1
|
Liu J, Zhang L, Liu L, Wu T, Wang L, Han Q. The potential capacities of FTY720: Novel therapeutic functions, targets, and mechanisms against diseases. Eur J Med Chem 2025; 290:117508. [PMID: 40120496 DOI: 10.1016/j.ejmech.2025.117508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Fingolimod (FTY720), an antagonist of sphingosine-1-phosphate (S1P), functions by binding to S1P receptors (S1PRs), excluding S1PR2. It received approval from the Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) in 2010. As the first non-selective oral agonist for S1PRs, FTY720's diverse and systemic receptor expression often leads to alterations in various signaling pathways and multiple systems, making it a subject of intense research. Recent studies have identified a wide range of novel or potential functions for FTY720 beyond its application in MS. These include effects on the blood-brain barrier (BBB), vascular system, organelles, and cell death, as well as potential applications in organ transplantation, immune disorders, oncological conditions, neurological and psychiatric disorders, viral infections, and hypersensitivity diseases. This paper reviews the novel roles, targets, and mechanisms of FTY720 that hold promise for clinical utility. Additionally, it summarizes FTY720's derivation and development process, the characterization and mechanism of the structure of FTY720-P bound to S1PRs, the clinical safety profile, future challenges, and potential strategies to address them. These insights aim to guide future research and applications of FTY720, maximizing its therapeutic potential.
Collapse
Affiliation(s)
- Juan Liu
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Lu Zhang
- Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, PR China
| | - Le Liu
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Tianfeng Wu
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Lin Wang
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Qingzhen Han
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China.
| |
Collapse
|
2
|
Kurt H, Akyol A, Son CD, Zheng C, Gado I, Meli M, Ferrandi EE, Bassanini I, Vasile F, Gurevich VV, Nebol A, Cagavi E, Morra G, Sensoy O. A small molecule enhances arrestin-3 binding to the β 2-adrenergic receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628161. [PMID: 39713392 PMCID: PMC11661165 DOI: 10.1101/2024.12.12.628161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
G protein-coupled receptor (GPCR) signaling is terminated by arrestin binding to a phosphorylated receptor. Binding propensity has been shown to be modulated by stabilizing the pre-activated state of arrestin through point mutations or C-tail truncation. Here, we hypothesize that pre-activated rotated states can be stabilized by small molecules, and this can promote binding to phosphorylation-deficient receptors, which underly a variety of human disorders. We performed virtual screening on druggable pockets identified on pre-activated conformations in Molecular Dynamics trajectories of arrestin-3, and found a compound targeting an activation switch, the back loop at the inter-domain interface. According to our model, consistent with available biochemical and structural data, the compound destabilized the ionic lock between the finger and the back loop, and enabled transition of the `gate loop` towards the pre-activated state, which stabilizes pre-activated inter-domain rotation. The predicted binding pocket is consistent with saturation-transfer difference NMR data indicating close contact between the piperazine moiety of the compound and C/finger loops. The compound increases in-cell arrestin-3 binding to phosphorylation-deficient and wild-type β2-adrenergic receptor, but not to muscarinic M2 receptor, as verified by FRET and NanoBiT. This study demonstrates that the back loop can be targeted to modulate interaction of arrestin with phosphorylation-deficient GPCRs in a receptor-specific manner.
Collapse
Affiliation(s)
- Han Kurt
- Istanbul Medipol University, Graduate School of Engineering and Natural Sciences, 34810, Istanbul, Turkey
- present address: University of Cagliari, Department of Physics, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Ali Akyol
- The Middle East Technical University, Department of Biological Sciences, Ankara 06800, Turkey
| | - Cagdas Devrim Son
- The Middle East Technical University, Department of Biological Sciences, Ankara 06800, Turkey
| | - Chen Zheng
- Vanderbilt University, Department of Pharmacology, 37232, Nashville, TN, USA
| | - Irene Gado
- University of Milano, Department of Chemistry, via Golgi 19, 20131 Milano, Italy
| | - Massimiliano Meli
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Erica Elisa Ferrandi
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Ivan Bassanini
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Francesca Vasile
- University of Milano, Department of Chemistry, via Golgi 19, 20131 Milano, Italy
| | | | - Aylin Nebol
- Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Regenerative and Restorative Medical Research Center (REMER), 34810, Istanbul, Turkey
- Istanbul Medipol University, Institute for Health Sciences, Medical Biology and Genetics Program, 34810, Istanbul, Turkey
- Istanbul Medipol University, School of Medicine, Department of Medical Biology, 34810, Istanbul, Turkey
| | - Esra Cagavi
- Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Regenerative and Restorative Medical Research Center (REMER), 34810, Istanbul, Turkey
- Istanbul Medipol University, Institute for Health Sciences, Medical Biology and Genetics Program, 34810, Istanbul, Turkey
- Istanbul Medipol University, School of Medicine, Department of Medical Biology, 34810, Istanbul, Turkey
| | - Giulia Morra
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Ozge Sensoy
- Istanbul Medipol University, School of Engineering and Natural Sciences, Department of Biomedical Engineering, 34810, Istanbul, Turkey
- Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Regenerative and Restorative Medical Research Center (REMER), 34810, Istanbul, Turkey
| |
Collapse
|
3
|
Gao PP, Li L, Chen TT, Li N, Li MQ, Zhang HJ, Chen YN, Zhang SH, Wei W, Sun WY. β-arrestin2: an emerging player and potential therapeutic target in inflammatory immune diseases. Acta Pharmacol Sin 2024:10.1038/s41401-024-01390-w. [PMID: 39349766 DOI: 10.1038/s41401-024-01390-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/01/2024] [Indexed: 03/17/2025]
Abstract
β-arrestin2, a pivotal protein within the arrestin family, is localized in the cytoplasm, plasma membrane and nucleus, and regulates G protein-coupled receptors (GPCRs) signaling. Recent evidence shows that β-arrestin2 plays a dual role in regulating GPCRs by mediating desensitization and internalization, and by acting as a scaffold for the internalization, kinase activation, and the modulation of various signaling pathways, including NF-κB, MAPK, and TGF-β pathways of non-GPCRs. Earlier studies have identified that β-arrestin2 is essential in regulating immune cell infiltration, inflammatory factor release, and inflammatory cell proliferation. Evidently, β-arrestin2 is integral to the pathological mechanisms of inflammatory immune diseases, such as inflammatory bowel disease, sepsis, asthma, rheumatoid arthritis, organ fibrosis, and tumors. Research on the modulation of β-arrestin2 offers a promising strategy for the development of pharmaceuticals targeting inflammatory immune diseases. This review meticulously describes the roles of β-arrestin2 in cells associated with inflammatory immune responses and explores its pathological relevance in various inflammatory immune diseases.
Collapse
Affiliation(s)
- Ping-Ping Gao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Ling Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Ting-Ting Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Nan Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Meng-Qi Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Hui-Juan Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Ya-Ning Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Shi-Hao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| |
Collapse
|
4
|
Gupta PK, Das A, Singh A, Rana S. Ternary model structural complex of C5a, C5aR2, and β-arrestin1. J Biomol Struct Dyn 2024; 42:7190-7206. [PMID: 37493401 DOI: 10.1080/07391102.2023.2239927] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/15/2023] [Indexed: 07/27/2023]
Abstract
Complement component fragment 5a (C5a) is one of the potent proinflammatory modulators of the complement system. C5a recruits two genomically related G protein-coupled receptors (GPCRs), like C5aR1 and C5aR2, constituting a binary complex. The C5a-C5aR1/C5aR2 binary complexes involve other transducer proteins like heterotrimeric G-proteins and β-arrestins to generate the fully active ternary complexes that trigger intracellular signaling through downstream effector molecules in tissues. In the absence of structural data, we had recently developed highly refined model structures of C5aR2 in its inactive (free), meta-active (complexed to the CT-peptide of C5a), and active (complexed to C5a) state embedded to a model palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayer. Compared to C5aR1, C5aR2 is established as a noncanonical GPCR, as it recruits and signals through β-arrestins rather than G-proteins. Notably, structural understanding of the ternary complex involving C5a-C5aR2-β-arrestin is currently unknown. The current study has attempted to fill the gap by generating a highly refined, fully active ternary model structural complex of the C5a-C5aR2-β-arrestin1 embedded in a model POPC bilayer. The computational modeling, 500 ns molecular dynamics (MD) studies, and the principal component analysis (PCA), including the molecular mechanics Poisson-Boltzmann surface area (MM PBSA) based data presented in this study, provide an experimentally testable hypothesis about C5a-C5aR2-β-arrestin1 extendable to other such ternary systems. The model ternary complex of C5a-C5aR2-β-arrestin1 will further enrich the current structural understanding related to the interaction of β-arrestins with the C5a-C5aR2 system.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pulkit Kr Gupta
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha, India
| | - Aurosikha Das
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha, India
| | - Aditi Singh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha, India
| |
Collapse
|
5
|
Nürnberg B, Beer-Hammer S, Reisinger E, Leiss V. Non-canonical G protein signaling. Pharmacol Ther 2024; 255:108589. [PMID: 38295906 DOI: 10.1016/j.pharmthera.2024.108589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024]
Abstract
The original paradigm of classical - also referred to as canonical - cellular signal transduction of heterotrimeric G proteins (G protein) is defined by a hierarchical, orthograde interaction of three players: the agonist-activated G protein-coupled receptor (GPCR), which activates the transducing G protein, that in turn regulates its intracellular effectors. This receptor-transducer-effector concept was extended by the identification of regulators and adapters such as the regulators of G protein signaling (RGS), receptor kinases like βARK, or GPCR-interacting arrestin adapters that are integrated into this canonical signaling process at different levels to enable fine-tuning. Finally, the identification of atypical signaling mechanisms of classical regulators, together with the discovery of novel modulators, added a new and fascinating dimension to the cellular G protein signal transduction. This heterogeneous group of accessory G protein modulators was coined "activators of G protein signaling" (AGS) proteins and plays distinct roles in canonical and non-canonical G protein signaling pathways. AGS proteins contribute to the control of essential cellular functions such as cell development and division, intracellular transport processes, secretion, autophagy or cell movements. As such, they are involved in numerous biological processes that are crucial for diseases, like diabetes mellitus, cancer, and stroke, which represent major health burdens. Although the identification of a large number of non-canonical G protein signaling pathways has broadened the spectrum of this cellular communication system, their underlying mechanisms, functions, and biological effects are poorly understood. In this review, we highlight and discuss atypical G protein-dependent signaling mechanisms with a focus on inhibitory G proteins (Gi) involved in canonical and non-canonical signal transduction, review recent developments and open questions, address the potential of new approaches for targeted pharmacological interventions.
