1
|
Luo Y, Sun L, Peng Y. The structural basis of the G protein-coupled receptor and ion channel axis. Curr Res Struct Biol 2025; 9:100165. [PMID: 40083915 PMCID: PMC11904507 DOI: 10.1016/j.crstbi.2025.100165] [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: 09/26/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Sensory neurons play an essential role in recognizing and responding to detrimental, irritating, and inflammatory stimuli from our surroundings, such as pain, itch, cough, and neurogenic inflammation. The transduction of these physiological signals is chiefly mediated by G protein-coupled receptors (GPCRs) and ion channels. The binding of ligands to GPCRs triggers a signaling cascade, recruiting G proteins or β-arrestins, which subsequently interact with ion channels (e.g., GIRK and TRP channels). This interaction leads to the sensitization and activation of these channels, initiating the neuron's protective mechanisms. This review delves into the complex interplay between GPCRs and ion channels that underpin these physiological processes, with a particular focus on the role of structural biology in enhancing our comprehension. Through unraveling the intricacies of the GPCR-ion channel axis, we aim to shed light on the sophisticated intermolecular dynamics within these pivotal membrane protein families, ultimately guiding the development of precise therapeutic interventions.
Collapse
Affiliation(s)
- Yulin Luo
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, L Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| |
Collapse
|
2
|
Lim HD, Bartuzi D, Keen AC, Rauffenbart C, Glenn J, Charlton SJ, Lovera S, Sands ZA, Ates A, Wood M, Canals M, Javitch JA, Carlsson J, Lane JR. Identification of a Lipid-Exposed Extrahelical Binding Site for Positive Allosteric Modulators of the Dopamine D 2 Receptor. ACS Chem Neurosci 2025. [PMID: 40372152 DOI: 10.1021/acschemneuro.5c00105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
Recently, the first small-molecule positive allosteric modulators (PAMs) of the dopamine D2 receptor (D2R) were identified. The more potent PAM potentiated the effects of D2R signaling in vitro and in an in vivo model predictive of anti-Parkinson's efficacy. We reveal, based on the results of our site-directed mutagenesis and molecular dynamics experiments, that this scaffold binds to a hitherto unexploited lipid-exposed extrahelical allosteric site in the D2R that lies in a cleft toward the intracellular aspect of the D2R defined by residues in transmembrane domains 1 and 7 and helix 8. By binding to this site, the PAM acts to potentiate the binding affinity of efficacious agonists, such as dopamine. Our simulations suggest that the PAM achieves this effect by stabilizing an active-like conformation of the receptor, similar to the G protein-bound state with TM5 and the tyrosine toggle switch playing the major role.
Collapse
Affiliation(s)
- Herman D Lim
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Damian Bartuzi
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala SE- 751 24, Sweden
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., Lublin 20093, Poland
| | - Alastair C Keen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 399 Royal Parade, Parkville, VIC 3052, Australia
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Caroline Rauffenbart
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Jacqueline Glenn
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Steven J Charlton
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Silvia Lovera
- UCB BioPharma SPRL, Chemin de Foriest, Braine-l'Alleud, Brussels B-1420, Belgium
| | - Zara A Sands
- UCB BioPharma SPRL, Chemin de Foriest, Braine-l'Alleud, Brussels B-1420, Belgium
| | - Ali Ates
- UCB BioPharma SPRL, Chemin de Foriest, Braine-l'Alleud, Brussels B-1420, Belgium
| | - Martyn Wood
- UCB BioPharma SPRL, Chemin de Foriest, Braine-l'Alleud, Brussels B-1420, Belgium
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Jonathan A Javitch
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala SE- 751 24, Sweden
| | - J Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
3
|
Krumm BE, Roth BL. Intracellular GPCR modulators enable precision pharmacology. NPJ DRUG DISCOVERY 2025; 2:8. [PMID: 40371403 PMCID: PMC12069105 DOI: 10.1038/s44386-025-00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/20/2025] [Indexed: 05/16/2025]
Abstract
G-protein-coupled receptors (GPCRs) have proven to be the most successful target class for drug discovery but their complicated signal transduction pathways cause difficulties for drug development. Recently, ligands have been identified that engage an intracellular binding site which promotes pathway biased signal in cooperation with orthosteric ligands. Here, we explore the topic of biased signaling and intracellular modulators to understand their application for precision pharmacology of Class A or Rhodopsin-Like GPCRs.
