Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-17T04:56:56.846Z Has data issue: false hasContentIssue false

Custom-designed proteins as novel therapeutic tools? The case of arrestins

Published online by Cambridge University Press:  23 April 2010

Vsevolod V. Gurevich*
Affiliation:
Vanderbilt University, Nashville, TN 37232, USA.
Eugenia V. Gurevich
Affiliation:
Vanderbilt University, Nashville, TN 37232, USA.
*
*Corresponding author: Vsevolod V. Gurevich, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA. E-mail: vsevolod.gurevich@vanderbilt.edu

Abstract

Multiple genetic disorders can be associated with excessive signalling by mutant G-protein-coupled receptors (GPCRs) that are either constitutively active or have lost sites where phosphorylation by GPCR kinases is necessary for desensitisation by cognate arrestins. Phosphorylation-independent arrestin1 can compensate for defects in phosphorylation of the GPCR rhodopsin in retinal rod cells, facilitating recovery, improving light responsiveness, and promoting photoreceptor survival. These proof-of-principle experiments show that, based on mechanistic understanding of the inner workings of a protein, one can modify its functional characteristics to generate custom-designed mutants that improve the balance of signalling in congenital and acquired disorders. Manipulations of arrestin elements responsible for scaffolding mitogen-activated protein kinase cascades and binding other signalling proteins involved in life-or-death decisions in the cell are likely to yield mutants that affect cell survival and proliferation in the desired direction. Although this approach is still in its infancy, targeted redesign of individual functions of many proteins offers a promise of a completely new therapeutic toolbox with huge potential.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Bockaert, J. and Pin, J.P. (1999) Molecular tinkering of G protein-coupled receptors: an evolutionary success. EMBO Journal 18, 1723-1729CrossRefGoogle ScholarPubMed
2Rompler, H. et al. (2007) G protein-coupled time travel: evolutionary aspects of GPCR research. Molecular Interventions 7, 17-25Google Scholar
3Fredriksson, R. et al. (2003) The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Molecular Pharmacology 63, 1256-1272CrossRefGoogle ScholarPubMed
4Rosenbaum, D.M., Rasmussen, S.G. and Kobilka, B.K. (2009) The structure and function of G-protein-coupled receptors. Nature 459, 356-363CrossRefGoogle ScholarPubMed
5Hofmann, K.P. et al. (2009) A G protein-coupled receptor at work: the rhodopsin model. Trends in Biochemical Sciences 34, 540-552CrossRefGoogle Scholar
6Hubbell, W.L. et al. (2003) Rhodopsin structure, dynamics, and activation: a perspective from crystallography, site-directed spin labeling, sulfhydryl reactivity, and disulfide cross-linking. Advances in Protein Chemistry 63, 243-290CrossRefGoogle ScholarPubMed
7Scheerer, P. et al. (2008) Crystal structure of opsin in its G-protein-interacting conformation. Nature 455, 497-502CrossRefGoogle ScholarPubMed
8Burns, M.E. and Pugh, E.N.J. (2009) RGS9 concentration matters in rod phototransduction. Biophysical Journal 97, 1538-1547CrossRefGoogle ScholarPubMed
9Gurevich, V.V. and Gurevich, E.V. (2006) The structural basis of arrestin-mediated regulation of G protein-coupled receptors. Pharmacology & Therapeutics 110, 465-502CrossRefGoogle ScholarPubMed
10Krupnick, J.G., Gurevich, V.V. and Benovic, J.L. (1997) Mechanism of quenching of phototransduction: binding competition between arrestin and transducin for phosphorhodopsin. Journal of Biological Chemistry 272, 18125-18131Google Scholar
11Goodman, O.B. et al. (1996) Beta-arrestin acts as a clathrin adaptor in endocytosis of the beta2-adrenergic receptor. Nature 383, 447-450CrossRefGoogle ScholarPubMed
12Laporte, S.A. et al. (2000) The interaction of beta-arrestin with the AP-2 adaptor is required for the clustering of beta 2-adrenergic receptor into clathrin-coated pits. Journal of Biological Chemistry 275, 23120-23126Google Scholar
13DeWire, S.M. et al. (2007) Beta-arrestins and cell signaling. Annual Review of Physiology 69, 483-510Google Scholar
14Kendall, R.T. and Luttrell, L.M. (2009) Diversity in arrestin function. Cellular and Molecular Life Sciences 66, 2953-2973CrossRefGoogle ScholarPubMed