Collapse
Affiliation(s)
- Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany.
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany
| | - Ellen Reisinger
- Gene Therapy for Hearing Impairment Group, Department of Otolaryngology - Head & Neck Surgery, University of Tübingen Medical Center, Elfriede-Aulhorn-Straße 5, D-72076 Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany
| |
Collapse
|
6
|
Wess J, Oteng AB, Rivera-Gonzalez O, Gurevich EV, Gurevich VV. β-Arrestins: Structure, Function, Physiology, and Pharmacological Perspectives. Pharmacol Rev 2023; 75:854-884. [PMID: 37028945 PMCID: PMC10441628 DOI: 10.1124/pharmrev.121.000302] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
The two β-arrestins, β-arrestin-1 and -2 (systematic names: arrestin-2 and -3, respectively), are multifunctional intracellular proteins that regulate the activity of a very large number of cellular signaling pathways and physiologic functions. The two proteins were discovered for their ability to disrupt signaling via G protein-coupled receptors (GPCRs) via binding to the activated receptors. However, it is now well recognized that both β-arrestins can also act as direct modulators of numerous cellular processes via either GPCR-dependent or -independent mechanisms. Recent structural, biophysical, and biochemical studies have provided novel insights into how β-arrestins bind to activated GPCRs and downstream effector proteins. Studies with β-arrestin mutant mice have identified numerous physiologic and pathophysiological processes regulated by β-arrestin-1 and/or -2. Following a short summary of recent structural studies, this review primarily focuses on β-arrestin-regulated physiologic functions, with particular focus on the central nervous system and the roles of β-arrestins in carcinogenesis and key metabolic processes including the maintenance of glucose and energy homeostasis. This review also highlights potential therapeutic implications of these studies and discusses strategies that could prove useful for targeting specific β-arrestin-regulated signaling pathways for therapeutic purposes. SIGNIFICANCE STATEMENT: The two β-arrestins, structurally closely related intracellular proteins that are evolutionarily highly conserved, have emerged as multifunctional proteins able to regulate a vast array of cellular and physiological functions. The outcome of studies with β-arrestin mutant mice and cultured cells, complemented by novel insights into β-arrestin structure and function, should pave the way for the development of novel classes of therapeutically useful drugs capable of regulating specific β-arrestin functions.
Collapse
Affiliation(s)
- Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Antwi-Boasiako Oteng
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Osvaldo Rivera-Gonzalez
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Eugenia V Gurevich
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Vsevolod V Gurevich
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| |
Collapse
|
7
|
Xie Q, Chu Y, Yuan W, Li Y, Li K, Wu X, Liu X, Xu R, Cui S, Qu X. Activation of insulin-like growth factor-1 receptor (IGF-1R) promotes growth of colorectal cancer through triggering the MEX3A-mediated degradation of RIG-I. Acta Pharm Sin B 2023; 13:2963-2975. [PMID: 37521868 PMCID: PMC10372823 DOI: 10.1016/j.apsb.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/21/2023] [Accepted: 04/01/2023] [Indexed: 08/01/2023] Open
Abstract
Insulin-like growth factor-1 receptor (IGF-1R) has been made an attractive anticancer target due to its overexpression in cancers. However, targeting it has often produced the disappointing results as the role played by cross talk with numerous downstream signalings. Here, we report a disobliging IGF-1R signaling which promotes growth of cancer through triggering the E3 ubiquitin ligase MEX3A-mediated degradation of RIG-I. The active β-arrestin-2 scaffolds this disobliging signaling to talk with MEX3A. In response to ligands, IGF-1Rβ activated the basal βarr2 into its active state by phosphorylating the interdomain domain on Tyr64 and Tyr250, opening the middle loop (Leu130‒Cys141) to the RING domain of MEX3A through the conformational changes of βarr2. The models of βarr2/IGF-1Rβ and βarr2/MEX3A could interpret the mechanism of the activated-IGF-1R in triggering degradation of RIG-I. The assay of the mutants βarr2Y64A and βarr2Y250A further confirmed the role of these two Tyr residues of the interlobe in mediating the talk between IGF-1Rβ and the RING domain of MEX3A. The truncated-βarr2 and the peptide ATQAIRIF, which mimicked the RING domain of MEX3A could prevent the formation of βarr2/IGF-1Rβ and βarr2/MEX3A complexes, thus blocking the IGF-1R-triggered RIG-I degradation. Degradation of RIG-I resulted in the suppression of the IFN-I-associated immune cells in the TME due to the blockade of the RIG-I-MAVS-IFN-I pathway. Poly(I:C) could reverse anti-PD-L1 insensitivity by recovery of RIG-I. In summary, we revealed a disobliging IGF-1R signaling by which IGF-1Rβ promoted cancer growth through triggering the MEX3A-mediated degradation of RIG-I.
Collapse
Affiliation(s)
- Qiaobo Xie
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yanyan Chu
- Ocean University of China, School of Medicine and Pharmacy, Qingdao 266075, China
| | - Wenmin Yuan
- Marine Biomedical Research Institute of Qingdao, Qingdao 266075, China
| | - Yanan Li
- Department of Toxicology and Sanitary Chemistry, Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Keqin Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xinfeng Wu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaohui Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Rui Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuxiang Cui
- Department of Toxicology and Sanitary Chemistry, Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xianjun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
8
|
Hofmann KP, Lamb TD. Rhodopsin, light-sensor of vision. Prog Retin Eye Res 2023; 93:101116. [PMID: 36273969 DOI: 10.1016/j.preteyeres.2022.101116] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/06/2022]
Abstract
The light sensor of vertebrate scotopic (low-light) vision, rhodopsin, is a G-protein-coupled receptor comprising a polypeptide chain with bound chromophore, 11-cis-retinal, that exhibits remarkable physicochemical properties. This photopigment is extremely stable in the dark, yet its chromophore isomerises upon photon absorption with 70% efficiency, enabling the activation of its G-protein, transducin, with high efficiency. Rhodopsin's photochemical and biochemical activities occur over very different time-scales: the energy of retinaldehyde's excited state is stored in <1 ps in retinal-protein interactions, but it takes milliseconds for the catalytically active state to form, and many tens of minutes for the resting state to be restored. In this review, we describe the properties of rhodopsin and its role in rod phototransduction. We first introduce rhodopsin's gross structural features, its evolution, and the basic mechanisms of its activation. We then discuss light absorption and spectral sensitivity, photoreceptor electrical responses that result from the activity of individual rhodopsin molecules, and recovery of rhodopsin and the visual system from intense bleaching exposures. We then provide a detailed examination of rhodopsin's molecular structure and function, first in its dark state, and then in the active Meta states that govern its interactions with transducin, rhodopsin kinase and arrestin. While it is clear that rhodopsin's molecular properties are exquisitely honed for phototransduction, from starlight to dawn/dusk intensity levels, our understanding of how its molecular interactions determine the properties of scotopic vision remains incomplete. We describe potential future directions of research, and outline several major problems that remain to be solved.
Collapse
Affiliation(s)
- Klaus Peter Hofmann
- Institut für Medizinische Physik und Biophysik (CC2), Charité, and, Zentrum für Biophysik und Bioinformatik, Humboldt-Unversität zu Berlin, Berlin, 10117, Germany.
| | - Trevor D Lamb
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
9
|
Seckler JM, Robinson EN, Lewis SJ, Grossfield A. Surveying nonvisual arrestins reveals allosteric interactions between functional sites. Proteins 2023; 91:99-107. [PMID: 35988049 PMCID: PMC9771995 DOI: 10.1002/prot.26413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/25/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022]
Abstract
Arrestins are important scaffolding proteins that are expressed in all vertebrate animals. They regulate cell-signaling events upon binding to active G-protein coupled receptors (GPCR) and trigger endocytosis of active GPCRs. While many of the functional sites on arrestins have been characterized, the question of how these sites interact is unanswered. We used anisotropic network modeling (ANM) together with our covariance compliment techniques to survey all the available structures of the nonvisual arrestins to map how structural changes and protein-binding affect their structural dynamics. We found that activation and clathrin binding have a marked effect on arrestin dynamics, and that these dynamics changes are localized to a small number of distant functional sites. These sites include α-helix 1, the lariat loop, nuclear localization domain, and the C-domain β-sheets on the C-loop side. Our techniques suggest that clathrin binding and/or GPCR activation of arrestin perturb the dynamics of these sites independent of structural changes.
Collapse
Affiliation(s)
- James M. Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Emily N. Robinson
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY, USA
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Alan Grossfield
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY, USA
| |
Collapse
|
10
|
Bous J, Fouillen A, Orcel H, Trapani S, Cong X, Fontanel S, Saint-Paul J, Lai-Kee-Him J, Urbach S, Sibille N, Sounier R, Granier S, Mouillac B, Bron P. Structure of the vasopressin hormone-V2 receptor-β-arrestin1 ternary complex. SCIENCE ADVANCES 2022; 8:eabo7761. [PMID: 36054364 PMCID: PMC10866553 DOI: 10.1126/sciadv.abo7761] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Arrestins interact with G protein-coupled receptors (GPCRs) to stop G protein activation and to initiate key signaling pathways. Recent structural studies shed light on the molecular mechanisms involved in GPCR-arrestin coupling, but whether this process is conserved among GPCRs is poorly understood. Here, we report the cryo-electron microscopy active structure of the wild-type arginine-vasopressin V2 receptor (V2R) in complex with β-arrestin1. It reveals an atypical position of β-arrestin1 compared to previously described GPCR-arrestin assemblies, associated with an original V2R/β-arrestin1 interface involving all receptor intracellular loops. Phosphorylated sites of the V2R carboxyl terminus are clearly identified and interact extensively with the β-arrestin1 N-lobe, in agreement with structural data obtained with chimeric or synthetic systems. Overall, these findings highlight a notable structural variability among GPCR-arrestin signaling complexes.
