1
|
Gurevich VV. Arrestins: A Small Family of Multi-Functional Proteins. Int J Mol Sci 2024; 25:6284. [PMID: 38892473 PMCID: PMC11173308 DOI: 10.3390/ijms25116284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The first member of the arrestin family, visual arrestin-1, was discovered in the late 1970s. Later, the other three mammalian subtypes were identified and cloned. The first described function was regulation of G protein-coupled receptor (GPCR) signaling: arrestins bind active phosphorylated GPCRs, blocking their coupling to G proteins. It was later discovered that receptor-bound and free arrestins interact with numerous proteins, regulating GPCR trafficking and various signaling pathways, including those that determine cell fate. Arrestins have no enzymatic activity; they function by organizing multi-protein complexes and localizing their interaction partners to particular cellular compartments. Today we understand the molecular mechanism of arrestin interactions with GPCRs better than the mechanisms underlying other functions. However, even limited knowledge enabled the construction of signaling-biased arrestin mutants and extraction of biologically active monofunctional peptides from these multifunctional proteins. Manipulation of cellular signaling with arrestin-based tools has research and likely therapeutic potential: re-engineered proteins and their parts can produce effects that conventional small-molecule drugs cannot.
Collapse
|
2
|
Eissa AM, Hassanin MH, Ibrahim IAAEH. Hepatic β-arrestins: potential roles in liver health and disease. Mol Biol Rep 2023; 50:10399-10407. [PMID: 37843713 PMCID: PMC10676313 DOI: 10.1007/s11033-023-08898-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023]
Abstract
Β-arrestins are intracellular scaffolding proteins that have multifaceted roles in different types of disorders. In this review article, we gave a summary about the discovery, characterization and classification of these proteins and their intracellular functions. Moreover, this review article focused on the hepatic expression of β-arrestins and their hepatocellular distribution and function in each liver cell type. Also, we showed that β-arrestins are key regulators of distinct types of hepatic disorders. On the other hand, we addressed some important points that have never been studied before regarding the role of β-arrestins in certain types of hepatic disorders which needs more research efforts to cover.
Collapse
Affiliation(s)
| | | | - Islam A A E H Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
3
|
Dagdeviren S, Lee RT, Wu N. Physiological and Pathophysiological Roles of Thioredoxin Interacting Protein: A Perspective on Redox Inflammation and Metabolism. Antioxid Redox Signal 2023; 38:442-460. [PMID: 35754346 PMCID: PMC9968628 DOI: 10.1089/ars.2022.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/12/2022] [Indexed: 11/12/2022]
Abstract
Significance: Thioredoxin interacting protein (TXNIP) is a member of the arrestin fold superfamily with important cellular functions, including cellular transport, mitochondrial energy generation, and protein cycling. It is the only arrestin-domain protein known to covalently bind to thioredoxin and plays roles in glucose metabolism, inflammation, apoptosis, and cancer. Recent Advances: The crystal structure of the TXNIP-thioredoxin complex provided details about this fascinating interaction. Recent studies showed that TXNIP is induced by endoplasmic reticulum (ER) stress, activates NLR family pyrin domain containing 3 (NLRP3) inflammasomes, and can regulate glucose transport into cells. The tumor suppressor role of TXNIP in various cancer types and the role of TXNIP in fructose absorption are now described. Critical Issues: The influence of TXNIP on redox state is more complex than its interaction with thioredoxin. Future Directions: It is incompletely understood which functions of TXNIP are thioredoxin-dependent. It is also unclear whether TXNIP binding can inhibit glucose transporters without endocytosis. TXNIP-regulated control of ER stress should also be investigated further. Antioxid. Redox Signal. 38, 442-460.
Collapse
Affiliation(s)
- Sezin Dagdeviren
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Ning Wu
- Van Andel Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
4
|
Szénási T, Turu G, Hunyady L. Interactions between β-arrestin proteins and the cytoskeletal system, and their relevance to neurodegenerative disorders. Front Endocrinol (Lausanne) 2023; 14:957981. [PMID: 36843600 PMCID: PMC9947276 DOI: 10.3389/fendo.2023.957981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/04/2023] [Indexed: 02/11/2023] Open
Abstract
β-arrestins, which have multiple cellular functions, were initially described as proteins that desensitize rhodopsin and other G protein-coupled receptors. The cytoskeletal system plays a role in various cellular processes, including intracellular transport, cell division, organization of organelles, and cell cycle. The interactome of β-arrestins includes the major proteins of the three main cytoskeletal systems: tubulins for microtubules, actins for the actin filaments, and vimentin for intermediate filaments. β-arrestins bind to microtubules and regulate their activity by recruiting signaling proteins and interacting with assembly proteins that regulate the actin cytoskeleton and the intermediate filaments. Altered regulation of the cytoskeletal system plays an essential role in the development of Alzheimer's, Parkinson's and other neurodegenerative diseases. Thus, β-arrestins, which interact with the cytoskeleton, were implicated in the pathogenesis progression of these diseases and are potential targets for the treatment of neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Tibor Szénási
- Institute of Enzymology, Research Center for Natural Sciences, Centre of Excellence of the Hungarian Academy of Sciences, Budapest, Hungary
| | - Gábor Turu
- Institute of Enzymology, Research Center for Natural Sciences, Centre of Excellence of the Hungarian Academy of Sciences, Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - László Hunyady
- Institute of Enzymology, Research Center for Natural Sciences, Centre of Excellence of the Hungarian Academy of Sciences, Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- *Correspondence: László Hunyady,
| |
Collapse
|
5
|
Chen H, Zhang S, Zhang X, Liu H. QR code model: a new possibility for GPCR phosphorylation recognition. Cell Commun Signal 2022; 20:23. [PMID: 35236365 PMCID: PMC8889771 DOI: 10.1186/s12964-022-00832-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/23/2022] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane proteins in the human body and are responsible for accurately transmitting extracellular information to cells. Arrestin is an important member of the GPCR signaling pathway. The main function of arrestin is to assist receptor desensitization, endocytosis and signal transduction. In these processes, the recognition and binding of arrestin to phosphorylated GPCRs is fundamental. However, the mechanism by which arrestin recognizes phosphorylated GPCRs is not fully understood. The GPCR phosphorylation recognition "bar code model" and "flute" model describe the basic process of receptor phosphorylation recognition in terms of receptor phosphorylation sites, arrestin structural changes and downstream signaling. These two models suggest that GPCR phosphorylation recognition is a process involving multiple factors. This process can be described by a "QR code" model in which ligands, GPCRs, G protein-coupled receptor kinase, arrestin, and phosphorylation sites work together to determine the biological functions of phosphorylated receptors. Video Abstract.
Collapse
Affiliation(s)
- Hao Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
6
|
Yuan D, Yan T, Luo S, Huang J, Tan J, Zhang J, Zhang VW, Lan Y, Hu T, Guo J, Huang M, Zeng D. Identification and Functional Characterization of a Novel Nonsense Variant in ARR3 in a Southern Chinese Family With High Myopia. Front Genet 2021; 12:765503. [PMID: 34966409 PMCID: PMC8710690 DOI: 10.3389/fgene.2021.765503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/10/2021] [Indexed: 01/28/2023] Open
Abstract
ARR3 has been associated with X-linked, female-limited, high myopia. However, using exome sequencing (ES), we identified the first high myopia case with hemizygous ARR3-related mutation in a male patient in a Southern Chinese family. This novel truncated mutation (ARR3: c.569C>G, p.S190*) co-segregated with the disease phenotype in affected family members and demonstrated that high myopia caused by ARR3 is not X-linked, female-limited, where a complicated X-linked inheritance pattern may exist. Thus, our case expanded the variant spectrum in ARR3 and provided additional information for genetic counseling, prenatal testing, and diagnosis. Moreover, we characterized the nonsense-mediated decay of the ARR3 mutant mRNA and discussed the possible underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Dejian Yuan
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China.,Liuzhou Key Laboratory of Birth Defects Prevention and Control, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Tizhen Yan
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China.,Liuzhou Key Laboratory of Birth Defects Prevention and Control, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Shiqiang Luo
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China.,Liuzhou Key Laboratory of Birth Defects Prevention and Control, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Jun Huang
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China.,Liuzhou Key Laboratory of Birth Defects Prevention and Control, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Jianqiang Tan
- Department of Medical Genetics, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China.,Liuzhou Key Laboratory of Birth Defects Prevention and Control, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Jianping Zhang
- Department of Ophthalmology, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Victor Wei Zhang
- AmCare Genomics Laboratory, Guangzhou, China.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Yueyuan Lan
- Department of Ophthalmology, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Taobo Hu
- Center of Breast Diseases, Peking University People's Hospital, Beijing, China
| | - Jing Guo
- Reproductive Medical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingwei Huang
- Aegicare (Sheznzhen) Technology Co., Ltd., Shenzhen, China
| | - Dingyuan Zeng
- Department of Gynecology, Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, China
| |
Collapse
|
7
|
Shome A, Mugisho OO, Niederer RL, Rupenthal ID. Blocking the inflammasome: A novel approach to treat uveitis. Drug Discov Today 2021; 26:2839-2857. [PMID: 34229084 DOI: 10.1016/j.drudis.2021.06.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022]
Abstract
Uveitis is a complex ocular inflammatory disease often accompanied by bacterial or viral infections (infectious uveitis) or underlying autoimmune diseases (non-infectious uveitis). Treatment of the underlying infection along with corticosteroid-mediated suppression of acute inflammation usually resolves infectious uveitis. However, to develop more effective therapies for non-infectious uveitis and to better address acute inflammation in infectious disease, an improved understanding of the underlying inflammatory pathways is needed. In this review, we discuss the disease aetiology, preclinical in vitro and in vivo uveitis models, the role of inflammatory pathways, as well as current and future therapies. In particular, we highlight the involvement of the inflammasome in the development of non-infectious uveitis and how it could be a future target for effective treatment of the disease.