15Gurevich, V.V. and Gurevich, E.V. (2008) Rich tapestry of G protein-coupled receptor signaling and regulatory mechanisms. Molecular Pharmacology 74, 312-316Google Scholar
16Tobin, A.B., Butcher, A.J. and Kong, K.C. (2008) Location, location, location…site-specific GPCR phosphorylation offers a mechanism for cell-type-specific signalling. Trends in Pharmacological Sciences 29, 413-420Google Scholar
17Kobilka, B.K. and Deupi, X. (2007) Conformational complexity of G-protein-coupled receptors. Trends in Pharmacological Sciences 28, 397-406CrossRefGoogle ScholarPubMed
18Kenakin, T. (2007) Functional selectivity through protean and biased agonism: who steers the ship? Molecular Pharmacology 72, 1393-1401Google Scholar
19Drake, M.T. et al. (2008) beta-arrestin-biased agonism at the beta2-adrenergic receptor. Journal of Biological Chemistry 283, 5669-5676Google Scholar
20Schöneberg, T. et al. (2004) Mutant G-protein-coupled receptors as a cause of human diseases. Pharmacology & Therapeutics 104, 173-206CrossRefGoogle ScholarPubMed
21Barak, L.S. et al. (2001) Constitutive arrestin-mediated desensitization of a human vasopressin receptor mutant associated with nephrogenic diabetes insipidus. Proceedings of the National Academy of Sciences of the United States of America 98, 93-98CrossRefGoogle ScholarPubMed
22Rim, J. and Oprian, D.D. (1995) Constitutive activation of opsin: interaction of mutants with rhodopsin kinase and arrestin. Biochemistry 34, 11938-11945Google Scholar
23Rao, V.R., Cohen, G.B. and Oprian, D.D. (1994) Rhodopsin mutation G90D and a molecular mechanism for congenital night blindness. Nature 367, 639-642CrossRefGoogle Scholar
24Apfelstedt-Sylla, E. et al. (1993) Ocular findings in a family with autosomal dominant retinitis pigmentosa and a frameshift mutation altering the carboxyl terminal sequence of rhodopsin. British Journal of Ophthalmology 77, 495-501CrossRefGoogle Scholar
25Kim, R.Y. et al. (1993) Dominant retinitis pigmentosa associated with two rhodopsin gene mutations. Leu-40-Arg and an insertion disrupting the 5-splice junction of exon 5. Archives of Ophthalmology 111, 1518-1524Google Scholar
26Restagno, G. et al. (1993) A large deletion at the 3 end of the rhodopsin gene in an Italian family with a diffuse form of autosomal dominant retinitis pigmentosa. Human Molecular Genetics 2, 207-208CrossRefGoogle Scholar
27Cideciyan, A.V. et al. (1998) Null mutation in the rhodopsin kinase gene slows recovery kinetics of rod and cone phototransduction in man. Proceedings of the National Academy of Sciences of the United States of America 95, 328-333Google Scholar
28Fuchs, S. et al. (1995) A homozygous 1-base pair deletion in the arrestin gene is a frequent cause of Oguchi disease in Japanese. Nature Genetics 10, 360-362CrossRefGoogle ScholarPubMed
29Xu, J. et al. (1997) Prolonged photoresponses in transgenic mouse rods lacking arrestin. Nature 389, 505-509Google Scholar
30Chen, J. et al. (1995) Mechanisms of rhodopsin inactivation in vivo as revealed by a COOsteopathic Hospitals-terminal truncation mutant. Science 267, 374-377Google Scholar
31Chen, C.K. et al. (1999) Abnormal photoresponses and light-induced apoptosis in rods lacking rhodopsin kinase. Proceedings of the National Academy of Sciences of the United States of America 96, 3718-3722CrossRefGoogle ScholarPubMed
32Mendez, A. et al. (2000) Rapid and reproducible deactivation of rhodopsin requires multiple phosphorylation sites. Neuron 28, 153-164CrossRefGoogle ScholarPubMed
33Allen, L.F. et al. (1991) G-protein-coupled receptor genes as protooncogenes: constitutively activating mutation of the alpha 1B-adrenergic receptor enhances mitogenesis and tumorigenicity. Proceedings of the National Academy of Sciences of the United States of America 88, 11354-11358CrossRefGoogle ScholarPubMed
34Gutkind, J.S. et al. (1991) Muscarinic acetylcholine receptor subtypes as agonist-dependent oncogenes. Proceedings of the National Academy of Sciences of the United States of America 88, 4703-4707CrossRefGoogle ScholarPubMed
35Julius, D. et al. (1989) Ectopic expression of the serotonin 1c receptor and the triggering of malignant transformation. Science 244, 1057-1062Google Scholar
36Young, D. et al. (1986) Isolation and characterization of a new cellular oncogene encoding a protein with multiple potential transmembrane domains. Cell 45, 711-719CrossRefGoogle ScholarPubMed