Collapse
Affiliation(s)
- Julien Bous
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Aurélien Fouillen
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Hélène Orcel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Stefano Trapani
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Xiaojing Cong
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Simon Fontanel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Julie Saint-Paul
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Joséphine Lai-Kee-Him
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Serge Urbach
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Nathalie Sibille
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Rémy Sounier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Bernard Mouillac
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier Cedex 5, France
| | - Patrick Bron
- CBS (Centre de Biologie Structurale), Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
11
|
Aamna B, Kumar Dan A, Sahu R, Behera SK, Parida S. Deciphering the signaling mechanisms of β-arrestin1 and β-arrestin2 in regulation of cancer cell cycle and metastasis. J Cell Physiol 2022; 237:3717-3733. [PMID: 35908197 DOI: 10.1002/jcp.30847] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/22/2022] [Accepted: 07/18/2022] [Indexed: 11/05/2022]
Abstract
β-Arrestins are ubiquitously expressed intracellular proteins with many functions which interact directly and indirectly with a wide number of cellular partners and mediate downstream signaling. Originally, β-arrestins were identified for their contribution to GPCR desensitization to agonist-mediated activation, followed by receptor endocytosis and ubiquitylation. However, current investigations have now recognized that in addition to GPCR arresting (hence the name arrestin). β-Arrestins are adaptor proteins that control the recruitment, activation, and scaffolding of numerous cytoplasmic signaling complexes and assist in G-protein receptor signaling, thus bringing them into close proximity. They have participated in various cellular processes such as cell proliferation, migration, apoptosis, and transcription via canonical and noncanonical pathways. Despite their significant recognition in several physiological processes, these activities are also involved in the onset and progression of various cancers. This review delivers a concise overview of the role of β-arrestins with a primary emphasis on the signaling processes which underlie the mechanism of β-arrestins in the onset of cancer. Understanding these processes has important implications for understanding the therapeutic intervention and treatment of cancer in the future.
Collapse
Affiliation(s)
- Bari Aamna
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed to be University), Bhubaneswar, Odisha, India
| | - Aritra Kumar Dan
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed to be University), Bhubaneswar, Odisha, India
| | - Raghaba Sahu
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Sagarika Parida
- Department of Botany, Centurion University of Technology and Management, Odisha, India
| |
Collapse
|
12
|
Asher WB, Terry DS, Gregorio GGA, Kahsai AW, Borgia A, Xie B, Modak A, Zhu Y, Jang W, Govindaraju A, Huang LY, Inoue A, Lambert NA, Gurevich VV, Shi L, Lefkowitz RJ, Blanchard SC, Javitch JA. GPCR-mediated β-arrestin activation deconvoluted with single-molecule precision. Cell 2022; 185:1661-1675.e16. [PMID: 35483373 PMCID: PMC9191627 DOI: 10.1016/j.cell.2022.03.042] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/11/2022] [Accepted: 03/29/2022] [Indexed: 01/14/2023]
Abstract
β-arrestins bind G protein-coupled receptors to terminate G protein signaling and to facilitate other downstream signaling pathways. Using single-molecule fluorescence resonance energy transfer imaging, we show that β-arrestin is strongly autoinhibited in its basal state. Its engagement with a phosphopeptide mimicking phosphorylated receptor tail efficiently releases the β-arrestin tail from its N domain to assume distinct conformations. Unexpectedly, we find that β-arrestin binding to phosphorylated receptor, with a phosphorylation barcode identical to the isolated phosphopeptide, is highly inefficient and that agonist-promoted receptor activation is required for β-arrestin activation, consistent with the release of a sequestered receptor C tail. These findings, together with focused cellular investigations, reveal that agonism and receptor C-tail release are specific determinants of the rate and efficiency of β-arrestin activation by phosphorylated receptor. We infer that receptor phosphorylation patterns, in combination with receptor agonism, synergistically establish the strength and specificity with which diverse, downstream β-arrestin-mediated events are directed.
Collapse
Affiliation(s)
- Wesley B Asher
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Daniel S Terry
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - G Glenn A Gregorio
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Alem W Kahsai
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Alessandro Borgia
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Bing Xie
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Arnab Modak
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ying Zhu
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Wonjo Jang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Alekhya Govindaraju
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Li-Yin Huang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | | | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Robert J Lefkowitz
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Scott C Blanchard
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Jonathan A Javitch
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
13
|
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
|
14
|
Plouffe B, Karamitri A, Flock T, Gallion JM, Houston S, Daly CA, Bonnefond A, Guillaume JL, Le Gouill C, Froguel P, Lichtarge O, Deupi X, Jockers R, Bouvier M. Structural Elements Directing G Proteins and β-Arrestin Interactions with the Human Melatonin Type 2 Receptor Revealed by Natural Variants. ACS Pharmacol Transl Sci 2022; 5:89-101. [PMID: 35846981 PMCID: PMC9281605 DOI: 10.1021/acsptsci.1c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
G protein-coupled receptors (GPCRs) can engage distinct subsets of signaling pathways, but the structural determinants of this functional selectivity remain elusive. The naturally occurring genetic variants of GPCRs, selectively affecting different pathways, offer an opportunity to explore this phenomenon. We previously identified 40 coding variants of the MTNR1B gene encoding the melatonin MT2 receptor (MT2). These mutations differently impact the β-arrestin 2 recruitment, ERK activation, cAMP production, and Gαi1 and Gαz activation. In this study, we combined functional clustering and structural modeling to delineate the molecular features controlling the MT2 functional selectivity. Using non-negative matrix factorization, we analyzed the signaling signatures of the 40 MT2 variants yielding eight clusters defined by unique signaling features and localized in distinct domains of MT2. Using computational homology modeling, we describe how specific mutations can selectively affect the subsets of signaling pathways and offer a proof of principle that natural variants can be used to explore and understand the GPCR functional selectivity.
Collapse
Affiliation(s)
- Bianca Plouffe
- Department
of Biochemistry and Molecular Medicine, Université de Montréal, H3T 1J4 Montréal, Québec, Canada,Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada,The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Angeliki Karamitri
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Tilman Flock
- Laboratory
of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland,Department
of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Jonathan M. Gallion
- Program
in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, 77030 Houston, Texas, United States
| | - Shane Houston
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Carole A. Daly
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Amélie Bonnefond
- Université
de Lille, INSERM/CNRS UMR 1283/8199—EGID, Institut Pasteur
de Lille, CHU de Lille, 59045 Lille, France
| | - Jean-Luc Guillaume
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Christian Le Gouill
- Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Phillipe Froguel
- Université
de Lille, INSERM/CNRS UMR 1283/8199—EGID, Institut Pasteur
de Lille, CHU de Lille, 59045 Lille, France
| | - Olivier Lichtarge
- Program
in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, 77030 Houston, Texas, United States,Department
of Molecular and Human Genetics, Baylor
College of Medicine, 77030 Houston, Texas, United States
| | - Xavier Deupi
- Laboratory
of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland,Condensed
Matter Theory Group, Division of Scientific Computing, Theory, and
Data, Paul Scherrer Institute, 5232 Villigen, Switzerland,. Phone: +41-563103337
| | - Ralf Jockers
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France,. Phone: +33-140516434
| | - Michel Bouvier
- Department
of Biochemistry and Molecular Medicine, Université de Montréal, H3T 1J4 Montréal, Québec, Canada,Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada,. Phone: 1-514-343-6319
| |
Collapse
|
15
|
Kim K, Han Y, Duan L, Chung KY. Scaffolding of Mitogen-Activated Protein Kinase Signaling by β-Arrestins. Int J Mol Sci 2022; 23:ijms23021000. [PMID: 35055186 PMCID: PMC8778048 DOI: 10.3390/ijms23021000] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 12/19/2022] Open
Abstract
β-arrestins were initially identified to desensitize and internalize G-protein-coupled receptors (GPCRs). Receptor-bound β-arrestins also initiate a second wave of signaling by scaffolding mitogen-activated protein kinase (MAPK) signaling components, MAPK kinase kinase, MAPK kinase, and MAPK. In particular, β-arrestins facilitate ERK1/2 or JNK3 activation by scaffolding signal cascade components such as ERK1/2-MEK1-cRaf or JNK3-MKK4/7-ASK1. Understanding the precise molecular and structural mechanisms of β-arrestin-mediated MAPK scaffolding assembly would deepen our understanding of GPCR-mediated MAPK activation and provide clues for the selective regulation of the MAPK signaling cascade for therapeutic purposes. Over the last decade, numerous research groups have attempted to understand the molecular and structural mechanisms of β-arrestin-mediated MAPK scaffolding assembly. Although not providing the complete mechanism, these efforts suggest potential binding interfaces between β-arrestins and MAPK signaling components and the mechanism for MAPK signal amplification by β-arrestin-mediated scaffolding. This review summarizes recent developments of cellular and molecular works on the scaffolding mechanism of β-arrestin for MAPK signaling cascade.
Collapse
|
16
|
Flöser A, Becker K, Kostenis E, König G, Krasel C, Kolb P, Bünemann M. Disentangling bias between G q, GRK2, and arrestin3 recruitment to the M 3 muscarinic acetylcholine receptor. eLife 2021; 10:58442. [PMID: 34851820 PMCID: PMC8635974 DOI: 10.7554/elife.58442] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptors (GPCRs) transmit extracellular signals to the inside by activation of intracellular effector proteins. Different agonists can promote differential receptor-induced signaling responses – termed bias – potentially by eliciting different levels of recruitment of effector proteins. As activation and recruitment of effector proteins might influence each other, thorough analysis of bias is difficult. Here, we compared the efficacy of seven agonists to induce G protein, G protein-coupled receptor kinase 2 (GRK2), as well as arrestin3 binding to the muscarinic acetylcholine receptor M3 by utilizing FRET-based assays. In order to avoid interference between these interactions, we studied GRK2 binding in the presence of inhibitors of Gi and Gq proteins and analyzed arrestin3 binding to prestimulated M3 receptors to avoid differences in receptor phosphorylation influencing arrestin recruitment. We measured substantial differences in the agonist efficacies to induce M3R-arrestin3 versus M3R-GRK2 interaction. However, the rank order of the agonists for G protein- and GRK2-M3R interaction was the same, suggesting that G protein and GRK2 binding to M3R requires similar receptor conformations, whereas requirements for arrestin3 binding to M3R are distinct.
Collapse
Affiliation(s)
- Anja Flöser
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany.,Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Katharina Becker
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Gabriele König
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Cornelius Krasel
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
17
|
El Fessikh M, Belghiti H, Elkarhat Z, Guerinech H, Dakka N, El Baghdadi J. Identification of p.Met215Ile mutation of the MC4R gene in a Moroccan woman with obesity. Clin Case Rep 2021; 9:e05059. [PMID: 34815872 PMCID: PMC8593808 DOI: 10.1002/ccr3.5059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 11/19/2022] Open
Abstract
Screening the MC4R gene showed one rare mutation p.Met215Ile in a Moroccan patient with morbid obesity, which leads to a change in the protein structure. The analysis of MC4R variants may be useful for future therapeutic approaches.