Collapse
Affiliation(s)
- Avik Shome
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Rachael L Niederer
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand; Auckland District Health Board, Auckland, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
8
|
Martin GJ, Lower SE, Suvorov A, Bybee SM. Molecular Evolution of Phototransduction Pathway Genes in Nocturnal and Diurnal Fireflies (Coleoptera: Lampyridae). INSECTS 2021; 12:insects12060561. [PMID: 34207188 PMCID: PMC8235688 DOI: 10.3390/insects12060561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/04/2021] [Accepted: 06/06/2021] [Indexed: 11/16/2022]
Abstract
Most organisms are dependent on sensory cues from their environment for survival and reproduction. Fireflies (Coleoptera: Lampyridae) represent an ideal system for studying sensory niche adaptation due to many species relying on bioluminescent communication; as well as a diversity of ecologies. Here; using transcriptomics; we examine the phototransduction pathway in this non-model organism; and provide some of the first evidence for positive selection in the phototransduction pathway beyond opsins in beetles. Evidence for gene duplications within Lampyridae are found in inactivation no afterpotential C and inactivation no afterpotential D. We also find strong support for positive selection in arrestin-2; inactivation no afterpotential D; and transient receptor potential-like; with weak support for positive selection in guanine nucleotide-binding protein G(q) subunit alpha and neither inactivation nor afterpotential C. Taken with other recent work in flies; butterflies; and moths; this represents an exciting new avenue of study as we seek to further understand diversification and constraint on the phototransduction pathway in light of organism ecology.
Collapse
Affiliation(s)
- Gavin J. Martin
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (A.S.); (S.M.B.)
- Monte L. Bean Museum, Brigham Young University, Provo, UT 84602, USA
- Correspondence:
| | - Sarah E. Lower
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA;
| | - Anton Suvorov
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (A.S.); (S.M.B.)
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Seth M. Bybee
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (A.S.); (S.M.B.)
- Monte L. Bean Museum, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
9
|
Wanka L, Behr V, Beck-Sickinger AG. Arrestin-dependent internalization of rhodopsin-like G protein-coupled receptors. Biol Chem 2021; 403:133-149. [PMID: 34036761 DOI: 10.1515/hsz-2021-0128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/28/2021] [Indexed: 01/14/2023]
Abstract
The internalization of G protein-coupled receptors (GPCRs) is an important mechanism regulating the signal strength and limiting the opportunity of receptor activation. Based on the importance of GPCRs, the detailed knowledge about the regulation of signal transduction is crucial. Here, current knowledge about the agonist-induced, arrestin-dependent internalization process of rhodopsin-like GPCRs is reviewed. Arrestins are conserved molecules that act as key players within the internalization process of many GPCRs. Based on highly conserved structural characteristics within the rhodopsin-like GPCRs, the identification of arrestin interaction sites in model systems can be compared and used for the investigation of internalization processes of other receptors. The increasing understanding of this essential regulation mechanism of receptors can be used for drug development targeting rhodopsin-like GPCRs. Here, we focus on the neuropeptide Y receptor family, as these receptors transmit various physiological processes such as food intake, energy homeostasis, and regulation of emotional behavior, and are further involved in pathophysiological processes like cancer, obesity and mood disorders. Hence, this receptor family represents an interesting target for the development of novel therapeutics requiring the understanding of the regulatory mechanisms influencing receptor mediated signaling.
Collapse
Affiliation(s)
- Lizzy Wanka
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Victoria Behr
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| |
Collapse
|
10
|
Benovic JL. Historical Perspective of the G Protein-Coupled Receptor Kinase Family. Cells 2021; 10:555. [PMID: 33806476 PMCID: PMC7999923 DOI: 10.3390/cells10030555] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
Agonist activation of G protein-coupled receptors promotes sequential interaction of the receptor with heterotrimeric G proteins, G protein-coupled receptor kinases (GRKs), and arrestins. GRKs play a central role in mediating the switch from G protein to arrestin interaction and thereby control processes such as receptor desensitization and trafficking and arrestin-mediated signaling. In this review, I provide a historical perspective on some of the early studies that identified the family of GRKs with a primary focus on the non-visual GRKs. These studies included identification, purification, and cloning of the β-adrenergic receptor kinase in the mid- to late-1980s and subsequent cloning and characterization of additional members of the GRK family. This helped to lay the groundwork for ensuing work focused on understanding the structure and function of these important enzymes.
Collapse
Affiliation(s)
- Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
11
|
Harris SS, Urs NM. Targeting β-Arrestins in the Treatment of Psychiatric and Neurological Disorders. CNS Drugs 2021; 35:253-264. [PMID: 33651366 DOI: 10.1007/s40263-021-00796-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
Therapies for psychiatric and neurological disorders have been in the development and refinement process for the past 5 decades. Yet, most of these therapies lack optimal therapeutic efficacy and have multiple debilitating side effects. Recent advances in understanding the pathophysiological processes of psychiatric and neurological disorders have revealed an important role for β-arrestins, which are important regulators of G-protein-coupled receptor (GPCR) function, including desensitization and intracellular signaling. These findings have pushed β-arrestins to the forefront as potential therapeutic targets. Here, we highlight current knowledge on β-arrestin functions in certain psychiatric and neurological disorders (schizophrenia, Parkinson's disease, and substance abuse disorders), and how this has been leveraged to develop new therapeutic strategies. Furthermore, we discuss the obstacles impacting the field of β-arrestin-based therapeutic development and future approaches that might help advance strategies to develop optimal β-arrestin-based therapies.
Collapse
Affiliation(s)
- Sharonda S Harris
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Dr, ARB-R5-140, Gainesville, FL, 32610, USA
| | - Nikhil M Urs
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Dr, ARB-R5-140, Gainesville, FL, 32610, USA.
| |
Collapse
|
12
|
Smith JS, Pack TF. Noncanonical interactions of G proteins and β‐arrestins: from competitors to companions. FEBS J 2021; 288:2550-2561. [DOI: 10.1111/febs.15749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 02/02/2021] [Indexed: 12/30/2022]
Affiliation(s)
- Jeffrey S. Smith
- Department of Dermatology Massachusetts General Hospital Boston MA USA
- Department of Dermatology Brigham and Women's Hospital Boston MA USA
- Department of Dermatology Beth Israel Deaconess Medical Center Boston MA USA
- Dermatology Program Boston Children's Hospital Boston MA USA
- Harvard Medical School Boston MA USA
| | | |
Collapse
|
13
|
Dissecting the structural features of β-arrestins as multifunctional proteins. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140603. [PMID: 33421644 DOI: 10.1016/j.bbapap.2021.140603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023]
Abstract
β-arrestins bind active G protein-coupled receptors (GPCRs) and play a crucial role in receptor desensitization and internalization. The classical paradigm of arrestin function has been expanded with the identification of many non-receptor-binding partners, which indicated the multifunctional role of β-arrestins in cellular functions. To elucidate the molecular mechanism of β-arrestin-mediated signaling, the structural features of β-arrestins were investigated using X-ray crystallography and cryogenic electron microscopy (cryo-EM). However, the intrinsic conformational flexibility of β-arrestins hampers the elucidation of structural interactions between β-arrestins and their binding partners using conventional structure determination tools. Therefore, structural information obtained using complementary structure analysis techniques would be necessary in combination with X-ray crystallography and cryo-EM data. In this review, we describe how β-arrestins interact with their binding partners from a structural point of view, as elucidated by both traditional methods (X-ray crystallography and cryo-EM) and complementary structure analysis techniques.