37Young, D. et al. (1988) Characterization of the rat mas oncogene and its high-level expression in the hippocampus and cerebral cortex of rat brain. Proceedings of the National Academy of Sciences of the United States of America 85, 5339-5342Google Scholar
38Jackson, T.R. et al. (1988) The mas oncogene encodes an angiotensin receptor. Nature 335, 437-440Google Scholar
39Guigoni, C. et al. (2005) Pathogenesis of levodopa-induced dyskinesia: focus on D1 and D3 dopamine receptors. Parkinsonism & Related Disorders 11, S25-29Google Scholar
40Kuhn, H., Hall, S.W. and Wilden, U. (1984) Light-induced binding of 48-kDa protein to photoreceptor membranes is highly enhanced by phosphorylation of rhodopsin. FEBS Letters 176, 473-478CrossRefGoogle ScholarPubMed
41Pfister, C. et al. (1985) Retinal S antigen identified as the 48K protein regulating light-dependent phosphodiesterase in rods. Science 228, 891-893CrossRefGoogle ScholarPubMed
42Lohse, M.J. et al. (1990) beta-Arrestin: a protein that regulates beta-adrenergic receptor function. Science 248, 1547-1550CrossRefGoogle ScholarPubMed
43Attramadal, H. et al. (1992) Beta-arrestin2, a novel member of the arrestin/beta-arrestin gene family. Journal of Biological Chemistry 267, 17882-17890Google Scholar
44Sterne-Marr, R. et al. (1993) Polypeptide variants of beta-arrestin and arrestin3. Journal of Biological Chemistry 268, 15640-15648CrossRefGoogle ScholarPubMed
45Craft, C.M., Whitmore, D.H. and Wiechmann, A.F. (1994) Cone arrestin identified by targeting expression of a functional family. Journal of Biological Chemistry 269, 4613-4619CrossRefGoogle ScholarPubMed
46Murakami, A. et al. (1993) X-arrestin: a new retinal arrestin mapping to the X chromosome. FEBS Letters 334, 203-209Google Scholar
47Lohse, M.J. et al. (1992) Receptor-specific desensitization with purified proteins. Kinase dependence and receptor specificity of beta-arrestin and arrestin in the beta 2-adrenergic receptor and rhodopsin systems. Journal of Biological Chemistry 267, 8558-8564CrossRefGoogle ScholarPubMed
48Carman, C.V. and Benovic, J.L. (1998) G-protein-coupled receptors: turn-ons and turn-offs. Current Opinion in Neurobiology 8, 335-344CrossRefGoogle ScholarPubMed
49Gurevich, V.V. and Benovic, J.L. (1992) Cell-free expression of visual arrestin. Truncation mutagenesis identifies multiple domains involved in rhodopsin interaction. Journal of Biological Chemistry 267, 21919-21923CrossRefGoogle ScholarPubMed
50Li, J. et al. (2004) Structure of bovine rhodopsin in a trigonal crystal form. Journal of Molecular Biology 343, 1409-1438CrossRefGoogle Scholar
51Gurevich, V.V. and Benovic, J.L. (1993) Visual arrestin interaction with rhodopsin: Sequential multisite binding ensures strict selectivity towards light-activated phosphorylated rhodopsin. Journal of Biological Chemistry 268, 11628-11638Google Scholar
52Gurevich, V.V. and Gurevich, E.V. (2004) The molecular acrobatics of arrestin activation. Trends in Pharmacological Sciences 25, 59-112Google Scholar
53Gurevich, V.V. and Benovic, J.L. (1995) Visual arrestin binding to rhodopsin: diverse functional roles of positively charged residues within the phosphorylation-recignition region of arrestin. Journal of Biological Chemistry 270, 6010-6016Google Scholar
54Gurevich, V.V. and Benovic, J.L. (1997) Mechanism of phosphorylation-recognition by visual arrestin and the transition of arrestin into a high affinity binding state. Molecular Pharmacology 51, 161-169Google Scholar
55Hirsch, J.A. et al. (1999) The 2.8 A crystal structure of visual arrestin: a model for arrestin's regulation. Cell 97, 257-269CrossRefGoogle Scholar
56Han, M. et al. (2001) Crystal structure of beta-arrestin at 1.9 A: possible mechanism of receptor binding and membrane translocation. Structure 9, 869-880CrossRefGoogle ScholarPubMed
57Milano, S.K. et al. (2002) Scaffolding functions of arrestin-2 revealed by crystal structure and mutagenesis. Biochemistry 41, 3321-3328Google Scholar
58Sutton, R.B. et al. (2005) Crystal structure of cone arrestin at 2.3Å: evolution of receptor specificity. Journal of Molecular Biology 354, 1069-1080Google Scholar
59Vishnivetskiy, S.A. et al. (1999) How does arrestin respond to the phosphorylated state of rhodopsin? Journal of Biological Chemistry 274, 11451-11454Google Scholar