Collapse
Affiliation(s)
- Meriem El Fessikh
- Genetics UnitMilitary Hospital Mohammed VRabatMorocco
- Laboratory of Human Pathologies Biology, and Genomic Center of Human PathologiesFaculty of SciencesMohammed V University in RabatRabatMorocco
| | - Hakim Belghiti
- Clinical Nutrition Unit, Hygiene and Collectivity Medicine WardMilitary Hospital Mohammed VRabatMorocco
| | - Zouhair Elkarhat
- Laboratory of Genomics and Human GeneticsPasteur Institute of CasablancaCasablancaMorocco
| | - Hassania Guerinech
- Clinical Nutrition Unit, Hygiene and Collectivity Medicine WardMilitary Hospital Mohammed VRabatMorocco
| | - Nadia Dakka
- Laboratory of Human Pathologies Biology, and Genomic Center of Human PathologiesFaculty of SciencesMohammed V University in RabatRabatMorocco
| | | |
Collapse
|
18
|
White AD, Peña KA, Clark LJ, Maria CS, Liu S, Jean-Alphonse FG, Lee JY, Lei S, Cheng Z, Tu CL, Fang F, Szeto N, Gardella TJ, Xiao K, Gellman SH, Bahar I, Sutkeviciute I, Chang W, Vilardaga JP. Spatial bias in cAMP generation determines biological responses to PTH type 1 receptor activation. Sci Signal 2021; 14:eabc5944. [PMID: 34609896 PMCID: PMC8682804 DOI: 10.1126/scisignal.abc5944] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Alex D White
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Graduate Program in Molecular Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Karina A Peña
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lisa J Clark
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Graduate Program in Molecular Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Christian Santa Maria
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, and University of California, San Francisco, CA 94158, USA
| | - Shi Liu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Frédéric G Jean-Alphonse
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ji Young Lee
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Saifei Lei
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zhiqiang Cheng
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, and University of California, San Francisco, CA 94158, USA
| | - Chia-Ling Tu
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, and University of California, San Francisco, CA 94158, USA
| | - Fei Fang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nicholas Szeto
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, and University of California, San Francisco, CA 94158, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kunhong Xiao
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Samuel H Gellman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Ieva Sutkeviciute
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wenhan Chang
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, and University of California, San Francisco, CA 94158, USA
| | - Jean-Pierre Vilardaga
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
19
|
Qu C, Park JY, Yun MW, He QT, Yang F, Kim K, Ham D, Li RR, Iverson TM, Gurevich VV, Sun JP, Chung KY. Scaffolding mechanism of arrestin-2 in the cRaf/MEK1/ERK signaling cascade. Proc Natl Acad Sci U S A 2021; 118:e2026491118. [PMID: 34507982 PMCID: PMC8449410 DOI: 10.1073/pnas.2026491118] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 01/14/2023] Open
Abstract
Arrestins were initially identified for their role in homologous desensitization and internalization of G protein-coupled receptors. Receptor-bound arrestins also initiate signaling by interacting with other signaling proteins. Arrestins scaffold MAPK signaling cascades, MAPK kinase kinase (MAP3K), MAPK kinase (MAP2K), and MAPK. In particular, arrestins facilitate ERK1/2 activation by scaffolding ERK1/2 (MAPK), MEK1 (MAP2K), and Raf (MAPK3). However, the structural mechanism underlying this scaffolding remains unknown. Here, we investigated the mechanism of arrestin-2 scaffolding of cRaf, MEK1, and ERK2 using hydrogen/deuterium exchange-mass spectrometry, tryptophan-induced bimane fluorescence quenching, and NMR. We found that basal and active arrestin-2 interacted with cRaf, while only active arrestin-2 interacted with MEK1 and ERK2. The ATP binding status of MEK1 or ERK2 affected arrestin-2 binding; ATP-bound MEK1 interacted with arrestin-2, whereas only empty ERK2 bound arrestin-2. Analysis of the binding interfaces suggested that the relative positions of cRaf, MEK1, and ERK2 on arrestin-2 likely facilitate sequential phosphorylation in the signal transduction cascade.
Collapse
Affiliation(s)
- Changxiu Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, China
| | - Ji Young Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Min Woo Yun
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Qing-Tao He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, China
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Kiae Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Donghee Ham
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Rui-Rui Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, China
| | - T M Iverson
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | | | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, China;
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| |
Collapse
|
20
|
Alhosaini K, Azhar A, Alonazi A, Al-Zoghaibi F. GPCRs: The most promiscuous druggable receptor of the mankind. Saudi Pharm J 2021; 29:539-551. [PMID: 34194261 PMCID: PMC8233523 DOI: 10.1016/j.jsps.2021.04.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
All physiological events in living organisms originated as specific chemical/biochemical signals on the cell surface and transmitted into the cytoplasm. This signal is translated within milliseconds-hours to a specific and unique order required to maintain optimum performance and homeostasis of living organisms. Examples of daily biological functions include neuronal communication and neurotransmission in the process of learning and memory, secretion (hormones, sweat, and saliva), muscle contraction, cellular growth, differentiation and migration during wound healing, and immunity to fight infections. Among the different transducers for such life-dependent signals is the large family of G protein-coupled receptors (GPCRs). GPCRs constitute roughly 800 genes, corresponding to 2% of the human genome. While GPCRs control a plethora of pathophysiological disorders, only approximately one-third of GPCR families have been deorphanized and characterized. Recent drug data show that around 40% of the recommended drugs available in the market target mainly GPCRs. In this review, we presented how such system signals, either through G protein or via other players, independent of G protein, function within the biological system. We also discussed drugs in the market or clinical trials targeting mainly GPCRs in various diseases, including cancer.
Collapse
Key Words
- AC, Adenylyl Cyclase
- Arrestin
- CCR, Chemokine Receptor
- COX, Cyclooxygenase
- DAG, Diacylglycerol
- Drugs
- ERK, Extracellular signal-Regulated Kinase
- G proteins
- GIP, Gastric Inhibitory Peptide
- GLP1R, Glucagon-Like Peptide-1 Receptor
- GPCR
- GRKs
- GRKs, G protein-coupled Receptor Kinases
- Heterodimerization
- IP3, Inositol 1,4,5-triphosphate
- MAPK, Mitogen-Activated Protein Kinase
- NMDA, N-Methyl D-Aspartate
- Nbs, Nanobodies
- PAR-1, Protease Activated Receptor 1
- PIP2, Phosphatidylinositol-4,5-bisphosphate
- PKA, Protein Kinase A
- Signaling
- cAMP, cyclic AMP
Collapse
Affiliation(s)
- Khaled Alhosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451, Saudi Arabia
| | - Asim Azhar
- Interdisciplinary Biotechnology Unit, AMU Aligarh, UP, India
| | - Asma Alonazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451, Saudi Arabia
| | - F Al-Zoghaibi
- Molecular BioMedicine Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, MBC:03, Riyadh 11211, Saudi Arabia
| |
Collapse
|
21
|
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
|
22
|
Fanelli F, Felline A, Marigo V. Structural aspects of rod opsin and their implication in genetic diseases. Pflugers Arch 2021; 473:1339-1359. [PMID: 33728518 DOI: 10.1007/s00424-021-02546-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 01/04/2023]
Abstract
Vision in dim-light conditions is triggered by photoactivation of rhodopsin, the visual pigment of rod photoreceptor cells. Rhodopsin is made of a protein, the G protein coupled receptor (GPCR) opsin, and the chromophore 11-cis-retinal. Vertebrate rod opsin is the GPCR best characterized at the atomic level of detail. Since the release of the first crystal structure 20 years ago, a huge number of structures have been released that, in combination with valuable spectroscopic determinations, unveiled most aspects of the photobleaching process. A number of spontaneous mutations of rod opsin have been found linked to vision-impairing diseases like autosomal dominant or autosomal recessive retinitis pigmentosa (adRP or arRP, respectively) and autosomal congenital stationary night blindness (adCSNB). While adCSNB is mainly caused by constitutive activation of rod opsin, RP shows more variegate determinants affecting different aspects of rod opsin function. The vast majority of missense rod opsin mutations affects folding and trafficking and is linked to adRP, an incurable disease that awaits light on its molecular structure determinants. This review article summarizes all major structural information available on vertebrate rod opsin conformational states and the insights gained so far into the structural determinants of adCSNB and adRP linked to rod opsin mutations. Strategies to design small chaperones with therapeutic potential for selected adRP rod opsin mutants will be discussed as well.
Collapse
Affiliation(s)
- Francesca Fanelli
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125, Modena, Italy. .,Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, via Campi 287, Modena, 41125, Italy.
| | - Angelo Felline
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125, Modena, Italy
| | - Valeria Marigo
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, via Campi 287, Modena, 41125, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 287, 41125, Modena, Italy
| |
Collapse
|
23
|
Ren R, Pang B, Han Y, Li Y. A Glimpse of the Structural Biology of the Metabolism of Sphingosine-1-Phosphate. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2021; 4:2515256421995601. [PMID: 37366379 PMCID: PMC10243590 DOI: 10.1177/2515256421995601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 06/28/2023]
Abstract
As a key sphingolipid metabolite, sphingosine-1-phosphate (S1P) plays crucial roles in vascular and immune systems. It regulates angiogenesis, vascular integrity and homeostasis, allergic responses, and lymphocyte trafficking. S1P is interconverted with sphingosine, which is also derived from the deacylation of ceramide. S1P levels and the ratio to ceramide in cells are tightly regulated by its metabolic pathways. Abnormal S1P production causes the occurrence and progression of numerous severe diseases, such as metabolic syndrome, cancers, autoimmune disorders such as multiple sclerosis, and kidney and cardiovascular diseases. In recent years, huge advances on the structure of S1P metabolic pathways have been accomplished. In this review, we have got a glimpse of S1P metabolism through structural and biochemical studies of: sphingosine kinases, S1P transporters and S1P receptors, and the development of therapeutics targeting S1P signaling. The progress we summarize here could provide fresh perspectives to further the exploration of S1P functions and facilitate the development of therapeutic molecules targeting S1P signaling with improved specificity and therapeutic effects.
Collapse
Affiliation(s)
- Ruobing Ren
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Bin Pang
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Yufei Han
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Yihao Li
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| |
Collapse
|
24
|
Camacho E, Marie N, Dupas Q, Martel C, Nowoczyn M, Elie N, Rochais C, Töth G, Allouche S. Impact of T161, Y318 and S363 alanine mutations on regulation of the human delta-opioid receptor (hDOPr) induced by peptidic and alkaloid agonists. Neuropharmacology 2020; 179:108286. [PMID: 32841607 DOI: 10.1016/j.neuropharm.2020.108286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 11/16/2022]
Abstract
Previously, we showed a differential regulation of the human delta-opioid receptor (hDOPr) by etorphine and [D-Pen2, D-Pen5] enkephalin (DPDPE). To understand the molecular basis of such differences, we introduced 3 alanine mutations at the residues T161. Y318 and S363. Both wild type (WT) and hDOPr mutants were expressed in HEK cells containing endogenous arrestins or CFP-tagged arrestin 3, then desensitization, internalization, recycling and phosphorylation were studied. In a context of endogenous arrestin expression, a major difference in DOPr desensitization was observed between agonists that was modified with the T161A mutation upon etorphine and with the S363A substitution upon DPDPE exposure. While both agonists induced a major receptor internalization, T161A and S363A impaired DOPr sequestration only for etorphine. However, similar level of S363 phosphorylation was measured between agonists. When CFP-tagged arrestin 3 was over-expressed, a similar profile of desensitization was measured for both agonists. In this context, all the 3 alanine mutations decreased etorphine-induced receptor desensitization. Using FRET, we showed similar interactions between WT hDOPr and arrestin 3 under DPDPE and etorphine stimulation which were delayed by both the Y318A and the S363A substitutions for etorphine. Finally, hDOPr recycling was qualitatively evaluated by microscopy and showed neither arrestin 3/hDOPr colocalization nor major impact of alanine mutations except for the S363A which impaired internalization and recycling for etorphine. The T161, Y318 and S363 residues of hDOPr could underlie the differential regulation promoted by DPDPE and etorphine.