Collapse
|
14
|
Pinealocytes can not transport neurotropic viruses. Pinealo-to-retinal connection in prepubertal rats originates from pineal neurons: Light and electron microscopic immunohistochemical studies. Neurosci Lett 2020; 744:135517. [PMID: 33246028 DOI: 10.1016/j.neulet.2020.135517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/13/2020] [Accepted: 11/18/2020] [Indexed: 11/20/2022]
Abstract
It is well established that the adult mammalian pineal body (PB), with the exception of rodents, contains nerve cell bodies. Based on our previous results we have proposed that there is a pinealo-to-retinal neuronal connection in adult hamsters and in prebubertal rats. By the time the animals reached puberty, labeled cells in the PB were not observed in rats. In the present experiment, we provide light and electron microscopic immunohistochemical evidence that the labeled cells in the PB of prepubertal rats are neurons. Pinealocytes cannot transport neurotropic viruses. Virus labeled cells do not show S-antigen immunoreactivity typical for pinealocytes of six-day-old rats. Electron microscopic investigation confirmed the neuronal nature of virus labeled cells. These neurons, similarly to that of hamsters, also establish pinealo-to-retinal connections in prepubertal rats.
Collapse
|
15
|
Charles J, Castellino FJ, Ploplis VA. Past and Present Behçet's Disease Animal Models. Curr Drug Targets 2020; 21:1652-1663. [PMID: 32682369 PMCID: PMC7746599 DOI: 10.2174/1389450121666200719010425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Behçet's disease (BD) is presumably an autoinflammatory disease of unknown etiology for which several animal models have been described over the years. Agents and methods used for the development of these models have ranged from the herpes simplex type one virus (hsv-1) pathogen to the use of transgenic mice. Other models have also been used to investigate a possible autoimmune component. Each model possesses its own unique set of benefits and shortcomings, with no one model fully being able to recapitulate the disease phenotype. Here, we review the proposed models and provide commentary on their effectiveness and usefulness in studying the disease.
Collapse
Affiliation(s)
- Jermilia Charles
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Francis J. Castellino
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Victoria A. Ploplis
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
16
|
Haider RS, Wilhelm F, Rizk A, Mutt E, Deupi X, Peterhans C, Mühle J, Berger P, Schertler GFX, Standfuss J, Ostermaier MK. Arrestin-1 engineering facilitates complex stabilization with native rhodopsin. Sci Rep 2019; 9:439. [PMID: 30679635 PMCID: PMC6346018 DOI: 10.1038/s41598-018-36881-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 11/23/2018] [Indexed: 01/14/2023] Open
Abstract
Arrestin-1 desensitizes the activated and phosphorylated photoreceptor rhodopsin by forming transient rhodopsin−arrestin-1 complexes that eventually decay to opsin, retinal and arrestin-1. Via a multi-dimensional screening setup, we identified and combined arrestin-1 mutants that form lasting complexes with light-activated and phosphorylated rhodopsin in harsh conditions, such as high ionic salt concentration. Two quadruple mutants, D303A + T304A + E341A + F375A and R171A + T304A + E341A + F375A share similar heterologous expression and thermo-stability levels with wild type (WT) arrestin-1, but are able to stabilize complexes with rhodopsin with more than seven times higher half-maximal inhibitory concentration (IC50) values for NaCl compared to the WT arrestin-1 protein. These quadruple mutants are also characterized by higher binding affinities to phosphorylated rhodopsin, light-activated rhodopsin and phosphorylated opsin, as compared with WT arrestin-1. Furthermore, the assessed arrestin-1 mutants are still specifically associating with phosphorylated or light-activated receptor states only, while binding to the inactive ground state of the receptor is not significantly altered. Additionally, we propose a novel functionality for R171 in stabilizing the inactive arrestin-1 conformation as well as the rhodopsin–arrestin-1 complex. The achieved stabilization of the active rhodopsin–arrestin-1 complex might be of great interest for future structure determination, antibody development studies as well as drug-screening efforts targeting G protein-coupled receptors (GPCRs).
Collapse
Affiliation(s)
- Raphael S Haider
- InterAx Biotech AG, PARK InnovAARE, Villigen, 5234, Switzerland.,Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland.,Institute of Molecular Cell Biology, Jena, 07745, Germany
| | - Florian Wilhelm
- InterAx Biotech AG, PARK InnovAARE, Villigen, 5234, Switzerland
| | - Aurélien Rizk
- InterAx Biotech AG, PARK InnovAARE, Villigen, 5234, Switzerland
| | - Eshita Mutt
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Xavier Deupi
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Christian Peterhans
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Jonas Mühle
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Philipp Berger
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Gebhard F X Schertler
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland.,ETH Zurich, Zurich, 8093, Switzerland
| | - Jörg Standfuss
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | | |
Collapse
|
17
|
Abstract
Arrestins have now been implicated in the actions of virtually every G protein-coupled receptor (GPCR) for which they have been examined. Originally discovered for their role in the turnoff of visual perception, their newly discovered pleotropic functions in the cellular and physiological actions of GPCRs not only illuminate new mechanisms of signal transduction but also offer new avenues for therapeutic utility. Below, in this introductory chapter, we provide a short historical description and synopsis of how arrestins conceptually became associated with the function of GPCRs.
Collapse
Affiliation(s)
- Marc G. Caron
- Department of Cell Biology Duke University Medical Center Durham NC USA,Department of Neurobiology Duke University Medical Center Durham NC USA,Department of Medicine Duke University Medical Center Durham NC USA
| | - Lawrence S. Barak
- Department of Cell Biology Duke University Medical Center Durham NC USA
| |
Collapse
|
18
|
Laporte SA, Scott MGH. β-Arrestins: Multitask Scaffolds Orchestrating the Where and When in Cell Signalling. Methods Mol Biol 2019; 1957:9-55. [PMID: 30919345 DOI: 10.1007/978-1-4939-9158-7_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The β-arrestins (β-arrs) were initially appreciated for the roles they play in the desensitization and endocytosis of G protein-coupled receptors (GPCRs). They are now also known to act as multifunctional adaptor proteins binding many non-receptor protein partners to control multiple signalling pathways. β-arrs therefore act as key regulatory hubs at the crossroads of external cell inputs and functional outputs in cellular processes ranging from gene transcription to cell growth, survival, cytoskeletal regulation, polarity, and migration. An increasing number of studies have also highlighted the scaffolding roles β-arrs play in vivo in both physiological and pathological conditions, which opens up therapeutic avenues to explore. In this introductory review chapter, we discuss the functional roles that β-arrs exert to control GPCR function, their dynamic scaffolding roles and how this impacts signal transduction events, compartmentalization of β-arrs, how β-arrs are regulated themselves, and how the combination of these events culminates in cellular regulation.
Collapse
Affiliation(s)
- Stéphane A Laporte
- Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montreal, QC, Canada. .,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada. .,RI-MUHC/Glen Site, Montréal, QC, Canada.
| | - Mark G H Scott
- Institut Cochin, INSERM U1016, Paris, France. .,CNRS, UMR 8104, Paris, France. .,Univ. Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
19
|
Gurevich VV, Gurevich EV. Arrestin mutations: Some cause diseases, others promise cure. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 161:29-45. [PMID: 30711028 PMCID: PMC6400060 DOI: 10.1016/bs.pmbts.2018.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Arrestins play a key role in homologous desensitization of G protein-coupled receptors (GPCRs) and regulate several other vital signaling pathways in cells. Considering the critical roles of these proteins in cellular signaling, surprisingly few disease-causing mutations in human arrestins were described. Most of these are loss-of-function mutations of visual arrestin-1 that cause excessive rhodopsin signaling and hence night blindness. Only one dominant arrestin-1 mutation was discovered so far. It reduces the thermal stability of the protein, which likely results in photoreceptor death via unfolded protein response. In case of the two nonvisual arrestins, only polymorphisms were described, some of which appear to be associated with neurological disorders and altered response to certain treatments. Structure-function studies revealed several ways of enhancing arrestins' ability to quench GPCR signaling. These enhanced arrestins have potential as tools for gene therapy of disorders associated with excessive signaling of mutant GPCRs.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States.
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
20
|
Komolov KE, Benovic JL. G protein-coupled receptor kinases: Past, present and future. Cell Signal 2017; 41:17-24. [PMID: 28711719 DOI: 10.1016/j.cellsig.2017.07.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 02/08/2023]
Abstract
This review is provided in recognition of the extensive contributions of Dr. Robert J. Lefkowitz to the G protein-coupled receptor (GPCR) field and to celebrate his 75th birthday. Since one of the authors trained with Bob in the 80s, we provide a history of work done in the Lefkowitz lab during the 80s that focused on dissecting the mechanisms that regulate GPCR signaling, with a particular emphasis on the GPCR kinases (GRKs). In addition, we highlight structure/function characteristics of GRK interaction with GPCRs as well as a review of two recent reports that provide a molecular model for GRK-GPCR interaction. Finally, we offer our perspective on some future studies that we believe will drive this field.