Collapse
Affiliation(s)
- Brian E. Krumm
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Bryan L. Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| |
Collapse
|
4
|
Dashevskii D, Luginina A, Maslov I, Shevelyova M, Khorn P, Dmitrieva D, Kapranov I, Belousov A, Permyakov S, Cherezov V, Borshchevskiy V, Mishin A. Unlocking GPCR-ligand interactions: Measuring binding affinities with thermal shift assay. Protein Sci 2025; 34:e70120. [PMID: 40247825 PMCID: PMC12006757 DOI: 10.1002/pro.70120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Abstract
G protein-coupled receptors (GPCRs) constitute the largest transmembrane protein superfamily, with over 800 representatives in the human genome. Recognized as pivotal targets in pharmacological research and drug discovery, these receptors play a crucial role in advancing therapeutics. Understanding the molecular mechanisms of receptor-ligand interactions is imperative for drug discovery applications. However, experimental procedures for measuring ligand binding are complicated by various factors, including the transmembrane nature of the receptors and the high cost associated with specialized instruments and consumables. Here we introduce an application of the thermal shift assay (TSA) to measuring ligand binding affinities for GPCRs. TSA is a cost-effective and user-friendly method that detects changes in protein stability induced by alterations in environmental conditions. Employing the human A2A adenosine receptor as a representative GPCR, we determined binding constants for four orthosteric ligands and allosteric sodium using three mathematical models for TSA data approximation and analysis. Models were additionally validated by two antagonists of cysteinyl leukotriene GPCR (CysLT1R), used as antiasthmatic drugs. Our results suggest that the TSA approach demonstrates a high degree of reproducibility and agreement with existing literature data, thereby affirming its suitability for investigating GPCR interactions with various types of ligands.
Collapse
Affiliation(s)
- Dmitrii Dashevskii
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Aleksandra Luginina
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Ivan Maslov
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Marina Shevelyova
- Pushchino Scientific Center for Biological Research of the Russian Academy of ScienceInstitute for Biological InstrumentationPushchinoRussia
| | - Polina Khorn
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Daria Dmitrieva
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Ivan Kapranov
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| | - Anatolii Belousov
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
- Sao Carlos Institute of PhysicsUniversity of Sao PauloSao CarlosSão PaoloBrazil
| | - Sergei Permyakov
- Pushchino Scientific Center for Biological Research of the Russian Academy of ScienceInstitute for Biological InstrumentationPushchinoRussia
| | - Vadim Cherezov
- Bridge Institute, Department of ChemistryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Valentin Borshchevskiy
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
- Frank Laboratory of Neutron PhysicsDubnaRussia
| | - Alexey Mishin
- Research Center for Molecular Mechanisms of Aging and Age‐Related DiseasesMoscow Institute of Physics and TechnologyMoscowRussia
| |
Collapse
|
5
|
Russell IC, Lee D, Wootten D, Sexton PM, Bumbak F. Cryoelectron microscopy as a tool for illuminating activation mechanisms of human class A orphan G protein-coupled receptors. Pharmacol Rev 2025; 77:100056. [PMID: 40286430 DOI: 10.1016/j.pharmr.2025.100056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are critically important medicinal targets, and the cryogenic electron microscopy (cryo-EM) revolution is providing novel high-resolution GPCR structures at a rapid pace. Orphan G protein-coupled receptors (oGPCRs) are a group of approximately 100 nonolfactory GPCRs for which endogenous ligands are unknown or not validated. The absence of modulating ligands adds difficulties to understanding the physiologic significance of oGPCRs and in the determination of high-resolution structures of isolated receptors that could facilitate drug discovery. Despite the challenges, cryo-EM structures of oGPCR-G protein complexes are emerging. This is being facilitated by numerous developments to stabilize GPCR-G protein complexes such as the use of dominant-negative G proteins, mini-G proteins, complex-stabilizing nanobodies or antibody fragments, and protein tethering methods. Moreover, many oGPCRs are constitutively active, which can facilitate complex formation in the absence of a known activating ligand. Consequently, in addition to providing templates for drug discovery, active oGPCR structures shed light on constitutive GPCR activation mechanisms. These comprise self-activation, whereby mobile extracellular portions of the receptor act as tethered agonists by occupying a canonical orthosteric-binding site in the transmembrane core, constitutive activity due to alterations to conserved molecular switches that stabilize inactive states of GPCRs, as well as receptors activated by cryptic ligands that are copurified with the receptor. Cryo-EM structures of oGPCRs are now being determined at a rapid pace and are expected to be invaluable tools for oGPCR drug discovery. SIGNIFICANCE STATEMENT: Orphan G protein-coupled receptors (GPCRs) provide large untapped potential for development of new medicines. Many of these receptors display constitutive activity, enabling structure determination and insights into observed GPCR constitutive activity including (1) self-activation by mobile receptor extracellular portions that function as tethered agonists, (2) modification of conserved motifs canonically involved in receptor quiescence and/or activation, and (3) activation by cryptic lipid ligands. Collectively, these studies advance fundamental understanding of GPCR function and provide opportunities for novel drug discovery.