Collapse
Affiliation(s)
- Elise Camacho
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Nicolas Marie
- Centre National de Recherche Scientifique, Unité Mixte de Recherche 8206, Institut National de La Santé et de La Recherche Médicale. U705, Université Paris Descartes, Laboratoire de Neuropsychopharmacologie des Addictions, 4 Avenue de L'observatoire, 75006, Paris, France
| | - Quentin Dupas
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Caroline Martel
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Marie Nowoczyn
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France
| | - Nicolas Elie
- Plateau D'Histo-Imagerie Quantitative, CmaBio(3), SF 4206 ICORE, Normandie Univ, Caen, France
| | - Christophe Rochais
- Centre D'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, 14000, Caen, France
| | - Geza Töth
- Biological Research Centre, Institute of Biochemistry, Hungarian Academy of Sciences, Szeged, Hungary
| | - Stéphane Allouche
- Laboratoire de Signalisation, électrophysiologie et Imagerie des Lésions D'ischémie-reperfusion Myocardique, Normandie Univ, UNICAEN, Caen, France.
| |
Collapse
|
25
|
Abstract
Heart failure is a major source of morbidity and mortality, driven, in part, by maladaptive sympathetic hyperactivity in response to poor cardiac output. Current therapies target β-adrenergic and angiotensin II G protein-coupled receptors to reduce adverse cardiac remodeling and improve clinical outcomes; however, there is a pressing need for new therapeutic approaches to preserve cardiac function. β-arrestin is a multifunctional protein which has come under analysis in recent years as a key player in G protein-coupled receptor signal transduction and a potential therapeutic target in heart failure. β-arrestin attenuates β-adrenergic and angiotensin II receptor signaling to limit the deleterious response to excessive sympathetic stimulation while simultaneously transactivating cardioprotective signaling cascades that preserve cardiac structure and function in response to injury. β-arrestin signaling may provide unique advantages compared to classic heart failure treatment approaches, but a number of challenges currently limit clinical applications. In this review, we discuss the role and functions of β-arrestin and the current attempts to develop G protein-coupled receptor agonists biased towards β-arrestin activation. Furthermore, we examine the functional diversity of cardiac β-arrestin isotypes to explore key considerations in the promise of β-arrestin as a pharmacotherapeutic target in heart failure.
Collapse
|
26
|
Zhuo Y, Gurevich VV, Vishnivetskiy SA, Klug CS, Marchese A. A non-GPCR-binding partner interacts with a novel surface on β-arrestin1 to mediate GPCR signaling. J Biol Chem 2020; 295:14111-14124. [PMID: 32753481 PMCID: PMC7549033 DOI: 10.1074/jbc.ra120.015074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/29/2020] [Indexed: 12/30/2022] Open
Abstract
The multifaceted adaptor protein β-arr1 (β-arrestin1) promotes activation of focal adhesion kinase (FAK) by the chemokine receptor CXCR4, facilitating chemotaxis. This function of β-arr1 requires the assistance of the adaptor protein STAM1 (signal-transducing adaptor molecule 1) because disruption of the interaction between STAM1 and β-arr1 reduces CXCR4-mediated activation of FAK and chemotaxis. To begin to understand the mechanism by which β-arr1 together with STAM1 activates FAK, we used site-directed spin-labeling EPR spectroscopy-based studies coupled with bioluminescence resonance energy transfer-based cellular studies to show that STAM1 is recruited to activated β-arr1 by binding to a novel surface on β-arr1 at the base of the finger loop, at a site that is distinct from the receptor-binding site. Expression of a STAM1-deficient binding β-arr1 mutant that is still able to bind to CXCR4 significantly reduced CXCL12-induced activation of FAK but had no impact on ERK-1/2 activation. We provide evidence of a novel surface at the base of the finger loop that dictates non-GPCR interactions specifying β-arrestin-dependent signaling by a GPCR. This surface might represent a previously unidentified switch region that engages with effector molecules to drive β-arrestin signaling.
Collapse
Affiliation(s)
- Ya Zhuo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Candice S Klug
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
27
|
Felline A, Seeber M, Fanelli F. webPSN v2.0: a webserver to infer fingerprints of structural communication in biomacromolecules. Nucleic Acids Res 2020; 48:W94-W103. [PMID: 32427333 PMCID: PMC7319592 DOI: 10.1093/nar/gkaa397] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/21/2020] [Accepted: 05/17/2020] [Indexed: 12/25/2022] Open
Abstract
A mixed Protein Structure Network (PSN) and Elastic Network Model-Normal Mode Analysis (ENM-NMA)-based strategy (i.e. PSN-ENM) was developed to investigate structural communication in bio-macromolecules. Protein Structure Graphs (PSGs) are computed on a single structure, whereas information on system dynamics is supplied by ENM-NMA. The approach was implemented in a webserver (webPSN), which was significantly updated herein. The webserver now handles both proteins and nucleic acids and relies on an internal upgradable database of network parameters for ions and small molecules in all PDB structures. Apart from the radical restyle of the server and some changes in the calculation setup, other major novelties concern the possibility to: a) compute the differences in nodes, links, and communication pathways between two structures (i.e. network difference) and b) infer links, hubs, communities, and metapaths from consensus networks computed on a number of structures. These new features are useful to identify commonalties and differences between two different functional states of the same system or structural-communication signatures in homologous or analogous systems. The output analysis relies on 3D-representations, interactive tables and graphs, also available for download. Speed and accuracy make this server suitable to comparatively investigate structural communication in large sets of bio-macromolecular systems. URL: http://webpsn.hpc.unimore.it.
Collapse
Affiliation(s)
- Angelo Felline
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Michele Seeber
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Francesca Fanelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy.,Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena 41125, Italy
| |
Collapse
|
28
|
Many faces of the GPCR-arrestin interaction. Arch Pharm Res 2020; 43:890-899. [DOI: 10.1007/s12272-020-01263-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/11/2020] [Indexed: 01/14/2023]
|
29
|
Min K, Yoon HJ, Park JY, Baidya M, Dwivedi-Agnihotri H, Maharana J, Chaturvedi M, Chung KY, Shukla AK, Lee HH. Crystal Structure of β-Arrestin 2 in Complex with CXCR7 Phosphopeptide. Structure 2020; 28:1014-1023.e4. [PMID: 32579945 DOI: 10.1016/j.str.2020.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/02/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022]
Abstract
β-Arrestins (βarrs) critically regulate G-protein-coupled receptor (GPCR) signaling and trafficking. βarrs have two isoforms, βarr1 and βarr2. Receptor phosphorylation is a key determinant for the binding of βarrs, and understanding the intricate details of receptor-βarr interaction is the next frontier in GPCR structural biology. The high-resolution structure of active βarr1 in complex with a phosphopeptide derived from GPCR has been revealed, but that of βarr2 remains elusive. Here, we present a 2.3-Å crystal structure of βarr2 in complex with a phosphopeptide (C7pp) derived from the carboxyl terminus of CXCR7. The structural analysis of C7pp-bound βarr2 reveals key differences from the previously determined active conformation of βarr1. One of the key differences is that C7pp-bound βarr2 shows a relatively small inter-domain rotation. Antibody-fragment-based conformational sensor and hydrogen/deuterium exchange experiments further corroborated the structural features of βarr2 and suggested that βarr2 adopts a range of inter-domain rotations.
Collapse
Affiliation(s)
- Kyungjin Min
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hye-Jin Yoon
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Young Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Mithu Baidya
- Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | | | - Jagannath Maharana
- Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Madhu Chaturvedi
- Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Arun K Shukla
- Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India.
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
30
|
Bellucci L, Felline A, Fanelli F. Dynamics and structural communication in the ternary complex of fully phosphorylated V2 vasopressin receptor, vasopressin, and β-arrestin 1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183355. [PMID: 32413442 DOI: 10.1016/j.bbamem.2020.183355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are critically regulated by arrestins, which not only desensitize G-protein signaling but also initiate a G protein-independent wave of signaling. The information from structure determination was herein exploited to build a structural model of the ternary complex, comprising fully phosphorylated V2 vasopressin receptor (V2R), the agonist arginine vasopressin (AVP), and β-arrestin 1 (β-arr1). Molecular simulations served to explore dynamics and structural communication in the ternary complex. Flexibility and mechanical profiles reflect fold of V2R and β-arr1. Highly conserved amino acids tend to behave as hubs in the structure network and contribute the most to the mechanical rigidity of V2R seven-helix bundle and of β-arr1. Two structurally and dynamically distinct receptor-arrestin interfaces assist the twist of the N- and C-terminal domains (ND and CD, respectively) of β-arr1 with respect to each other, which is linked to arrestin activation. While motion of the ND is essentially assisted by the fully phosphorylated C-tail of V2R (V2RCt), that of CD is assisted by the second and third intracellular loops and the cytosolic extensions of helices 5 and 6. In the presence of the receptor, the β-arr1 inter-domain twist angle correlates with the modes describing the essential subspace of the ternary complex. β-arr1 motions are also influenced by the anchoring to the membrane of the C-edge-loops in the β-arr1-CD. Overall fluctuations reveal a coupling between motions of the agonist binding site and of β-arr1-ND, which are in allosteric communication between each other. Mechanical rigidity points, often acting as hubs in the structure network and distributed along the main axis of the receptor helix bundle, contribute to establish a preferential communication pathway between agonist ligand and the ND of arrestin. Such communication, mediated by highly conserved amino acids, involves also the first amino acid in the arrestin C-tail, which is highly dynamic and is involved in clathrin-mediated GPCR internalization.