Collapse
Affiliation(s)
- Konstantin E Komolov
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States.
| |
Collapse
|
21
|
Bansal S, Barathi VA, Iwata D, Agrawal R. Experimental autoimmune uveitis and other animal models of uveitis: An update. Indian J Ophthalmol 2016; 63:211-8. [PMID: 25971165 PMCID: PMC4448233 DOI: 10.4103/0301-4738.156914] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Over the past several decades, animal models of autoimmune uveitis directed at eye-specific antigens (Ags) have been developed. These have allowed researchers to understand the basic mechanisms that lead to these diseases and also recently helped the researchers in translational research for therapeutic interventions. Experimental autoimmune uveitis (EAU) is an animal disease model of human endogenous uveitis and can be induced in susceptible animals by immunization with retinal Ags. Ever since the first description of EAU in mice in 1988, several animal models of uveitis has been described by researchers. Disease-specific model for cytomegalovirus retinitis and tubercular uveitis has evolved our understanding of these complex entities. Endotoxin induced uveitis is another useful model for anterior uveitis, which is not an autoimmune process and is triggered by injection of bacterial endotoxin (lipopolysaccharides) resulting in a rapid short lasting uveitis. The current article will give an insight into the various EAU animal models and their current implications in translational research. The article will also highlight the different grading systems for EAU in the animal model.
Collapse
Affiliation(s)
| | | | | | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Singapore Eye Research Institute, Singapore; Institute of Ophthalmology, University College London, London,
| |
Collapse
|
22
|
Nogueras-Ortiz C, Yudowski GA. The Multiple Waves of Cannabinoid 1 Receptor Signaling. Mol Pharmacol 2016; 90:620-626. [PMID: 27338082 PMCID: PMC11037448 DOI: 10.1124/mol.116.104539] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/22/2016] [Indexed: 12/15/2022] Open
Abstract
The cannabinoid 1 receptor (CB1R) is one of the most abundant G protein-coupled receptors (GPCRs) in the central nervous system, with key roles during neurotransmitter release and synaptic plasticity. Upon ligand activation, CB1Rs may signal in three different spatiotemporal waves. The first wave, which is transient (<10 minutes) and initiated by heterotrimeric G proteins, is followed by a second wave (>5 minutes) that is mediated by β-arrestins. The third and final wave occurs at intracellular compartments and could be elicited by G proteins or β-arrestins. This complexity presents multiple challenges, including the correct classification of receptor ligands, the identification of the signaling pathways regulated by each wave, and the underlying molecular mechanisms and physiologic impacts of these waves. Simultaneously, it provides new opportunities to harness the therapeutic potential of the cannabinoid system and other GPCRs. Over the last several years, we have significantly expanded our understanding of the mechanisms and pathways downstream from the CB1R. The identification of receptor mutations that can bias signaling to specific pathways and the use of siRNA technology have been key tools to identifying which signaling cascades are controlled by G proteins or β-arrestins. Here, we review our current knowledge on CB1R signaling, with particular emphasis on the mechanisms and cascades mediated by β-arrestins downstream from the CB1R.
Collapse
Affiliation(s)
- Carlos Nogueras-Ortiz
- Institute of Neurobiology(C.N.-O., G.A.Y.) and Department of Anatomy and Neurobiology (G.A.Y.), University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Guillermo A Yudowski
- Institute of Neurobiology(C.N.-O., G.A.Y.) and Department of Anatomy and Neurobiology (G.A.Y.), University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
23
|
Deming JD, Pak JS, Shin JA, Brown BM, Kim MK, Aung MH, Lee EJ, Pardue MT, Craft CM. Arrestin 1 and Cone Arrestin 4 Have Unique Roles in Visual Function in an All-Cone Mouse Retina. Invest Ophthalmol Vis Sci 2016; 56:7618-28. [PMID: 26624493 DOI: 10.1167/iovs.15-17832] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Previous studies discovered cone phototransduction shutoff occurs normally for Arr1-/- and Arr4-/-; however, it is defective when both visual arrestins are simultaneously not expressed (Arr1-/-Arr4-/-). We investigated the roles of visual arrestins in an all-cone retina (Nrl-/-) since each arrestin has differential effects on visual function, including ARR1 for normal light adaptation, and ARR4 for normal contrast sensitivity and visual acuity. METHODS We examined Nrl-/-, Nrl-/-Arr1-/-, Nrl-/-Arr4-/-, and Nrl-/-Arr1-/-Arr4-/- mice with photopic electroretinography (ERG) to assess light adaptation and retinal responses, immunoblot and immunohistochemical localization analysis to measure retinal expression levels of M- and S-opsin, and optokinetic tracking (OKT) to measure the visual acuity and contrast sensitivity. RESULTS Study results indicated that Nrl-/- and Nrl-/-Arr4-/- mice light adapted normally, while Nrl-/-Arr1-/- and Nrl-/-Arr1-/-Arr4-/- mice did not. Photopic ERG a-wave, b-wave, and flicker amplitudes followed a general pattern in which Nrl-/-Arr4-/- amplitudes were higher than the amplitudes of Nrl-/-, while the amplitudes of Nrl-/-Arr1-/- and Nrl-/-Arr1-/-Arr4-/- were lower. All three visual arrestin knockouts had faster implicit times than Nrl-/- mice. M-opsin expression is lower when ARR1 is not expressed, while S-opsin expression is lower when ARR4 is not expressed. Although M-opsin expression is mislocalized throughout the photoreceptor cells, S-opsin is confined to the outer segments in all genotypes. Contrast sensitivity is decreased when ARR4 is not expressed, while visual acuity was normal except in Nrl-/-Arr1-/-Arr4-/-. CONCLUSIONS Based on the opposite visual phenotypes in an all-cone retina in the Nrl-/-Arr1-/- and Nrl-/-Arr4-/- mice, we conclude that ARR1 and ARR4 perform unique modulatory roles in cone photoreceptors.
Collapse
Affiliation(s)
- Janise D Deming
- Mary D. Allen Laboratory for Vision Research, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, USC Eye Institute, Los Angeles, California, United States
| | - Joseph S Pak
- Mary D. Allen Laboratory for Vision Research, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, USC Eye Institute, Los Angeles, California, United States
| | - Jung-A Shin
- Mary D. Allen Laboratory for Vision Research, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, USC Eye Institute, Los Angeles, California, United States 2Department of Anatomy, School of Medicine, Ewha Womans
| | - Bruce M Brown
- Mary D. Allen Laboratory for Vision Research, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, USC Eye Institute, Los Angeles, California, United States
| | - Moon K Kim
- Rehabilitation Research & Development Center of Excellence, Atlanta VA Medical Center, Decatur, Georgia, United States
| | - Moe H Aung
- Neuroscience/Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Eun-Jin Lee
- Mary D. Allen Laboratory for Vision Research, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, USC Eye Institute, Los Angeles, California, United States 5Department of Biomedical Engineering, University of Sou
| | - Machelle T Pardue
- Rehabilitation Research & Development Center of Excellence, Atlanta VA Medical Center, Decatur, Georgia, United States 4Neuroscience/Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Cheryl Mae Craft
- Mary D. Allen Laboratory for Vision Research, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, USC Eye Institute, Los Angeles, California, United States 6Department of Cell & Neurobiology, Keck School of Medic
| |
Collapse
|
24
|
Jean-Charles PY, Freedman NJ, Shenoy SK. Chapter Nine - Cellular Roles of Beta-Arrestins as Substrates and Adaptors of Ubiquitination and Deubiquitination. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:339-69. [PMID: 27378762 DOI: 10.1016/bs.pmbts.2016.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
β-Arrestin1 and β-arrestin2 are homologous adaptor proteins that are ubiquitously expressed in mammalian cells. They belong to a four-member family of arrestins that regulate the vast family of seven-transmembrane receptors that couple to heterotrimeric G proteins (7TMRs or GPCRs), and that modulate 7TMR signal transduction. β-Arrestins were originally identified in the context of signal inhibition via the 7TMRs because they competed with and thereby blocked G protein coupling to 7TMRs. Currently, in addition to their role as desensitizers of signaling, β-arrestins are appreciated as multifunctional adaptors that mediate trafficking and signal transduction of not only 7TMRs, but a growing list of additional receptors, ion channels, and nonreceptor proteins. β-Arrestins' interactions with their multifarious partners are based on their dynamic conformational states rather than particular domain-domain interactions. β-Arrestins adopt activated conformations upon 7TMR association. In addition, β-arrestins undergo various posttranslational modifications that are choreographed by activated 7TMRs, including phosphorylation, ubiquitination, acetylation, nitrosylation, and SUMOylation. Ubiquitination of β-arrestins is critical for their high-affinity interaction with 7TMRs as well as with endocytic adaptor proteins and signaling kinases. β-Arrestins also function as critical adaptors for ubiquitination and deubiquitination of various cellular proteins, and thereby affect the longevity of signal transducers and the intensity of signal transmission.