60Vishnivetskiy, S.A. et al. (2000) An additional phosphate-binding element in arrestin molecule. Implications for the mechanism of arrestin activation. Journal of Biological Chemistry 275, 41049-41057CrossRefGoogle ScholarPubMed
61Kovoor, A. et al. (1999) Targeted construction of phosphorylation-independent b-arrestin mutants with constitutive activity in cells. Journal of Biological Chemistry 274, 6831-6834Google Scholar
62Celver, J. et al. (2002) Conservation of the phosphate-sensitive elements in the arrestin family of proteins. Journal of Biological Chemistry 277, 9043-9048CrossRefGoogle ScholarPubMed
63Gray-Keller, M.P. et al. (1997) Arrestin with a single amino acid sustitution quenches light-activated rhodopsin in a phosphorylation-independent fashion. Biochemistry 36, 7058-7063Google Scholar
64Pan, L., Gurevich, E.V. and Gurevich, V.V. (2003) The nature of the arrestin x receptor complex determines the ultimate fate of the internalized receptor. Journal of Biological Chemistry 278, 11623-11632Google Scholar
65Song, X. et al. (2009) Enhanced arrestin facilitates recovery and protects rod photoreceptors deficient in rhodopsin phosphorylation. Current Biology 19, 700-705Google Scholar
66Gurevich, V.V. (1998) The selectivity of visual arrestin for light-activated phosphorhodopsin is controlled by multiple nonredundant mechanisms. Journal of Biological Chemistry 273, 15501-15506Google Scholar
67Hanson, S.M. et al. (2006) Differential interaction of spin-labeled arrestin with inactive and active phosphorhodopsin. Proceedings of the National Academy of Sciences of the United States of America 103, 4900-4905CrossRefGoogle ScholarPubMed
68Vishnivetskiy, S.A. et al. (2010) The role of arrestin alpha-helix I in receptor binding. Journal of Molecular Biology 395, 42-54Google Scholar
69Gurevich, V.V. et al. (1997) Agonist-receptor-arrestin, an alternative ternary complex with high agonist affinity. Journal of Biological Chemistry 272, 28849-28852CrossRefGoogle ScholarPubMed
70Krasel, C. et al. (2005) Beta-arrestin binding to the beta2-adrenergic receptor requires both receptor phosphorylation and receptor activation. Journal of Biological Chemistry 280, 9528-9535Google Scholar
71Moore, C.A., Milano, S.K. and Benovic, J.L. (2007) Regulation of receptor trafficking by GRKs and arrestins. Annual Review of Physiology 69, 451-482Google Scholar
72Gurevich, V.V. et al. (1994) Visual arrestin binding to rhodopsin: Intramolecular interaction between the basic N-terminus and acidic C-terminus of arrestin may regulate binding selectivity. Journal of Biological Chemistry 269, 8721-8727CrossRefGoogle ScholarPubMed
73Puig, J. et al. (1995) Synthetic phosphopeptide from rhodopsin sequence induces retinal arrestin binding to photoactivated unphosphorylated rhodopsin. FEBS Letters 362, 185-188CrossRefGoogle ScholarPubMed
74Celver, J. et al. (2001) Threonine 180 is requred for G protein-coupled receptor kinase 3 and b-arrestin mediated desensitization of the m-opioid receptor in Xenopus oocytes. Journal of Biological Chemistry 276, 4894-4900Google Scholar
75Macey, T.A., Lowe, J.D. and Chavkin, C. (2006) Mu opioid receptor activation of ERK1/2 is GRK3 and arrestin dependent in striatal neurons. Journal of Biological Chemistry 281, 34515-34524Google Scholar
76Gurevich, V.V. et al. (2007) How rod arrestin achieved perfection: regulation of its availability and binding selectivity. In Signal Transduction in the Retina (Fliesler, S.J. and Kisselev, O., eds), pp. 55-88, CRC Press, Boca Raton, FL, USAGoogle Scholar
77Baylor, D.A., Lamb, T.D. and Yau, K.W. (1979) Responses of retinal rods to single photons. Journal of Physiology 288, 613-634Google Scholar
78Krispel, C.M. et al. (2006) RGS expression rate-limits recovery of rod photoresponses. Neuron 51, 409-416Google Scholar
79Doan, T. et al. (2006) Multiple phosphorylation sites confer reproducibility of the rod's single-photon responses. Science 313, 530-533CrossRefGoogle ScholarPubMed
80Vishnivetskiy, S.A. et al. (2007) Regulation of arrestin binding by rhodopsin phosphorylation level. Journal of Biological Chemistry 282, 32075-32083Google Scholar
81Burns, M.E. et al. (2006) Deactivation of phosphorylated and nonphosphorylated rhodopsin by arrestin splice variants. Journal of Neuroscience 26, 1036-1044CrossRefGoogle ScholarPubMed
