAM-1325 Antibody Microarray Services Page No. 1. Introduction ………………………………………………………………………………….……….... 4 2. Highly Validated Antibodies ………………………………………………...……………………….. 5 3. Quality Control Procedures ………………………………………………………………………….. 7 4. Non-Competitive Single Fluorescent Dye Combination Labelling …………………………….… 10 5. Detection of Protein Expression and Site-specific Phosphorylation with the KAM-1325 Array. 10 6. Detection of Covalent Modifications with the KAM-1325 Array ………………………………… 13 7. Detection of Protein-Protein Interactions with the KAM-1325 Array …………………..…….…. 14 8. Detection of Protein Kinase Activation and Drug Interactions with the KAM-1325 Array ..…… 15 9. KAM-1325 Antibody Microarray Reports ……………………………………………………….….. 16 10. Pricing Information ……………………………………………………………………………..……… 18 11. Follow-Up Services ………………………………………………………………………….…..……. 19 Sample Preparation 12. Quantity of Lysate ……………………………………………………………………………..……… 21 13. Lysis Buffer …………………………………………………………………………………..……… 21 14. Fractionations …………………………………………………………………………………..……… 23 15. Protein Lysate Preparation with and without Chemical Cleavage ……………………..…….…. 24 A. Preparation of Lysates from Cells with Chemical Cleavage ………………….……..…… 25 B. Preparation of Lysates from Cells without Chemical Cleavage …………….…………… 26 C. Preparation of Lysates from Tissues with Chemical
    [Show full text]
  • Characterization of Phototransduction Gene Knockouts Revealed Important Signaling Networks in the Light-Induced Retinal Degeneration
    Hindawi Publishing Corporation Journal of Biomedicine and Biotechnology Volume 2008, Article ID 327468, 7 pages doi:10.1155/2008/327468 Research Article Characterization of Phototransduction Gene Knockouts Revealed Important Signaling Networks in the Light-Induced Retinal Degeneration Jayalakshmi Krishnan,1 Gwang Lee,1, 2 Sang-Uk Han,1, 3 and Sangdun Choi1, 4 1 Department of Molecular Science and Technology, College of Natural Sciences, Ajou University , Suwon 443-749, South Korea 2 Brain Disease Research Center, Ajou University School of Medicine, Suwon 443-749, South Korea 3 Department of Surgery, Ajou University School of Medicine, Suwon 443-749, South Korea 4 Department of Biological Sciences, College of Natural Science, Ajou University, Suwon 443-749, South Korea Correspondence should be addressed to Sangdun Choi, [email protected] Received 24 August 2007; Accepted 17 December 2007 Recommended by Daniel Howard Understanding the molecular pathways mediating neuronal function in retinas can be greatly facilitated by the identification of genes regulated in the retinas of different mutants under various light conditions. We attempted to conduct a gene chip analysis − − study on the genes regulated during rhodopsin kinase (Rhok / ) and arrestin (Sag−/−) knockout and double knockouts in mice retina. Hence, mice were exposed to constant illumination of 450 lux or 6,000 lux on dilated pupils for indicated periods. The retinas were removed after the exposure and processed for microarray analysis. Double knockout was associated with immense changes in gene expression regulating a number of apoptosis inducing transcription factors. Subsequently, network analysis re- vealed that during early exposure the transcription factors, p53, c-MYC, c-FOS, JUN, and, in late phase, NF-κB, appeared to be essential for the initiation of light-induced retinal rod loss, and some other classical pro- and antipoptotic genes appeared to be significantly important as well.
    [Show full text]