Collapse
Affiliation(s)
- P-Y Jean-Charles
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, North Carolina, United States
| | - N J Freedman
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, North Carolina, United States; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States
| | - S K Shenoy
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, North Carolina, United States; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States.
| |
Collapse
|
25
|
Wisler JW, Harris EM, Raisch M, Mao L, Kim J, Rockman HA, Lefkowitz RJ. The role of β-arrestin2-dependent signaling in thoracic aortic aneurysm formation in a murine model of Marfan syndrome. Am J Physiol Heart Circ Physiol 2015; 309:H1516-27. [PMID: 26371162 PMCID: PMC4666970 DOI: 10.1152/ajpheart.00291.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/15/2015] [Indexed: 01/14/2023]
Abstract
Ang II type 1a receptor (AT1aR)-mediated activation of MAPKs contributes to thoracic aortic aneurysm (TAA) development in Marfan syndrome (MFS). β-Arrestin2 (βarr2) is known to mediate AT1aR-dependent MAPK activation, as well as proproliferative and profibrotic signaling in aortic vascular smooth muscle cells. Therefore, we investigated whether βarr2-dependent signaling contributes to TAA formation in MFS. We used a murine model of MFS [fibrillin (Fbn)(C1039G/+)] to generate an MFS murine model in combination with genetic βarr2 deletion (Fbn(C1039G/+)/βarr2(-/-)). Fbn(C1039G/+)/βarr2(-/-) mice displayed delayed aortic root dilation compared with Fbn(C1039G/+) mice. The mRNA and protein expression of several mediators of TAA formation, including matrix metalloproteinase (MMP)-2 and -9, was reduced in the aorta of Fbn(C1039G/+)/βarr2(-/-) mice relative to Fbn(C1039G/+) mice. Activation of ERK1/2 was also decreased in the aortas of Fbn(C1039G/+)/βarr2(-/-) mice compared with Fbn(C1039G/+) animals. Small interfering RNA targeting βarr2 inhibited angiotensin-stimulated expression of proaneurysmal signaling mediators in primary aortic root smooth muscle cells. Angiotensin-stimulated expression of the proaneurysmal signaling mediators MMP-2 and -9 was inhibited by blockade of ERK1/2 or the EGF receptor, whereas blockade of the transforming growth factor-β receptor had no effect. These results suggest that βarr2 contributes to TAA formation in MFS by regulating ERK1/2-dependent expression of proaneurysmal genes and proteins downstream of the AT1aR. Importantly, this demonstration of the unique signaling mechanism by which βarr2 contributes to aneurysm formation identifies multiple novel, potential therapeutic targets in MFS.
Collapse
MESH Headings
- Angiotensins/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Arrestins/genetics
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Disease Models, Animal
- ErbB Receptors/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/genetics
- Fibrillins
- Fibrosis
- MAP Kinase Signaling System
- Marfan Syndrome/genetics
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 9/genetics
- Mice
- Mice, Knockout
- Microfilament Proteins/genetics
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- RNA, Messenger/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Signal Transduction
- Transcriptome
- beta-Arrestins
Collapse
Affiliation(s)
- James W Wisler
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Emily M Harris
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Michael Raisch
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina
| | - Lan Mao
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina; Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina
| | - Robert J Lefkowitz
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Biochemistry, Duke University Medical Center, Durham, North Carolina; and Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
26
|
Deming JD, Pak JS, Brown BM, Kim MK, Aung MH, Eom YS, Shin JA, Lee EJ, Pardue MT, Craft CM. Visual Cone Arrestin 4 Contributes to Visual Function and Cone Health. Invest Ophthalmol Vis Sci 2015; 56:5407-16. [PMID: 26284544 DOI: 10.1167/iovs.15-16647] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Visual arrestins (ARR) play a critical role in shutoff of rod and cone phototransduction. When electrophysiological responses are measured for a single mouse cone photoreceptor, ARR1 expression can substitute for ARR4 in cone pigment desensitization; however, each arrestin may also contribute its own, unique role to modulate other cellular functions. METHODS A combination of ERG, optokinetic tracking, immunohistochemistry, and immunoblot analysis was used to investigate the retinal phenotypes of Arr4 null mice (Arr4-/-) compared with age-matched control, wild-type mice. RESULTS When 2-month-old Arr4-/- mice were compared with wild-type mice, they had diminished visual acuity and contrast sensitivity, yet enhanced ERG flicker response and higher photopic ERG b-wave amplitudes. In contrast, in older Arr4-/- mice, all ERG amplitudes were significantly reduced in magnitude compared with age-matched controls. Furthermore, in older Arr4-/- mice, the total cone numbers decreased and cone opsin protein immunoreactive expression levels were significantly reduced, while overall photoreceptor outer nuclear layer thickness was unchanged. CONCLUSIONS Our study demonstrates that Arr4-/- mice display distinct phenotypic differences when compared to controls, suggesting that ARR4 modulates essential functions in high acuity vision and downstream cellular signaling pathways that are not fulfilled or substituted by the coexpression of ARR1, despite its high expression levels in all mouse cones. Without normal ARR4 expression levels, cones slowly degenerate with increasing age, making this a new model to study age-related cone dystrophy.
Collapse
Affiliation(s)
- Janise D Deming
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Joseph S Pak
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Bruce M Brown
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Moon K Kim
- Rehabilitation Research & Development Center of Excellence, Atlanta VA Medical Center, Decatur, Georgia, United States
| | - Moe H Aung
- Neuroscience/Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Yun Sung Eom
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 4Dornsife College of Letters, Arts and Sciences, Univers
| | - Jung-A Shin
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 5Department of Anatomy, School of Medicine, Ewha Womans
| | - Eun-Jin Lee
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 6Department of Biomedical Engineering, University of Sou
| | - Machelle T Pardue
- Rehabilitation Research & Development Center of Excellence, Atlanta VA Medical Center, Decatur, Georgia, United States 3Neuroscience/Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Cheryl Mae Craft
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 7Department of Cell & Neurobiology, Keck School of Medic
| |
Collapse
|
27
|
Deming JD, Shin JA, Lim K, Lee EJ, Van Craenenbroeck K, Craft CM. Dopamine receptor D4 internalization requires a beta-arrestin and a visual arrestin. Cell Signal 2015; 27:2002-13. [PMID: 26169958 DOI: 10.1016/j.cellsig.2015.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 06/30/2015] [Indexed: 11/25/2022]
Abstract
PURPOSE The G-protein coupled receptor (GPCR) Dopamine Receptor D4 (DRD4) plays an essential role in cAMP regulation and gap junctional coupling in the photoreceptors, where DRD4 expression is under circadian control. Previous in vitro transfection studies of human DRD4 desensitization have reported that DRD4 is not internalized upon dopamine stimulation when beta-arrestin is co-transfected with DRD4. We hypothesized that the visual arrestins, ARR1 and ARR4, play a modulatory role in DRD4 desensitization in the photoreceptors. METHODS To test this hypothesis, immunohistochemistry analysis of mouse retinas was used to determine the cellular localization of beta-arrestins and DRD4 in photoreceptors. In vitro studies were performed in HEK293T cells transiently transfected with human DRD4 and arrestins. First, co-immunoprecipitation experiments were executed to test protein-protein interactions and to investigate the effect of dopamine stimulation. Second, immunohistochemistry analysis was implemented to study DRD4 internalization and translocation of ARR4. RESULTS Immunohistochemistry studies of mouse retinas confirmed the expression of beta-arrestin 2, ARR1 and ARR4, as well as DRD4 in mouse cone photoreceptor inner segments. Co-immunoprecipitation experiments revealed a dopamine-dependent protein-protein interaction between human DRD4 and ARR4. In vitro internalization experiments showed that no detectable internalization of DRD4 was observed with any single arrestin co-transfected. However, a dopamine-dependent internalization of DRD4 was observed with three out of six sets of two arrestins co-transfected with DRD4. Each of these pairs of arrestins contained one visual arrestin and one beta-arrestin, and no internalization was observed with either two visual arrestins or two beta-arrestins. Additional time-course experiments revealed that in vitro, ARR4 translocates to co-localize with DRD4 at the plasma membrane in response to 30min of dopamine stimulation. CONCLUSIONS The results have functional implications and we hypothesize that the desensitization and internalization of DRD4 in photoreceptors are synergistically mediated by both visual and beta-arrestins. These results are additionally unique because they demonstrate for the first time that at least one G-protein coupled receptor, DRD4, requires two arrestins for desensitization and internalization, and opens up the possibility that other G-protein coupled receptors may require more than one arrestin for desensitization and/or internalization.