Collapse
Affiliation(s)
- Luca Bellucci
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy; NEST, Istituto Nanoscienze-CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Angelo Felline
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy
| | - Francesca Fanelli
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy; Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, via Campi 287, 41125 Modena, Italy.
| |
Collapse
|
31
|
Huang W, Masureel M, Qu Q, Janetzko J, Inoue A, Kato HE, Robertson MJ, Nguyen KC, Glenn JS, Skiniotis G, Kobilka BK. Structure of the neurotensin receptor 1 in complex with β-arrestin 1. Nature 2020; 579:303-308. [PMID: 31945771 PMCID: PMC7100716 DOI: 10.1038/s41586-020-1953-1] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/08/2020] [Indexed: 01/14/2023]
Abstract
Arrestin proteins bind to active, phosphorylated G-protein-coupled receptors (GPCRs), thereby preventing G-protein coupling, triggering receptor internalization and affecting various downstream signalling pathways1,2. Although there is a wealth of structural information detailing the interactions between GPCRs and G proteins, less is known about how arrestins engage GPCRs. Here we report a cryo-electron microscopy structure of full-length human neurotensin receptor 1 (NTSR1) in complex with truncated human β-arrestin 1 (βarr1(ΔCT)). We find that phosphorylation of NTSR1 is critical for the formation of a stable complex with βarr1(ΔCT), and identify phosphorylated sites in both the third intracellular loop and the C terminus that may promote this interaction. In addition, we observe a phosphatidylinositol-4,5-bisphosphate molecule forming a bridge between the membrane side of NTSR1 transmembrane segments 1 and 4 and the C-lobe of arrestin. Compared with a structure of a rhodopsin-arrestin-1 complex, in our structure arrestin is rotated by approximately 85° relative to the receptor. These findings highlight both conserved aspects and plasticity among arrestin-receptor interactions.
Collapse
Affiliation(s)
- Weijiao Huang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthieu Masureel
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Qianhui Qu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - John Janetzko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hideaki E Kato
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Komaba Institute for Science, The University of Tokyo, Tokyo, Japan
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Khanh C Nguyen
- Departments of Medicine and Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | - Jeffrey S Glenn
- Departments of Medicine and Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Photon Science, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA.
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
32
|
Staus DP, Hu H, Robertson MJ, Kleinhenz ALW, Wingler LM, Capel WD, Latorraca NR, Lefkowitz RJ, Skiniotis G. Structure of the M2 muscarinic receptor-β-arrestin complex in a lipid nanodisc. Nature 2020; 579:297-302. [PMID: 31945772 PMCID: PMC7367492 DOI: 10.1038/s41586-020-1954-0] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023]
Abstract
Following agonist activation, G protein-coupled receptors (GPCRs) recruit β-arrestin, which desensitizes heterotrimeric G protein signaling and promotes receptor endocytosis1. Additionally, β-arrestin directly regulates many cell signaling pathways that can induce cellular responses distinct from that of G proteins2. Here we present a cryo-electron microscopy (cryoEM) structure of β-arrestin1 (βarr1) in complex with muscarinic acetylcholine-2-receptor (M2R) reconstituted in lipid nanodiscs. The M2R-βarr1 structure shows a multimodal network of flexible interactions, including binding of the βarr1 N-domain to phosphorylated receptor residues and βarr1 finger loop insertion into the M2R seven-transmembrane bundle, which adopts a conformation similar to that in the M2R-heterotrimeric Go protein structure3. Moreover, the cryoEM map reveals that the βarr1 C-domain edge engages the lipid bilayer. Through atomistic simulations, biophysical, biochemical, and cellular assays, we show that the C-edge is critical for stable complex formation, βarr1 recruitment, receptor internalization, and desensitization of G protein activation. Taken together, these data suggest the cooperative interactions of β-arrestin with both the receptor and phospholipid bilayer contribute to its functional versatility.
Collapse
Affiliation(s)
- Dean P Staus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
| | - Hongli Hu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,School of Life and Health Sciences, Kobilka Institute of Innovative Drug Discovery, The Chinese University of Hong Kong, Shenzhen, China
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alissa L W Kleinhenz
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA.,School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Laura M Wingler
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
| | - William D Capel
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Naomi R Latorraca
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Computer Science, Stanford University, Stanford, CA, USA.,Biophysics Program, Stanford University, Stanford, CA, USA
| | - Robert J Lefkowitz
- Department of Medicine, Duke University Medical Center, Durham, NC, USA. .,Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA. .,Department of Biochemistry, Duke University Medical Center, Durham, NC, USA.
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
33
|
Zou Y, Ewalt J, Ng HL. Recent Insights from Molecular Dynamics Simulations for G Protein-Coupled Receptor Drug Discovery. Int J Mol Sci 2019; 20:E4237. [PMID: 31470676 PMCID: PMC6747122 DOI: 10.3390/ijms20174237] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are critical drug targets. GPCRs convey signals from the extracellular to the intracellular environment through G proteins. Some ligands that bind to GPCRs activate different downstream signaling pathways. G protein activation, or -arrestin biased signaling, involves ligands binding to receptors and stabilizing conformations that trigger a specific pathway. -arrestin biased signaling has become a hot target for structure-based drug discovery. However, challenges include that there are few crystal structures available in the Protein Data Bank and that GPCRs are highly dynamic. Hence, molecular dynamics (MD) simulations are especially valuable for obtaining detailed mechanistic information, including identification of allosteric sites and understanding modulators' interactions with receptors and ligands. Here, we highlight recent MD simulation studies and enhanced sampling methods used to study biased G protein-coupled receptor signaling and their conformational dynamics as well as applications to drug discovery.
Collapse
Affiliation(s)
- Ye Zou
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - John Ewalt
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Ho-Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
34
|
Structure-function analysis of β-arrestin Kurtz reveals a critical role of receptor interactions in downregulation of GPCR signaling in vivo. Dev Biol 2019; 455:409-419. [PMID: 31325455 DOI: 10.1016/j.ydbio.2019.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 06/15/2019] [Accepted: 07/16/2019] [Indexed: 01/14/2023]
Abstract
Arrestins control signaling via the G protein coupled receptors (GPCRs), serving as both signal terminators and transducers. Previous studies identified several structural elements in arrestins that contribute to their functions as GPCR regulators. However, the importance of these elements in vivo is unclear, and the developmental roles of arrestins are not well understood. We carried out an in vivo structure-function analysis of Kurtz (Krz), the single ortholog of mammalian β-arrestins in the Drosophila genome. A combination of Krz mutations affecting the GPCR-phosphosensing and receptor core-binding ("finger loop") functions (Krz-KKVL/A) resulted in a complete loss of Krz activity during development. Endosome recruitment and bioluminescence resonance energy transfer (BRET) assays revealed that the KKVL/A mutations abolished the GPCR-binding ability of Krz. We found that the isolated "finger loop" mutation (Krz-VL/A), while having a negligible effect on GPCR internalization, severely affected Krz function, suggesting that tight receptor interactions are necessary for proper termination of signaling in vivo. Genetic analysis as well as live imaging demonstrated that mutations in Krz led to hyperactivity of the GPCR Mist (also known as Mthl1), which is activated by its ligand Folded gastrulation (Fog) and is responsible for cellular contractility and epithelial morphogenesis. Krz mutations affected two developmental events that are under the control of Fog-Mist signaling: gastrulation and morphogenesis of the wing. Overall, our data reveal the functional importance in vivo of direct β-arrestin/GPCR binding, which is mediated by the recognition of the phosphorylated receptor tail and receptor core interaction. These Krz-GPCR interactions are critical for setting the correct level of Fog-Mist signaling during epithelial morphogenesis.
Collapse
|
35
|
Tsai CJ, Marino J, Adaixo R, Pamula F, Muehle J, Maeda S, Flock T, Taylor NMI, Mohammed I, Matile H, Dawson RJP, Deupi X, Stahlberg H, Schertler G. Cryo-EM structure of the rhodopsin-Gαi-βγ complex reveals binding of the rhodopsin C-terminal tail to the gβ subunit. eLife 2019; 8:e46041. [PMID: 31251171 PMCID: PMC6629373 DOI: 10.7554/elife.46041] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/26/2019] [Indexed: 12/22/2022] Open
Abstract
One of the largest membrane protein families in eukaryotes are G protein-coupled receptors (GPCRs). GPCRs modulate cell physiology by activating diverse intracellular transducers, prominently heterotrimeric G proteins. The recent surge in structural data has expanded our understanding of GPCR-mediated signal transduction. However, many aspects, including the existence of transient interactions, remain elusive. We present the cryo-EM structure of the light-sensitive GPCR rhodopsin in complex with heterotrimeric Gi. Our density map reveals the receptor C-terminal tail bound to the Gβ subunit of the G protein, providing a structural foundation for the role of the C-terminal tail in GPCR signaling, and of Gβ as scaffold for recruiting Gα subunits and G protein-receptor kinases. By comparing available complexes, we found a small set of common anchoring points that are G protein-subtype specific. Taken together, our structure and analysis provide new structural basis for the molecular events of the GPCR signaling pathway.
Collapse
Affiliation(s)
- Ching-Ju Tsai
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
| | - Jacopo Marino
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
| | - Ricardo Adaixo
- Center for Cellular Imaging and NanAnalytics (C-CINA), BiozentrumUniversity of BaselBaselSwitzerland
| | - Filip Pamula
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
| | - Jonas Muehle
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
| | - Shoji Maeda
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
| | - Tilman Flock
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
- Department of BiologyETH ZurichZürichSwitzerland
| | - Nicholas MI Taylor
- Center for Cellular Imaging and NanAnalytics (C-CINA), BiozentrumUniversity of BaselBaselSwitzerland
| | - Inayatulla Mohammed
- Center for Cellular Imaging and NanAnalytics (C-CINA), BiozentrumUniversity of BaselBaselSwitzerland
| | - Hugues Matile
- Pharma Research and Early Development, Therapeutic modalities, Roche Innovation Center BaselHoffmann-La Roche LtdBaselSwitzerland
| | - Roger JP Dawson
- Pharma Research and Early Development, Therapeutic modalities, Roche Innovation Center BaselHoffmann-La Roche LtdBaselSwitzerland
| | - Xavier Deupi
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
- Condensed Matter Theory GroupPaul Scherrer InstituteVilligenSwitzerland
| | - Henning Stahlberg
- Center for Cellular Imaging and NanAnalytics (C-CINA), BiozentrumUniversity of BaselBaselSwitzerland
| | - Gebhard Schertler
- Division of Biology and Chemistry / Laboratory of Biomolecular ResearchPaul Scherrer InstituteVilligenSwitzerland
- Department of BiologyETH ZurichZürichSwitzerland
| |
Collapse
|
36
|
Russo S, De Azevedo WF. Advances in the Understanding of the Cannabinoid Receptor 1 – Focusing on the Inverse Agonists Interactions. Curr Med Chem 2019; 26:1908-1919. [DOI: 10.2174/0929867325666180417165247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 12/31/2022]
Abstract
Background:
Cannabinoid Receptor 1 (CB1) is a membrane protein prevalent in
the central nervous system, whose crystallographic structure has recently been solved. Studies
will be needed to investigate CB1 complexes with its ligands and its role in the development
of new drugs.