82Palczewski, K. et al. (1991) Phosphorylated rhodopsin and heparin induce similar conformational changes in arrestin. Journal of Biological Chemistry 266, 18649-18654Google Scholar
83Pulvermuller, A. et al. (1997) Functional differences in the interaction of arrestin and its splice variant, p44, with rhodopsin. Biochemistry 36, 9253-9260CrossRefGoogle ScholarPubMed
84Nair, K.S. et al. (2005) Light-dependent redistribution of arrestin in vertebrate rods is an energy-independent process governed by protein-protein interactions. Neuron 46, 555-567CrossRefGoogle ScholarPubMed
85Hanson, S.M. et al. (2007) Each rhodopsin molecule binds its own arrestin. Proceedings of the National Academy of Sciences of the United States of America 104, 3125-3128CrossRefGoogle ScholarPubMed
86Strissel, K.J. et al. (2006) Arrestin translocation is induced at a critical threshold of visual signaling and is superstoichiometric to bleached rhodopsin. Journal of Neuroscience 26, 1146-1153Google Scholar
87Penn, R.B. et al. (2001) Arrestin specificity for G protein-coupled receptors in human airway smooth muscle. Journal of Biological Chemistry 276, 32648-32656Google Scholar
88Gurevich, V.V. and Gurevich, E.V. (2008) GPCR monomers and oligomers: it takes all kinds. Trends in Neurosciences 31, 74-81CrossRefGoogle ScholarPubMed
89Gurevich, E.V., Benovic, J.L. and Gurevich, V.V. (2002) Arrestin2 and arrestin3 are differentially expressed in the rat brain during postnatal development. Neuroscience 109, 421-436Google Scholar
90Gurevich, E.V., Benovic, J.L. and Gurevich, V.V. (2004) Arrestin2 expression selectively increases during neural differentiation. Journal of Neurochemistry 91, 1404-1416Google Scholar
91Luo, J., Busillo, J.M. and Benovic, J.L. (2008) M3 muscarinic acetylcholine receptor-mediated signaling is regulated by distinct mechanisms. Molecular Pharmacology 74, 338-347Google Scholar
92Gurevich, V.V. et al. (1995) Arrestin interaction with G protein-coupled receptors. Direct binding studies of wild type and mutant arrestins with rhodopsin, b2-adrenergic, and m2 muscarinic cholinergic receptors. Journal of Biological Chemistry 270, 720-731Google Scholar
93Hanson, S.M. et al. (2007) Arrestin mobilizes signaling proteins to the cytoskeleton and redirects their activity. Journal of Molecular Biology 368, 375-387Google Scholar
94Hanson, S.M. and Gurevich, V.V. (2006) The differential engagement of arrestin surface charges by the various functional forms of the receptor. Journal of Biological Chemistry 281, 3458-3462Google Scholar
95Song, X. et al. (2006) Visual and both non-visual arrestins in their “inactive” conformation bind JNK3 and Mdm2 and relocalize them from the nucleus to the cytoplasm. Journal of Biological Chemistry 281, 21491-21499CrossRefGoogle ScholarPubMed
96Carter, J.M. et al. (2005) Conformational differences between arrestin2 and pre-activated mutants as revealed by hydrogen exchange mass spectrometry. Journal of Molecular Biology 351, 865-878Google Scholar
97Barak, L.S. et al. (1997) A beta-arrestin/green fluorescent protein biosensor for detecting G protein-coupled receptor activation. Journal of Biological Chemistry 272, 27497-27500Google Scholar
98Miller, A.M. and Dean, D.A. (2009) Tissue-specific and transcription factor-mediated nuclear entry of DNA. Advanced Drug Delivery Reviews 61, 603-613Google Scholar
99Beck, C. et al. (2004) Tissue-specific targeting for cardiovascular gene transfer. Potential vectors and future challenges. Current Gene Therapy 4, 457-467CrossRefGoogle ScholarPubMed
100Sadeghi, H. and Hitt, M.M. (2005) Transcriptionally targeted adenovirus vectors. Current Gene Therapy 5, 411-427Google Scholar
101Lundberg, C. et al. (2008) Applications of lentiviral vectors for biology and gene therapy of neurological disorders. Current Gene Therapy 8, 461-473Google Scholar
102Baum, C. et al. (2006) Retrovirus vectors: toward the plentivirus? Molecular Therapy 13, 1050-1063CrossRefGoogle ScholarPubMed
103Ohguro, H. et al. (1994) Topographic study of arrestin using differential chemical modifications and hydrogen/deuterium exchange. Protein Science, 2428-2434Google Scholar
104Pulvermuller, A. et al. (2000) Interactions of metarhodopsin II. Arrestin peptides compete with arrestin and transducin. Journal of Biological Chemistry 275, 37679-37685Google Scholar