Collapse
Affiliation(s)
- Janise D Deming
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA.
| | - Jung-A Shin
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA; Department of Anatomy, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea.
| | - Kayleen Lim
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA.
| | - Eun-Jin Lee
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA; Department of Biomedical Engineering, University of Southern California Viterbi School of Engineering, 1042 Downey Way, Los Angeles, CA 90089-1111, USA.
| | - Kathleen Van Craenenbroeck
- Laboratory of GPCR Expression and Signal Transduction (L-GEST), Ghent University-UGent, K.L. Ledeganckstraat 35, B-9000 Gent, Belgium.
| | - Cheryl Mae Craft
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA; Department of Cell & Neurobiology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Center 135H, Los Angeles, CA 90033, USA.
| |
Collapse
|
28
|
Palczewski K. Chemistry and biology of the initial steps in vision: the Friedenwald lecture. Invest Ophthalmol Vis Sci 2014; 55:6651-72. [PMID: 25338686 DOI: 10.1167/iovs.14-15502] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Visual transduction is the process in the eye whereby absorption of light in the retina is translated into electrical signals that ultimately reach the brain. The first challenge presented by visual transduction is to understand its molecular basis. We know that maintenance of vision is a continuous process requiring the activation and subsequent restoration of a vitamin A-derived chromophore through a series of chemical reactions catalyzed by enzymes in the retina and retinal pigment epithelium (RPE). Diverse biochemical approaches that identified key proteins and reactions were essential to achieve a mechanistic understanding of these visual processes. The three-dimensional arrangements of these enzymes' polypeptide chains provide invaluable insights into their mechanisms of action. A wealth of information has already been obtained by solving high-resolution crystal structures of both rhodopsin and the retinoid isomerase from pigment RPE (RPE65). Rhodopsin, which is activated by photoisomerization of its 11-cis-retinylidene chromophore, is a prototypical member of a large family of membrane-bound proteins called G protein-coupled receptors (GPCRs). RPE65 is a retinoid isomerase critical for regeneration of the chromophore. Electron microscopy (EM) and atomic force microscopy have provided insights into how certain proteins are assembled to form much larger structures such as rod photoreceptor cell outer segment membranes. A second challenge of visual transduction is to use this knowledge to devise therapeutic approaches that can prevent or reverse conditions leading to blindness. Imaging modalities like optical coherence tomography (OCT) and scanning laser ophthalmoscopy (SLO) applied to appropriate animal models as well as human retinal imaging have been employed to characterize blinding diseases, monitor their progression, and evaluate the success of therapeutic agents. Lately two-photon (2-PO) imaging, together with biochemical assays, are revealing functional aspects of vision at a new molecular level. These multidisciplinary approaches combined with suitable animal models and inbred mutant species can be especially helpful in translating provocative cell and tissue culture findings into therapeutic options for further development in animals and eventually in humans. A host of different approaches and techniques is required for substantial progress in understanding fundamental properties of the visual system.
Collapse
Affiliation(s)
- Krzysztof Palczewski
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
29
|
Wu JX, Shan FX, Zheng JN, Pei DS. β-arrestin promotes c-Jun N-terminal kinase mediated apoptosis via a GABA(B)R·β-arrestin·JNK signaling module. Asian Pac J Cancer Prev 2014; 15:1041-6. [PMID: 24568448 DOI: 10.7314/apjcp.2014.15.2.1041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Evidence is growing that the GABAB receptor, which belongs to the G protein-coupled receptor (GPCR) superfamily, is involved in tumorigenesis. Recent studies have shown that β-arrestin can serve as a scaffold to recruit signaling protein c-Jun N-terminal knase (JNK) to GPCR. Here we investigated whether β-arrestin recruits JNK to the GABAB receptor and facilitates its activation to affect the growth of cancer cells. Our results showed that β-arrestin expression is decreased in breast cancer cells in comparison with controls. β-arrestin could enhance interactions of the GABABR·β-arrestin·JNK signaling module in MCF-7 and T-47D cells. Further studies revealed that increased expression of β-arrestin enhances the phosphorylation of JNK and induces cancer cells apoptosis. Collectively, these results indicate that β-arrestin promotes JNK mediated apoptosis via a GABABR·β-arrestin·JNK signaling module.
Collapse
Affiliation(s)
- Jin-Xia Wu
- The First Clinical Medical College, Nanjing Medical University, Nanjing, China E-mail : ;
| | | | | | | |
Collapse
|
30
|
Abstract
G-protein-coupled receptors (GPCRs) are the primary interaction partners for arrestins. The visual arrestins, arrestin1 and arrestin4, physiologically bind to only very few receptors, i.e., rhodopsin and the color opsins, respectively. In contrast, the ubiquitously expressed nonvisual variants β-arrestin1 and 2 bind to a large number of receptors in a fairly nonspecific manner. This binding requires two triggers, agonist activation and receptor phosphorylation by a G-protein-coupled receptor kinase (GRK). These two triggers are mediated by two different regions of the arrestins, the "phosphorylation sensor" in the core of the protein and a less well-defined "activation sensor." Binding appears to occur mostly in a 1:1 stoichiometry, involving the N-terminal domain of GPCRs, but in addition a second GPCR may loosely bind to the C-terminal domain when active receptors are abundant.Arrestin binding initially uncouples GPCRs from their G-proteins. It stabilizes receptors in an active conformation and also induces a conformational change in the arrestins that involves a rotation of the two domains relative to each other plus changes in the polar core. This conformational change appears to permit the interaction with further downstream proteins. The latter interaction, demonstrated mostly for β-arrestins, triggers receptor internalization as well as a number of nonclassical signaling pathways.Open questions concern the exact stoichiometry of the interaction, possible specificity with regard to the type of agonist and of GRK involved, selective regulation of downstream signaling (=biased signaling), and the options to use these mechanisms as therapeutic targets.
Collapse
Affiliation(s)
- Martin J Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Straße 9, 97078, Würzburg, Germany,
| | | |
Collapse
|
31
|
Smith WC. The role of arrestins in visual and disease processes of the eye. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:243-65. [PMID: 23764057 DOI: 10.1016/b978-0-12-394440-5.00010-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Visual arrestins are well known for their function in quenching the phototransduction process in rods and cones. Perhaps not as well known is their participation in multiple other processes in the normal and disease states of the eye. This chapter covers the range of the known functions of the visual arrestins, beginning with their classical role in quenching light-activated visual pigments. The role of visual arrestins is also reviewed from the perspective of their dynamic mobility whereby they redistribute significantly between the compartments of highly polarized photoreceptor cells. Additional roles of the visual arrestins are also reviewed based on new interacting partners that have been discovered over the past decade. Finally, the contribution of the visual arrestins to diseases of the visual system is explored.
Collapse
Affiliation(s)
- W Clay Smith
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
32
|
Kommaddi RP, Shenoy SK. Arrestins and protein ubiquitination. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:175-204. [PMID: 23764054 DOI: 10.1016/b978-0-12-394440-5.00007-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The adaptor proteins, β-arrestins 1 and 2, were originally identified as inhibitors of G protein signaling at the seven-transmembrane receptors (7TMRs, also called G protein-coupled receptors or GPCRs). Subsequent studies have established β-arrestins as critical multifunctional 7TMR adaptors that mediate receptor trafficking and activate G protein-independent signaling pathways. 7TMR activation leads not only to the recruitment of arrestin proteins upon phosphorylation by GPCR kinases but also to β-arrestin ubiquitination. This posttranslational modification of β-arrestin is appended by specific E3 ubiquitin ligases and reversed by deubiquitinases, which are also recruited in a receptor- and agonist-specific manner. β-Arrestin ubiquitination allows it to form protein complexes with activated 7TMRs, endocytic proteins such as clathrin, and phosphorylated ERK1/2. β-Arrestin ubiquitination is dependent on its activated conformation and likely regulates timing and subcellular localization of various protein interactions during receptor trafficking and signaling. β-Arrestins also serve as adaptors that escort E3 ubiquitin ligases to mediate ubiquitination of a wide list of substrate proteins including 7TMRs and provide an added layer of regulation for defining substrate specificity in the cellular ubiquitination pathway.
Collapse
Affiliation(s)
- Reddy Peera Kommaddi
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
33
|
Lefkowitz RJ. Arrestins Come of Age. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:3-18. [DOI: 10.1016/b978-0-12-394440-5.00001-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
34
|
Walther C, Ferguson SSG. Arrestins: role in the desensitization, sequestration, and vesicular trafficking of G protein-coupled receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:93-113. [PMID: 23764051 DOI: 10.1016/b978-0-12-394440-5.00004-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the years, β-arrestins have emerged as multifunctional molecular scaffolding proteins regulating almost every imaginable G protein-coupled receptor (GPCR) function. Originally discovered as GPCR-desensitizing molecules, they have been shown to also serve as important regulators of GPCR signaling, sequestration, and vesicular trafficking. This broad functional role implicates β-arrestins as key regulatory proteins for cellular function. Hence, this chapter summarizes the current understanding of the β-arrestin family's unique ability to control the kinetics as well as the extent of GPCR activity at the level of desensitization, sequestration, and subsequent intracellular trafficking.
Collapse
Affiliation(s)
- Cornelia Walther
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, Western University Canada, London, Ontario, Canada
| | | |
Collapse
|
35
|
Abstract
The model of experimental autoimmune uveitis (EAU) in mice and in rats is described. EAU targets immunologically privileged retinal antigens and serves as a model of autoimmune uveitis in humans as well as a model for autoimmunity in a more general sense. EAU is a well-characterized, robust, and reproducible model that is easily followed and quantitated. It is inducible with synthetic peptides derived from retinal autoantigens in commonly available strains of rats and mice. The ability to induce EAU in various gene-manipulated, including HLA-transgenic, mouse strains makes the EAU model suitable for the study of basic mechanisms as well as in clinically relevant interventions.