Objective:
Our goal here is to review the studies on CB1, starting with general aspects and
focusing on the recent structural studies, with emphasis on the inverse agonists bound structures.
Methods:
We start with a literature review, and then we describe recent studies on CB 1 crystallographic
structure and docking simulations. We use this structural information to depict
protein-ligand interactions. We also describe the molecular docking method to obtain complex
structures of CB 1 with inverse agonists.
Results:
Analysis of the crystallographic structure and docking results revealed the residues
responsible for the specificity of the inverse agonists for CB 1. Most of the intermolecular interactions
involve hydrophobic residues, with the participation of the residues Phe 170 and
Leu 359 in all complex structures investigated in the present study. For the complexes with
otenabant and taranabant, we observed intermolecular hydrogen bonds involving residues His
178 (otenabant) and Thr 197 and Ser 383 (taranabant).
Conclusion:
Analysis of the structures involving inverse agonists and CB 1 revealed the pivotal
role played by residues Phe 170 and Leu 359 in their interactions and the strong intermolecular
hydrogen bonds highlighting the importance of the exploration of intermolecular interactions
in the development of novel inverse agonists.
Collapse
Affiliation(s)
- Silvana Russo
- Laboratory of Computational Systems Biology, School of Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, Porto Alegre-RS 90619-900, Brazil
| | - Walter Filgueira De Azevedo
- Laboratory of Computational Systems Biology, School of Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, Porto Alegre-RS 90619-900, Brazil
| |
Collapse
|
37
|
Seyedabadi M, Ghahremani MH, Albert PR. Biased signaling of G protein coupled receptors (GPCRs): Molecular determinants of GPCR/transducer selectivity and therapeutic potential. Pharmacol Ther 2019; 200:148-178. [PMID: 31075355 DOI: 10.1016/j.pharmthera.2019.05.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
G protein coupled receptors (GPCRs) convey signals across membranes via interaction with G proteins. Originally, an individual GPCR was thought to signal through one G protein family, comprising cognate G proteins that mediate canonical receptor signaling. However, several deviations from canonical signaling pathways for GPCRs have been described. It is now clear that GPCRs can engage with multiple G proteins and the line between cognate and non-cognate signaling is increasingly blurred. Furthermore, GPCRs couple to non-G protein transducers, including β-arrestins or other scaffold proteins, to initiate additional signaling cascades. Receptor/transducer selectivity is dictated by agonist-induced receptor conformations as well as by collateral factors. In particular, ligands stabilize distinct receptor conformations to preferentially activate certain pathways, designated 'biased signaling'. In this regard, receptor sequence alignment and mutagenesis have helped to identify key receptor domains for receptor/transducer specificity. Furthermore, molecular structures of GPCRs bound to different ligands or transducers have provided detailed insights into mechanisms of coupling selectivity. However, receptor dimerization, compartmentalization, and trafficking, receptor-transducer-effector stoichiometry, and ligand residence and exposure times can each affect GPCR coupling. Extrinsic factors including cell type or assay conditions can also influence receptor signaling. Understanding these factors may lead to the development of improved biased ligands with the potential to enhance therapeutic benefit, while minimizing adverse effects. In this review, evidence for ligand-specific GPCR signaling toward different transducers or pathways is elaborated. Furthermore, molecular determinants of biased signaling toward these pathways and relevant examples of the potential clinical benefits and pitfalls of biased ligands are discussed.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Pharmacology, School of Medicine, Bushehr University of Medical Sciences, Iran; Education Development Center, Bushehr University of Medical Sciences, Iran
| | | | - Paul R Albert
- Ottawa Hospital Research Institute, Neuroscience, University of Ottawa, Canada.
| |
Collapse
|
38
|
Gurevich VV, Gurevich EV. The structural basis of the arrestin binding to GPCRs. Mol Cell Endocrinol 2019; 484:34-41. [PMID: 30703488 PMCID: PMC6377262 DOI: 10.1016/j.mce.2019.01.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/04/2019] [Accepted: 01/17/2019] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of signaling proteins targeted by more clinically used drugs than any other protein family. GPCR signaling via G proteins is quenched (desensitized) by the phosphorylation of the active receptor by specific GPCR kinases (GRKs) followed by tight binding of arrestins to active phosphorylated receptors. Thus, arrestins engage two types of receptor elements: those that contain GRK-added phosphates and those that change conformation upon activation. GRKs attach phosphates to serines and threonines in the GPCR C-terminus or any one of the cytoplasmic loops. In addition to these phosphates, arrestins engage the cavity that appears between trans-membrane helices upon receptor activation and several other non-phosphorylated elements. The residues that bind GPCRs are localized on the concave side of both arrestin domains. Arrestins undergo a global conformational change upon receptor binding (become activated). Arrestins serve as important hubs of cellular signaling, emanating from activated GPCRs and receptor-independent.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
39
|
Bond RA, Lucero Garcia-Rojas EY, Hegde A, Walker JKL. Therapeutic Potential of Targeting ß-Arrestin. Front Pharmacol 2019; 10:124. [PMID: 30894814 PMCID: PMC6414794 DOI: 10.3389/fphar.2019.00124] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/31/2019] [Indexed: 12/22/2022] Open
Abstract
ß-arrestins are multifunctional proteins that modulate heptahelical 7 transmembrane receptors, also known as G protein-coupled receptors (GPCRs), a superfamily of receptors that regulate most physiological processes. ß-arrestin modulation of GPCR function includes termination of G protein-dependent signaling, initiation of ß-arrestin-dependent signaling, receptor trafficking to degradative or recycling pathways, receptor transactivation, transcriptional regulation, and localization of second messenger regulators. The pleiotropic influence ß-arrestins exert on these receptors regulates a breadth of physiological functions, and additionally, ß-arrestins are involved in the pathophysiology of numerous and wide-ranging diseases, making them prime therapeutic targets. In this review, we briefly describe the mechanisms by which ß-arrestins regulate GPCR signaling, including the functional cellular mechanisms modulated by ß-arrestins and relate this to observed pathophysiological responses associated with ß-arrestins. We focus on the role for ß-arrestins in transducing cell signaling; a pathway that is complementary to the classical G protein-coupling pathway. The existence of these GPCR dual signaling pathways offers an immense therapeutic opportunity through selective targeting of one signaling pathway over the other. Finally, we will consider several mechanisms by which the potential of dual signaling pathway regulation can be harnessed and the implications for improved disease treatments.
Collapse
Affiliation(s)
- Richard A Bond
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Emilio Y Lucero Garcia-Rojas
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Akhil Hegde
- School of Nursing, Duke University, Durham, NC, United States
| | | |
Collapse
|
40
|
Dores MR, Trejo J. Endo-lysosomal sorting of G-protein-coupled receptors by ubiquitin: Diverse pathways for G-protein-coupled receptor destruction and beyond. Traffic 2018; 20:101-109. [PMID: 30353650 DOI: 10.1111/tra.12619] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/14/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
Ubiquitin is covalently attached to substrate proteins in the form of a single ubiquitin moiety or polyubiquitin chains and has been generally linked to protein degradation, however, distinct types of ubiquitin linkages are also used to control other critical cellular processes like cell signaling. Over forty mammalian G protein-coupled receptors (GPCRs) have been reported to be ubiquitinated, but despite the diverse and rich complexity of GPCR signaling, ubiquitin has been largely ascribed to receptor degradation. Indeed, GPCR ubiquitination targets the receptors for degradation by lysosome, which is mediated by the Endosomal Sorting Complexes Required for Transport (ESCRT) machinery, and the proteasome. This has led to the view that ubiquitin and ESCRTs primarily function as the signal to target GPCRs for destruction. Contrary to this conventional view, studies indicate that ubiquitination of certain GPCRs and canonical ubiquitin-binding ESCRTs are not required for receptor degradation and revealed that diverse and complex pathways exist to regulate endo-lysosomal sorting of GPCRs. In other studies, GPCR ubiquitination has been shown to drive signaling and not receptor degradation and further revealed novel insight into the mechanisms by which GPCRs trigger the activity of the ubiquitination machinery. Here, we discuss the diverse pathways by which ubiquitin controls GPCR endo-lysosomal sorting and beyond.
Collapse
Affiliation(s)
- Michael R Dores
- Department of Biology, Hofstra University, Hempstead, New York
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
41
|
GPCR drug discovery: integrating solution NMR data with crystal and cryo-EM structures. Nat Rev Drug Discov 2018; 18:59-82. [PMID: 30410121 DOI: 10.1038/nrd.2018.180] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The 826 G protein-coupled receptors (GPCRs) in the human proteome regulate key physiological processes and thus have long been attractive drug targets. With the crystal structures of more than 50 different human GPCRs determined over the past decade, an initial platform for structure-based rational design has been established for drugs that target GPCRs, which is currently being augmented with cryo-electron microscopy (cryo-EM) structures of higher-order GPCR complexes. Nuclear magnetic resonance (NMR) spectroscopy in solution is one of the key approaches for expanding this platform with dynamic features, which can be accessed at physiological temperature and with minimal modification of the wild-type GPCR covalent structures. Here, we review strategies for the use of advanced biochemistry and NMR techniques with GPCRs, survey projects in which crystal or cryo-EM structures have been complemented with NMR investigations and discuss the impact of this integrative approach on GPCR biology and drug discovery.
Collapse
|
42
|
Haider RS, Godbole A, Hoffmann C. To sense or not to sense-new insights from GPCR-based and arrestin-based biosensors. Curr Opin Cell Biol 2018; 57:16-24. [PMID: 30408632 DOI: 10.1016/j.ceb.2018.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/18/2018] [Indexed: 12/27/2022]
Abstract
Advances in resolving crystal structures of GPCRs and their binding partners as well as improvements in live-cell microscopy and the fluorescent proteins pallet has greatly driven new ideas for designing optical sensors for the same. Sensors have been developed to monitor ligand binding as well as the ensuing ligand-induced conformational changes in GPCRs, G-proteins and arrestins. In this review we will highlight the functionality of such sensor designs starting from monitoring ligand binding to receptor activation and interaction with arrestins. Furthermore, we will highlight the importance of sensor designs to monitor receptor-dependent arrestin conformations and give an idea about the various detected arrestin conformations and their possible implications.