105Dinculescu, A. et al. (2002) Insertional mutagenesis and immunochemical analysis of visual arrestin interaction with rhodopsin. Journal of Biological Chemistry 277, 11703-11708Google Scholar
106Vishnivetskiy, S.A. et al. (2004) Mapping the arrestin-receptor interface: structural elements responsible for receptor specificity of arrestin proteins. Journal of Biological Chemistry 279, 1262-1268Google Scholar
107Gurevich, V.V. et al. (1993) Binding of wild type and chimeric arrestins to the m2 muscarinic cholinergic receptor. Journal of Biological Chemistry 268, 16879-16882Google Scholar
108Gurevich, E.V. and Gurevich, V.V. (2006) Arrestins are ubiquitous regulators of cellular signaling pathways. Genome Biology 7, 236Google Scholar
109Oakley, R.H. et al. (2000) Differential affinities of visual arrestin, barrestin1, and barrestin2 for G protein-coupled receptors delineate two major classes of receptors. Journal of Biological Chemistry 275, 17201-17210Google Scholar
110DeGraff, J.L., Gurevich, V.V. and Benovic, J.L. (2002) The third intracellular loop of alpha2-adrenergic receptors determines subtype specificity of arrestin interaction. Journal of Biological Chemistry 277, 43247-43252Google Scholar
111Macey, T.A., Gurevich, V.V. and Neve, K.A. (2004) Preferential Interaction between the dopamine D2 receptor and Arrestin2 in neostriatal neurons. Molecular Pharmacology 66, 1635-1642Google Scholar
112Dale, L.B. et al. (2001) Agonist-stimulated and tonic internalization of metabotropic glutamate receptor 1A in human embryonic kidney 293 cells: agonist-stimulated endocytosis is beta-arrestin1 isoform-specific. Molecular Pharmacology 60, 1243-1253Google Scholar
113Mundell, S.J. et al. (2000) Arrestin isoforms dictate differential kinetics of A2B adenosine receptor trafficking. Biochemistry 39, 12828-12836Google Scholar
114Ahn, S. et al. (2004) Reciprocal regulation of angiotensin receptor-activated extracellular signal-regulated kinases by beta-arrestins 1 and 2. Journal of Biological Chemistry 279, 7807-7811Google Scholar
115Gurevich, V.V. and Gurevich, E.V. (2003) The new face of active receptor bound arrestin attracts new partners. Structure 11, 1037-1042CrossRefGoogle ScholarPubMed
116Song, X., Gurevich, E.V. and Gurevich, V.V. (2007) Cone arrestin binding to JNK3 and Mdm2: conformational preference and localization of interaction sites. Journal of Neurochemistry 103, 1053-1062Google Scholar
117Song, X. et al. (2009) How does arrestin assemble MAP kinases into a signaling complex? Journal of Biological Chemistry 284, 685-695Google Scholar
118Hanson, S.M. et al. (2006) Visual arrestin binding to microtubules involves a distinct conformational change. Journal of Biological Chemistry 281, 9765-9772Google Scholar
119Wu, N. et al. (2006) Arrestin binding to calmodulin: a direct interaction between two ubiquitous signaling proteins. Journal of Molecular Biology 364, 955-963CrossRefGoogle ScholarPubMed
120Meng, D. et al. (2009) MEK1 binds directly to betaarrestin1, influencing both its phosphorylation by ERK and the timing of its isoprenaline-stimulated internalization. Journal of Biological Chemistry 284, 11425-11435Google Scholar
121Gurevich, V.V., Gurevich, E.V. and Cleghorn, W.M. (2008) Arrestins as multi-functional signaling adaptors. Handbook of Experimental Pharmacology 186, 15-37Google Scholar
122Kim, Y.M. and Benovic, J.L. (2002) Differential roles of arrestin-2 interaction with clathrin and adaptor protein 2 in G protein-coupled receptor trafficking. Journal of Biological Chemistry 277, 30760-30768Google Scholar
123Kang, D.S. et al. (2009) Structure of an arrestin2/clathrin complex reveals a novel clathrin binding domain that modulates receptor trafficking. Journal of Biological Chemistry 284, 29860-29872Google Scholar
124Baillie, G.S. et al. (2007) Mapping binding sites for the PDE4D5 cAMP-specific phosphodiesterase to the N- and C-domains of beta-arrestin using spot-immobilized peptide arrays. Biochemical Journal 404, 71-80Google Scholar
125Luttrell, L.M. et al. (2001) Activation and targeting of extracellular signal-regulated kinases by beta-arrestin scaffolds. Proceedings of the National Academy of Sciences of the United States of America 98, 2449-2454Google Scholar