Collapse
|
36
|
Gurevich VV, Hanson SM, Song X, Vishnivetskiy SA, Gurevich EV. The functional cycle of visual arrestins in photoreceptor cells. Prog Retin Eye Res 2011; 30:405-430. [PMID: 21824527 PMCID: PMC3196764 DOI: 10.1016/j.preteyeres.2011.07.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 01/14/2023]
Abstract
Visual arrestin-1 plays a key role in the rapid and reproducible shutoff of rhodopsin signaling. Its highly selective binding to light-activated phosphorylated rhodopsin is an integral part of the functional perfection of rod photoreceptors. Structure-function studies revealed key elements of the sophisticated molecular mechanism ensuring arrestin-1 selectivity and paved the way to the targeted manipulation of the arrestin-1 molecule to design mutants that can compensate for congenital defects in rhodopsin phosphorylation. Arrestin-1 self-association and light-dependent translocation in photoreceptor cells work together to keep a constant supply of active rhodopsin-binding arrestin-1 monomer in the outer segment. Recent discoveries of arrestin-1 interaction with other signaling proteins suggest that it is a much more versatile signaling regulator than previously thought, affecting the function of the synaptic terminals and rod survival. Elucidation of the fine molecular mechanisms of arrestin-1 interactions with rhodopsin and other binding partners is necessary for the comprehensive understanding of rod function and for devising novel molecular tools and therapeutic approaches to the treatment of visual disorders.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave, PRB, Rm 417D, Nashville, TN 37232, USA.
| | | | | | | | | |
Collapse
|
37
|
Comparative modeling of retinol-binding protein-3 and retinal S-antigen in Eales' disease and prediction of their binding sites using computational methods. J Ocul Biol Dis Infor 2011; 3:88-91. [PMID: 22833777 DOI: 10.1007/s12177-011-9060-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 07/12/2011] [Indexed: 10/18/2022] Open
Abstract
Retinal S-antigen and interphotoreceptor retinoid-binding protein-3 play a significant role in the etiopathogenesis of Eales' disease. Protein 3D structures are functionally very important and play a significant role in progression of the disease, hence these 3D structures are better target for further drug designing and relative studies. We developed 3D model structure of retinol-binding protein-3 and retinal S-antigen protein of human involved in Eales' disease. Functional site prediction is a very important and related step; hence, in the current course of analysis, we predicted putative functional site residues in the target proteins. Molecular models of these proteins of Eales' disease as documented in this study may provide a valuable aid for designing an inhibitor or better ligand against Eales' disease and could play a significant role in drug design.
Collapse
|
38
|
A variant of arrestin-1 binds rod outer segment membranes in a light-independent manner. Arch Biochem Biophys 2011; 507:219-31. [DOI: 10.1016/j.abb.2010.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 12/13/2010] [Accepted: 12/15/2010] [Indexed: 11/21/2022]
|
39
|
Le monoxyde d’azote, bio-marqueur de l’uvéite auto-immune expérimentale induite par l’antigène S. J Fr Ophtalmol 2010; 33:693-700. [DOI: 10.1016/j.jfo.2010.09.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 08/16/2010] [Accepted: 09/20/2010] [Indexed: 01/05/2023]
|
40
|
Kenakin T, Miller LJ. Seven transmembrane receptors as shapeshifting proteins: the impact of allosteric modulation and functional selectivity on new drug discovery. Pharmacol Rev 2010; 62:265-304. [PMID: 20392808 PMCID: PMC2879912 DOI: 10.1124/pr.108.000992] [Citation(s) in RCA: 469] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
It is useful to consider seven transmembrane receptors (7TMRs) as disordered proteins able to allosterically respond to a number of binding partners. Considering 7TMRs as allosteric systems, affinity and efficacy can be thought of in terms of energy flow between a modulator, conduit (the receptor protein), and a number of guests. These guests can be other molecules, receptors, membrane-bound proteins, or signaling proteins in the cytosol. These vectorial flows of energy can yield standard canonical guest allostery (allosteric modification of drug effect), effects along the plane of the cell membrane (receptor oligomerization), or effects directed into the cytosol (differential signaling as functional selectivity). This review discusses these apparently diverse pharmacological effects in terms of molecular dynamics and protein ensemble theory, which tends to unify 7TMR behavior toward cells. Special consideration will be given to functional selectivity (biased agonism and biased antagonism) in terms of mechanism of action and potential therapeutic application. The explosion of technology that has enabled observation of diverse 7TMR behavior has also shown how drugs can have multiple (pluridimensional) efficacies and how this can cause paradoxical drug classification and nomenclatures.
Collapse
Affiliation(s)
- Terry Kenakin
- GlaxoSmithKline, 5 Moore Drive, Mailtstop V-287, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
41
|
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.
Collapse
|
42
|
Brown BM, Ramirez T, Rife L, Craft CM. Visual Arrestin 1 contributes to cone photoreceptor survival and light adaptation. Invest Ophthalmol Vis Sci 2009; 51:2372-80. [PMID: 20019357 DOI: 10.1167/iovs.09-4895] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To evaluate morphologic and functional contributions of Arrestin 1 (Arr1) and Arrestin 4 (Arr4) in cone photoreceptors, the authors examined the phenotypes of visual arrestin knockout mice (Arr1(-/-), Arr4(-/-), Arr1(-/-)Arr4(-/-) [Arr-DKO]) reared in darkness. METHODS Retinal rods and cones were evaluated in wild-type (WT), Arr1(-/-), Arr4(-/-), and Arr-DKO mice using quantitative morphologic analysis, immunoblot, immunohistochemistry, TUNEL, and electroretinographic (ERG) techniques. RESULTS Compared with either Arr4(-/-) or WT, Arr1(-/-) and Arr-DKO mice had increased apoptotic nuclei in their retinal outer nuclear layer (ONL) at postnatal day (P) 22. By P60, cone density was significantly diminished, but the ONL appeared normal. After 1 minute of background illumination, cone ERG b-wave amplitudes were similar in WT and all Arr KO mice. However, by 3 minutes and continuing through 15 minutes of light adaptation, the cone b-wave amplitudes of WT and Arr4(-/-) mice increased significantly over those of the Arr1(-/-) and Arr-DKO mice, which demonstrated no cone b-wave amplitude increase. In contrast, ERG flicker analysis after the 15-minute light adaptation period demonstrated no loss in amplitude for either Arr1(-/-) or Arr4(-/-) mice, whereas Arr-DKO had significantly lower amplitudes. When Arr1 expression was restored in Arr1(-/-) mice (+p48(Arr1-/-)), normal cone density and light-adapted ERG b-wave amplitudes were observed. CONCLUSIONS In the adult dark-reared Arr1(-/-) and Arr-DKO mice, viable cones diminish over time. Arr1 expression is essential for cone photoreceptor survival and light adaptation, whereas either Arr1 or Arr4 is necessary for maintaining normal flicker responses.
Collapse
Affiliation(s)
- Bruce M Brown
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033-9224, USA
| | | | | | | |
Collapse
|
43
|
Wang L, Martin B, Brenneman R, Luttrell LM, Maudsley S. Allosteric modulators of g protein-coupled receptors: future therapeutics for complex physiological disorders. J Pharmacol Exp Ther 2009; 331:340-8. [PMID: 19667132 PMCID: PMC2775272 DOI: 10.1124/jpet.109.156380] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 08/07/2009] [Indexed: 01/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are one of the most important classes of proteins in the genome, not only because of their tremendous molecular diversity but because they are the targets of nearly 50% of current pharmacotherapeutics. The majority of these drugs affect GPCR activity by binding to a similar molecular site as the endogenous cognate ligand for the receptor. These "orthosterically" targeted drugs currently dominate the existing pharmacopeia. Over the past two decades, novel opportunities for drug discovery have risen from a greater understanding of the complexity of GPCR signaling. A striking example of this is the appreciation that many GPCRs possess functional allosteric binding sites. Allosteric modulator ligands bind receptor domains topographically distinct from the orthosteric site, altering the biological activity of the orthosteric ligand by changing its binding affinity, functional efficacy, or both. This additional receptor signaling complexity can be embraced and exploited for the next generation of GPCR-targeted therapies. Despite the challenges associated with detecting and quantifying the myriad of possible allosteric effects on GPCR activity, allosteric ligands offer the prospect of engendering a facile stimulus-bias in orthosteric ligand signaling, paving the way for not only receptor-selective but also signaling pathway-selective therapies. Allosteric modulators possess specific advantages when considering the treatment of multifactorial syndromes, such as metabolic diseases or age-related cognitive impairment, because they may not greatly affect neurotransmitter or hormone release patterns, thus maintaining the integrity of complex signaling networks that underlie perception, memory patterns, or neuroendocrinological axes while introducing therapeutically beneficial signal bias.