Collapse
Affiliation(s)
- Raphael Silvanus Haider
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll Straße 2, D-07745 Jena, Germany
| | - Amod Godbole
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll Straße 2, D-07745 Jena, Germany
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll Straße 2, D-07745 Jena, Germany.
| |
Collapse
|
43
|
Wanka L, Babilon S, Kaiser A, Mörl K, Beck-Sickinger AG. Different mode of arrestin-3 binding at the human Y 1 and Y 2 receptor. Cell Signal 2018; 50:58-71. [DOI: 10.1016/j.cellsig.2018.06.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 01/04/2023]
|
44
|
Bandyopadhyay A, Van Eps N, Eger BT, Rauscher S, Yedidi RS, Moroni T, West GM, Robinson KA, Griffin PR, Mitchell J, Ernst OP. A Novel Polar Core and Weakly Fixed C-Tail in Squid Arrestin Provide New Insight into Interaction with Rhodopsin. J Mol Biol 2018; 430:4102-4118. [PMID: 30120952 DOI: 10.1016/j.jmb.2018.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 08/03/2018] [Accepted: 08/09/2018] [Indexed: 12/31/2022]
Abstract
Photoreceptors of the squid Loligo pealei contain a G-protein-coupled receptor (GPCR) signaling system that activates phospholipase C in response to light. Analogous to the mammalian visual system, signaling of the photoactivated GPCR rhodopsin is terminated by binding of squid arrestin (sArr). sArr forms a light-dependent, high-affinity complex with squid rhodopsin, which does not require prior receptor phosphorylation for interaction. This is at odds with classical mammalian GPCR desensitization where an agonist-bound phosphorylated receptor is needed to break stabilizing constraints within arrestins, the so-called "three-element interaction" and "polar core" network, before a stable receptor-arrestin complex can be established. Biophysical and mass spectrometric analysis of the squid rhodopsin-arrestin complex indicates that in contrast to mammalian arrestins, the sArr C-tail is not involved in a stable three-element interaction. We determined the crystal structure of C-terminally truncated sArr that adopts a basal conformation common to arrestins and is stabilized by a series of weak but novel polar core interactions. Unlike mammalian arrestin-1, deletion of the sArr C-tail does not influence kinetic properties of complex formation of sArr with the receptor. Hydrogen-deuterium exchange studies revealed the footprint of the light-activated rhodopsin on sArr. Furthermore, double electron-electron resonance spectroscopy experiments provide evidence that receptor-bound sArr adopts a conformation different from the one known for arrestin-1 and molecular dynamics simulations reveal the residues that account for the weak three-element interaction. Insights gleaned from studying this system add to our general understanding of GPCR-arrestin interaction.
Collapse
Affiliation(s)
| | - Ned Van Eps
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Bryan T Eger
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Rauscher
- Department of Chemical and Physical Sciences, University of Toronto, Mississauga, Ontario L5L 1C6, Canada
| | - Ravikiran S Yedidi
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tina Moroni
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Graham M West
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Kelly Ann Robinson
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Patrick R Griffin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Jane Mitchell
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Oliver P Ernst
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
45
|
Chen Q, Iverson TM, Gurevich VV. Structural Basis of Arrestin-Dependent Signal Transduction. Trends Biochem Sci 2018; 43:412-423. [PMID: 29636212 PMCID: PMC5959776 DOI: 10.1016/j.tibs.2018.03.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/03/2018] [Accepted: 03/12/2018] [Indexed: 12/29/2022]
Abstract
Arrestins are a small family of proteins with four isoforms in humans. Remarkably, two arrestins regulate signaling from >800 G protein-coupled receptors (GPCRs) or nonreceptor activators by simultaneously binding an activator and one out of hundreds of other signaling proteins. When arrestins are bound to GPCRs or other activators, the affinity for these signaling partners changes. Thus, it is proposed that an activator alters arrestin's ability to transduce a signal. The comparison of all available arrestin structures identifies several common conformational rearrangements associated with activation. In particular, it identifies elements that are directly involved in binding to GPCRs or other activators, elements that likely engage distinct downstream effectors, and elements that likely link the activator-binding sites with the effector-binding sites.
Collapse
Affiliation(s)
- Qiuyan Chen
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47906, USA
| | - Tina M Iverson
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232-0146, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37232-0146, USA; Center for Structural Biology, Vanderbilt University, Nashville, TN 37232-0146, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232-0146, USA.
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232-0146, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232-0146, USA.
| |
Collapse
|
46
|
Eichel K, Jullié D, Barsi-Rhyne B, Latorraca NR, Masureel M, Sibarita JB, Dror RO, von Zastrow M. Catalytic activation of β-arrestin by GPCRs. Nature 2018; 557:381-386. [PMID: 29720660 PMCID: PMC6058965 DOI: 10.1038/s41586-018-0079-1] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 03/20/2018] [Indexed: 12/21/2022]
Abstract
β-arrestins are critical regulator and transducer proteins for G-protein-coupled receptors (GPCRs). β-arrestin is widely believed to be activated by forming a stable and stoichiometric GPCR-β-arrestin scaffold complex, which requires and is driven by the phosphorylated tail of the GPCR. Here we demonstrate a distinct and additional mechanism of β-arrestin activation that does not require stable GPCR-β-arrestin scaffolding or the GPCR tail. Instead, it occurs through transient engagement of the GPCR core, which destabilizes a conserved inter-domain charge network in β-arrestin. This promotes capture of β-arrestin at the plasma membrane and its accumulation in clathrin-coated endocytic structures (CCSs) after dissociation from the GPCR, requiring a series of interactions with membrane phosphoinositides and CCS-lattice proteins. β-arrestin clustering in CCSs in the absence of the upstream activating GPCR is associated with a β-arrestin-dependent component of the cellular ERK (extracellular signal-regulated kinase) response. These results delineate a discrete mechanism of cellular β-arrestin function that is activated catalytically by GPCRs.
Collapse
Affiliation(s)
- Kelsie Eichel
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Damien Jullié
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Benjamin Barsi-Rhyne
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Naomi R Latorraca
- Biophysics Program, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Matthieu Masureel
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jean-Baptiste Sibarita
- Interdisciplinary Institute for Neuroscience, UMR 5297, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Ron O Dror
- Biophysics Program, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Mark von Zastrow
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, CA, USA.
- Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
47
|
Abstract
G protein-coupled receptors (GPCRs), which mediate processes as diverse as olfaction and maintenance of metabolic homeostasis, have become the single most effective class of therapeutic drug targets. As a result, understanding the molecular basis for their activity is of paramount importance. Recent technological advances have made GPCR structural biology increasingly tractable, offering views of these receptors in unprecedented atomic detail. Structural and biophysical data have shown that GPCRs function as complex allosteric machines, communicating ligand-binding events through conformational change. Changes in receptor conformation lead to activation of effector proteins, such as G proteins and arrestins, which are themselves conformational switches. Here, we review how structural biology has illuminated the agonist-induced cascade of conformational changes that culminate in a cellular response to GPCR activation.
Collapse
Affiliation(s)
- Sarah C Erlandson
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Conor McMahon
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
48
|
Elgeti M, Kazmin R, Rose AS, Szczepek M, Hildebrand PW, Bartl FJ, Scheerer P, Hofmann KP. The arrestin-1 finger loop interacts with two distinct conformations of active rhodopsin. J Biol Chem 2018; 293:4403-4410. [PMID: 29363577 DOI: 10.1074/jbc.m117.817890] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/17/2018] [Indexed: 11/06/2022] Open
Abstract
Signaling of the prototypical G protein-coupled receptor (GPCR) rhodopsin through its cognate G protein transducin (Gt) is quenched when arrestin binds to the activated receptor. Although the overall architecture of the rhodopsin/arrestin complex is known, many questions regarding its specificity remain unresolved. Here, using FTIR difference spectroscopy and a dual pH/peptide titration assay, we show that rhodopsin maintains certain flexibility upon binding the "finger loop" of visual arrestin (prepared as synthetic peptide ArrFL-1). We found that two distinct complexes can be stabilized depending on the protonation state of E3.49 in the conserved (D)ERY motif. Both complexes exhibit different interaction modes and affinities of ArrFL-1 binding. The plasticity of the receptor within the rhodopsin/ArrFL-1 complex stands in contrast to the complex with the C terminus of the Gt α-subunit (GαCT), which stabilizes only one specific substate out of the conformational ensemble. However, Gt α-subunit binding and both ArrFL-1-binding modes involve a direct interaction to conserved R3.50, as determined by site-directed mutagenesis. Our findings highlight the importance of receptor conformational flexibility and cytoplasmic proton uptake for modulation of rhodopsin signaling and thereby extend the picture provided by crystal structures of the rhodopsin/arrestin and rhodopsin/ArrFL-1 complexes. Furthermore, the two binding modes of ArrFL-1 identified here involve motifs of conserved amino acids, which indicates that our results may have elucidated a common modulation mechanism of class A GPCR-G protein/-arrestin signaling.
Collapse
Affiliation(s)
- Matthias Elgeti
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany,
| | - Roman Kazmin
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Alexander S Rose
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Group ProteInformatics
| | - Michal Szczepek
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Group Protein X-ray Crystallography and Signal Transduction
| | - Peter W Hildebrand
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Institut für Medizinische Physik und Biophysik, Universität Leipzig, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Franz J Bartl
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Institut für Biologie, Biophysikalische Chemie, Humboldt-Universität zu Berlin, Invalidenstrasse 42, 10115 Berlin, Germany
| | - Patrick Scheerer
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Group Protein X-ray Crystallography and Signal Transduction
| | - Klaus Peter Hofmann
- From the Institut für Medizinische Physik und Biophysik (CC2), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
49
|
Hilger D, Masureel M, Kobilka BK. Structure and dynamics of GPCR signaling complexes. Nat Struct Mol Biol 2018; 25:4-12. [PMID: 29323277 PMCID: PMC6535338 DOI: 10.1038/s41594-017-0011-7] [Citation(s) in RCA: 624] [Impact Index Per Article: 89.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/21/2017] [Indexed: 12/16/2022]
Abstract
G-protein-coupled receptors (GPCRs) relay numerous extracellular signals by triggering intracellular signaling through coupling with G proteins and arrestins. Recent breakthroughs in the structural determination of GPCRs and GPCR-transducer complexes represent important steps toward deciphering GPCR signal transduction at a molecular level. A full understanding of the molecular basis of GPCR-mediated signaling requires elucidation of the dynamics of receptors and their transducer complexes as well as their energy landscapes and conformational transition rates. Here, we summarize current insights into the structural plasticity of GPCR-G-protein and GPCR-arrestin complexes that underlies the regulation of the receptor's intracellular signaling profile.
Collapse
Affiliation(s)
- Daniel Hilger
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthieu Masureel
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|