126McDonald, P.H. et al. (2000) Beta-arrestin 2: a receptor-regulated MAPK scaffold for the activation of JNK3. Science 290, 1574-1577Google Scholar
127Miller, W.E. et al. (2001) Identification of a motif in the carboxyl terminus of beta -arrestin2 responsible for activation of JNK3. Journal of Biological Chemistry 276, 27770-27777Google Scholar
128Scott, M.G. et al. (2002) Differential nucleocytoplasmic shuttling of beta-arrestins. Characterization of a leucine-rich nuclear export signal in beta-arrestin2. Journal of Biological Chemistry 277, 37693-37701Google Scholar
129Wang, P. et al. (2003) Subcellular localization of beta-arrestins is determined by their intact N domain and the nuclear export signal at the C terminus. Journal of Biological Chemistry 278, 11648-11653Google Scholar
130Miller, W.E. and Lefkowitz, R.J. (2001) Expanding roles for beta-arrestins as scaffolds and adapters in GPCR signaling and trafficking. Current Opinion in Cell Biology 13, 139-145Google Scholar
131Li, X. et al. (2009) A scanning peptide array approach uncovers association sites within the JNK/betaarrestin signalling complex. FEBS Letters 583, 3310-3316Google Scholar
132Schleicher, A., Kuhn, H. and Hofmann, K.P. (1989) Kinetics, binding constant, and activation energy of the 48-kDa protein-rhodopsin complex by extra-metarhodopsin II. Biochemistry 28, 1770-1775Google Scholar
133Xiao, K. et al. (2004) Activation-dependent conformational changes in {beta}-arrestin 2. Journal of Biological Chemistry 279, 55744-55753CrossRefGoogle ScholarPubMed
134Nobles, K.N. et al. (2007) The active conformation of beta-arrestin1: direct evidence for the phosphate sensor in the N-domain and conformational differences in the active states of beta-arrestins1 and -2. Journal of Biological Chemistry 282, 21370-21381Google Scholar
135Vishnivetskiy, S.A. et al. (2002) Transition of arrestin into the active receptor-binding state requires an extended interdomain hinge. Journal of Biological Chemistry 277, 43961-43967Google Scholar
136Bhandari, D. et al. (2007) Arrestin-2 interacts with the ubiquitin-protein isopeptide ligase atrophin-interacting protein 4 and mediates endosomal sorting of the chemokine receptor CXCR4. Journal of Biological Chemistry 282, 36971-36979Google Scholar
137Gaidarov, I. and Keen, J.H. (1999) Phosphoinositide-AP-2 interactions required for targeting to plasma membrane clathrin-coated pits. Journal of Cell Biology 146, 755-764CrossRefGoogle ScholarPubMed
138Krupnick, J.G. et al. (1997) Modulation of the arrestin-clathrin interaction in cells. Characterization of beta-arrestin dominant-negative mutants. Journal of Biological Chemistry. 272, 32507-32512Google Scholar
139Perry, S.J. et al. (2002) Targeting of cyclic AMP degradation to beta 2-adrenergic receptors by beta-arrestins. Science 298, 834-836CrossRefGoogle ScholarPubMed
140Li, Y. et al. (2009) Regulation of amygdalar PKA by beta-arrestin-2/phosphodiesterase-4 complex is critical for fear conditioning. Proceedings of the National Academy of Sciences of the United States of America 106, 21918-21923Google Scholar
141Lin, F.T. et al. (1999) Feedback regulation of beta-arrestin1 function by extracellular signal-regulated kinases. Journal of Biological Chemistry 274, 15971-15974Google Scholar
142Shenoy, S.K. et al. (2001) Regulation of receptor fate by ubiquitination of activated beta 2-adrenergic receptor and beta-arrestin. Science 294, 1307-1313Google Scholar
143Shenoy, S.K. et al. (2008) Nedd4 mediates agonist-dependent ubiquitination, lysosomal targeting, and degradation of the beta2-adrenergic receptor. Journal of Biological Chemistry 283, 22166-22176Google Scholar
144Berthouze, M. et al. (2009) The deubiquitinases USP33 and USP20 coordinate beta2 adrenergic receptor recycling and resensitization. EMBO Journal 28, 1684-1696Google Scholar
145Shenoy, S.K. and Lefkowitz, R.J. (2005) Receptor-specific ubiquitination of beta-arrestin directs assembly and targeting of seven-transmembrane receptor signalosomes. Journal of Biological Chemistry 280, 15315-15324Google Scholar
146Mosser, V.A. et al. (2008) Differential role of beta-arrestin ubiquitination in agonist-promoted down-regulation of M1 vs M2 muscarinic acetylcholine receptors. Journal of Molecular Signaling 3, 20Google Scholar