Collapse
Affiliation(s)
- Liyun Wang
- Receptor Pharmacology Unit, and Metabolism Unit, the National Institutes of Health, National Institute on Aging, Baltimore, Maryland 21224, USA
| | | | | | | | | |
Collapse
|
44
|
Roles of G protein and β-arrestin in dopamine D 2 receptor-mediated ERK activation. Biochem Biophys Res Commun 2008; 377:705-709. [DOI: 10.1016/j.bbrc.2008.10.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 10/10/2008] [Indexed: 11/21/2022]
|
45
|
Abstract
Upon their discovery, beta-arrestins 1 and 2 were named for their capacity to sterically hinder the G protein coupling of agonist-activated seven-transmembrane receptors, ultimately resulting in receptor desensitization. Surprisingly, recent evidence shows that beta-arrestins can also function to activate signaling cascades independently of G protein activation. By serving as multiprotein scaffolds, the beta-arrestins bring elements of specific signaling pathways into close proximity. beta-Arrestin regulation has been demonstrated for an ever-increasing number of signaling molecules, including the mitogen-activated protein kinases ERK, JNK, and p38 as well as Akt, PI3 kinase, and RhoA. In addition, investigators are discovering new roles for beta-arrestins in nuclear functions. Here, we review the signaling capacities of these versatile adapter molecules and discuss the possible implications for cellular processes such as chemotaxis and apoptosis.
Collapse
Affiliation(s)
- Scott M DeWire
- Howard Hughes Medical Institute and Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
46
|
Wang Q, Limbird LE. Regulation of alpha2AR trafficking and signaling by interacting proteins. Biochem Pharmacol 2006; 73:1135-45. [PMID: 17229402 PMCID: PMC1885238 DOI: 10.1016/j.bcp.2006.12.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 12/11/2006] [Accepted: 12/20/2006] [Indexed: 01/23/2023]
Abstract
The continuing discovery of new G protein-coupled receptor (GPCR) interacting proteins and clarification of the functional consequences of these interactions has revealed multiple roles for these events. Some of these interactions serve to scaffold GPCRs to particular cellular micro-compartments or to tether them to defined signaling molecules, while other GPCR-protein interactions control GPCR trafficking and the kinetics of GPCR-mediated signaling transduction. This review provides a general overview of the variety of GPCR-protein interactions reported to date, and then focuses on one prototypical GPCR, the alpha(2)AR, and the in vitro and in vivo significance of its reciprocal interactions with arrestin and spinophilin. It seems appropriate to recognize the life and career of Arthur Hancock with a summary of studies that both affirm and surprise our preconceived notions of how nature is designed, as his career-long efforts similarly affirmed the complexity of human biology and attempted to surprise pathological changes in that biology with novel, discovery-based therapeutic interventions. Dr. Hancock's love of life, of family, and of commitment to making the world a better place are a model of the life well lived, and truly missed by those who were privileged to know, and thus love, him.
Collapse
Affiliation(s)
- Qin Wang
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL35294
| | - Lee E. Limbird
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN 37208
| |
Collapse
|
47
|
Wang Q, Lu R, Zhao J, Limbird LE. Arrestin serves as a molecular switch, linking endogenous alpha2-adrenergic receptor to SRC-dependent, but not SRC-independent, ERK activation. J Biol Chem 2006; 281:25948-55. [PMID: 16809338 DOI: 10.1074/jbc.m605415200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our previous studies have demonstrated that neither receptor endocytosis nor arrestin is required for ERK activation by the alpha2-adrenergic receptor (Wang, Q., Zhao, J., Brady, A. E., Feng, J., Allen, P. B., Lefkowitz, R. J., Greengard, P., and Limbird, L. E. (2004) Science 304, 1940-1944). The present studies address whether arrestin plays a role in determining the route of alpha2AR-evoked ERK signaling activation, taking advantage of endogenous expression of the alpha(2A)AR subtype in mouse embryonic fibroblasts (MEFs) and the availability of MEFs without arrestin expression (derived from Arr2,3-/- mice). Our data demonstrate that the endogenous alpha(2A)AR evokes ERK phosphorylation through both a Src-dependent and a Src-independent pathway, both of which are G protein dependent and converge on the Ras-Raf-MEK pathway. Arrestin is essential to recruit Src to this process, as alpha(2A)AR-mediated ERK signaling in Arr2,3-/- MEFs does not involve Src. Stimulation of alpha(2A)AR enhances arrestin-Src interaction and promotes activation of Src. alpha2 agonists have similar potencies in stimulating Src-dependent and Src-independent ERK phosphorylation in wild-type and Arr2,3-/- cells, respectively. However, Src-independent alpha(2A)AR-mediated ERK stimulation has both a longer duration of activation and a more rapid translocation of pERK into the nucleus when compared with Src-dependent activation. These data not only affirm the role of arrestin as an escort for signaling molecules such as Src family kinases but also demonstrate the impact of arrestin-dependent modulation on both the temporal and spatial properties of ERK activation.
Collapse
Affiliation(s)
- Qin Wang
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | |
Collapse
|
48
|
Gurevich VV, Gurevich EV. The structural basis of arrestin-mediated regulation of G-protein-coupled receptors. Pharmacol Ther 2006; 110:465-502. [PMID: 16460808 PMCID: PMC2562282 DOI: 10.1016/j.pharmthera.2005.09.008] [Citation(s) in RCA: 371] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Accepted: 09/22/2005] [Indexed: 12/23/2022]
Abstract
The 4 mammalian arrestins serve as almost universal regulators of the largest known family of signaling proteins, G-protein-coupled receptors (GPCRs). Arrestins terminate receptor interactions with G proteins, redirect the signaling to a variety of alternative pathways, and orchestrate receptor internalization and subsequent intracellular trafficking. The elucidation of the structural basis and fine molecular mechanisms of the arrestin-receptor interaction paved the way to the targeted manipulation of this interaction from both sides to produce very stable or extremely transient complexes that helped to understand the regulation of many biologically important processes initiated by active GPCRs. The elucidation of the structural basis of arrestin interactions with numerous non-receptor-binding partners is long overdue. It will allow the construction of fully functional arrestins in which the ability to interact with individual partners is specifically disrupted or enhanced by targeted mutagenesis. These "custom-designed" arrestin mutants will be valuable tools in defining the role of various interactions in the intricate interplay of multiple signaling pathways in the living cell. The identification of arrestin-binding sites for various signaling molecules will also set the stage for designing molecular tools for therapeutic intervention that may prove useful in numerous disorders associated with congenital or acquired disregulation of GPCR signaling.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
49
|
Abstract
G protein-coupled receptors (GPCR) interact not only with heterotrimeric G proteins but also with accessory proteins called GPCR interacting proteins (GIP). These proteins have important functions. They are implicated in GPCR targeting to specific cellular compartments, in their assembling into large functional complexes called "receptosomes," in their trafficking to and from the plasma membrane, and in the fine-tuning of their signaling properties. There are several types of GIPs. Some are transmembrane proteins such as another GPCR (homodimerization and heterodimerization), ionic channels, ionotropic receptors, and single transmembrane proteins. The latter is implicated in the fine-tuning of receptor pharmacology or signaling. Other GIPs are soluble proteins interacting mainly with the "magic" C-terminal tail. Among them, PDZ domain-containing proteins are the most abundant. They generally, but not always, interact with the extreme C-terminal domain of GPCRs. Some GIPs interact with specific sequences of the C-terminal such as the Homer binding sequence (-PPxxFR-), the dopamine receptor interacting protein (DRIP) binding sequence (-FxxxFxxxF-), etc. Finally, only few GIPs have been found thus far to interact with the third intracellular loop of GPCRs. The future will tell us if this situation is only due to technical reasons.
Collapse
Affiliation(s)
- Joël Bockaert
- UPR CNRS 2580, CCIPE, 141 Rue de la Cardonille, 34094 Montpellier Cedex 5, France.
| | | | | | | |
Collapse
|
50
|
Claing A. Regulation of G protein-coupled receptor endocytosis by ARF6 GTP-binding proteins. Biochem Cell Biol 2004; 82:610-7. [PMID: 15674428 DOI: 10.1139/o04-113] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The function of G protein-coupled receptors is regulated by a broad variety of membrane-bound and intracellular proteins. These act in concert to activate signaling pathways that will lead to the desensitization of activated receptors and, for most receptor types, their trafficking to intracellular compartments. This review focuses mainly on the endocytic pathways used by a G protein-coupled receptor and on the proteins that play an essential role in the regulation of the internalization process, most specifically the ADP-ribosylation factors. This family of proteins has been shown to be important for vesicle trafficking between different cellular membranes. The latest findings regarding the molecular mechanisms that regulate internalization of an agonist-stimulated receptor are presented here. Finally, a perspective on how ARF6 proteins might regulate the internalization process is also proposed.Key words: G protein-coupled receptors, endocytosis, ADP-ribosylation factor.
Collapse
Affiliation(s)
- Audrey Claing
- Department of Pharmacology, School of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada.
| |
Collapse
|