147Pals-Rylaarsdam, R. et al. (1997) Internalization of the m2 muscarinic acetylcholine receptor: arrestin-independent and -dependent pathways. Journal of Biological Chemistry 272, 23682-23689Google Scholar
148Lee, K.B. et al. (2000) Arrestin binding to the M2 muscarinic acetylcholine receptor is precluded by an inhibitory element in the third intracellular loop of the receptor. Journal of Biological Chemistry 275, 9284-9289Google Scholar
149Shenoy, S.K. et al. (2007) Ubiquitination of beta-arrestin links seven-transmembrane receptor endocytosis and ERK activation. Journal of Biological Chemistry 282, 29549-29546CrossRefGoogle ScholarPubMed
150Schubert, C. et al. (1999) Visual arrestin activity may be regulated by self-association. Journal of Biological Chemistry 274, 21186-21190Google Scholar
151Imamoto, Y. et al. (2003) Concentration-dependent tetramerization of bovine visual arrestin. Biophysical Journal 85, 1186-1195CrossRefGoogle ScholarPubMed
152Hanson, S.M. et al. (2007) Structure and function of the visual arrestin oligomer. EMBO Journal 26, 1726-1736Google Scholar
153Granzin, J. et al. (1998) X-ray crystal structure of arrestin from bovine rod outer segments. Nature 391, 918-921Google Scholar
154Hanson, S.M. et al. (2008) A model for the solution structure of the rod arrestin tetramer. Structure 16, 924-934Google Scholar
155Hanson, S.M. et al. (2008) Opposing effects of inositol hexakisphosphate on rod arrestin and arrestin2 self-association. Biochemistry 47, 1070-1075Google Scholar
156Milano, S.K. et al. (2006) Nonvisual arrestin oligomerization and cellular localization are regulated by inositol hexakisphosphate binding. Journal of Biological Chemistry 281, 9812-9823Google Scholar
157Storez, H. et al. (2005) Homo- and hetero-oligomerization of beta-arrestins in living cells. Journal of Biological Chemistry 280, 40210-40215CrossRefGoogle ScholarPubMed
158Kang, J. et al. (2005) A nuclear function of beta-arrestin1 in GPCR signaling: regulation of histone acetylation and gene transcription. Cell 123, 833-847Google Scholar
159Boularan, C. et al. (2007) beta-arrestin 2 oligomerization controls the Mdm2-dependent inhibition of p53. Proceedings of the National Academy of Sciences of the United States of America 104, 18061-18066Google Scholar
160Xu, T.R. et al. (2008) Mutations of beta-arrestin 2 that limit self-association also interfere with interactions with the beta2-adrenoceptor and the ERK1/2 MAPKs: implications for beta2-adrenoceptor signalling via the ERK1/2 MAPKs. Biochemical Journal 413, 51-60Google Scholar
161DeFea, K.A. (2008) Beta-arrestin multimers: does a crowd help or hinder function? Biochemical Journal 413, e1-3Google Scholar
162Conn, P.J., Christopoulos, A. and Lindsley, C.W. (2009) Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nature Reviews Drug Discovery 8, 41-54Google Scholar
163Kenakin, T.P. (2009) 7TM receptor allostery: putting numbers to shapeshifting proteins. Trends in Pharmacological Sciences 30, 460-469CrossRefGoogle ScholarPubMed
Song, X. et al. (2009) Enhanced arrestin facilitates recovery and protects rods lacking rhodopsin phosphorylation. Current Biology 19, 700-705CrossRefGoogle ScholarPubMed
Tobin, A.B., Butcher, A.J. and Kong, K.C. (2008) Location, location, location…site-specific GPCR phosphorylation offers a mechanism for cell-type-specific signaling. Trends in Pharmacological Sciences 29, 413-420Google Scholar
DeWire, S.M. et al. (2007) Beta-arrestins and cell signaling. Annual Revew of Physiology 69, 483-510Google Scholar
Gurevich, V.V. and Gurevich, E.V. (2006) The structural basis of arrestin-mediated regulation of G-protein-coupled receptors. Pharmacology & Therapeutics 110, 465-502Google Scholar
Song, X. et al. (2009) Enhanced arrestin facilitates recovery and protects rods lacking rhodopsin phosphorylation. Current Biology 19, 700-705CrossRefGoogle ScholarPubMed
Tobin, A.B., Butcher, A.J. and Kong, K.C. (2008) Location, location, location…site-specific GPCR phosphorylation offers a mechanism for cell-type-specific signaling. Trends in Pharmacological Sciences 29, 413-420Google Scholar
DeWire, S.M. et al. (2007) Beta-arrestins and cell signaling. Annual Revew of Physiology 69, 483-510Google Scholar
Gurevich, V.V. and Gurevich, E.V. (2006) The structural basis of arrestin-mediated regulation of G-protein-coupled receptors. Pharmacology & Therapeutics 110, 465-502Google Scholar