Collapse
Affiliation(s)
- Isabella C Russell
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Dongju Lee
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Denise Wootten
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| | - Patrick M Sexton
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| | - Fabian Bumbak
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| |
Collapse
|
6
|
Conflitti P, Lyman E, Sansom MSP, Hildebrand PW, Gutiérrez-de-Terán H, Carloni P, Ansell TB, Yuan S, Barth P, Robinson AS, Tate CG, Gloriam D, Grzesiek S, Eddy MT, Prosser S, Limongelli V. Functional dynamics of G protein-coupled receptors reveal new routes for drug discovery. Nat Rev Drug Discov 2025; 24:251-275. [PMID: 39747671 PMCID: PMC11968245 DOI: 10.1038/s41573-024-01083-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
G protein-coupled receptors (GPCRs) are the largest human membrane protein family that transduce extracellular signals into cellular responses. They are major pharmacological targets, with approximately 26% of marketed drugs targeting GPCRs, primarily at their orthosteric binding site. Despite their prominence, predicting the pharmacological effects of novel GPCR-targeting drugs remains challenging due to the complex functional dynamics of these receptors. Recent advances in X-ray crystallography, cryo-electron microscopy, spectroscopic techniques and molecular simulations have enhanced our understanding of receptor conformational dynamics and ligand interactions with GPCRs. These developments have revealed novel ligand-binding modes, mechanisms of action and druggable pockets. In this Review, we highlight such aspects for recently discovered small-molecule drugs and drug candidates targeting GPCRs, focusing on three categories: allosteric modulators, biased ligands, and bivalent and bitopic compounds. Although studies so far have largely been retrospective, integrating structural data on ligand-induced receptor functional dynamics into the drug discovery pipeline has the potential to guide the identification of drug candidates with specific abilities to modulate GPCR interactions with intracellular effector proteins such as G proteins and β-arrestins, enabling more tailored selectivity and efficacy profiles.
Collapse
Affiliation(s)
- Paolo Conflitti
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Edward Lyman
- Department of Physics and Astronomy, University of Delaware, Newark, DE, USA
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Paolo Carloni
- INM-9/IAS-5 Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - T Bertie Ansell
- Department of Biochemistry, University of Oxford, Oxford, UK
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Shuguang Yuan
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - David Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark
| | - Stephan Grzesiek
- Focal Area Structural Biology and Biophysics, Biozentrum, University of Basel, Basel, Switzerland
| | - Matthew T Eddy
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Scott Prosser
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - Vittorio Limongelli
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland.
| |
Collapse
|
7
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [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: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
8
|
Kugawa M, Kawakami K, Kise R, Suomivuori CM, Tsujimura M, Kobayashi K, Kojima A, Inoue WJ, Fukuda M, Matsui TE, Fukunaga A, Koyanagi J, Kim S, Ikeda H, Yamashita K, Saito K, Ishikita H, Dror RO, Inoue A, Kato HE. Structural insights into lipid chain-length selectivity and allosteric regulation of FFA2. Nat Commun 2025; 16:2809. [PMID: 40140663 PMCID: PMC11947310 DOI: 10.1038/s41467-025-57983-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
The free fatty acid receptor 2 (FFA2) is a G protein-coupled receptor (GPCR) that selectively recognizes short-chain fatty acids to regulate metabolic and immune functions. As a promising therapeutic target, FFA2 has been the focus of intensive development of synthetic ligands. However, the mechanisms by which endogenous and synthetic ligands modulate FFA2 activity remain unclear. Here, we present the structures of the human FFA2-Gi complex activated by the synthetic orthosteric agonist TUG-1375 and the positive allosteric modulator/allosteric agonist 4-CMTB, along with the structure of the inactive FFA2 bound to the antagonist GLPG0974. Structural comparisons with FFA1 and mutational studies reveal how FFA2 selects specific fatty acid chain lengths. Moreover, our structures reveal that GLPG0974 functions as an allosteric antagonist by binding adjacent to the orthosteric pocket to block agonist binding, whereas 4-CMTB binds the outer surface of transmembrane helices 6 and 7 to directly activate the receptor. Supported by computational and functional studies, these insights illuminate diverse mechanisms of ligand action, paving the way for precise GPCR-targeted drug design.
Collapse
Affiliation(s)
- Mai Kugawa
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kouki Kawakami
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Carl-Mikael Suomivuori
- Department of Computer Science, Stanford University, Stanford, CA, USA
- 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
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Masaki Tsujimura
- Department of Advanced Interdisciplinary Studies, The University of Tokyo, Meguro, Tokyo, Japan
| | - Kazuhiro Kobayashi
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Asato Kojima
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Wakana J Inoue
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Masahiro Fukuda
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Toshiki E Matsui
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Ayami Fukunaga
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Junki Koyanagi
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Suhyang Kim
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Hisako Ikeda
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Keitaro Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Keisuke Saito
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Applied Chemistry, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Hiroshi Ishikita
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Applied Chemistry, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, USA
- 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
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
- FOREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| | - Hideaki E Kato
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan.
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan.
- FOREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| |
Collapse
|
9
|
Schettler F, Gattor AO, Koch P, Keller M. Characterization of [ 3H]Propionylated Human Peptide YY-A New Probe for Neuropeptide Y Y 2 Receptor Binding Studies. ACS Pharmacol Transl Sci 2025; 8:785-799. [PMID: 40109743 PMCID: PMC11915035 DOI: 10.1021/acsptsci.4c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
The neuropeptide Y (NPY) Y2 receptor (Y2R) is a G-protein-coupled receptor that is involved in the regulation of various physiological processes such as neurotransmitter release, bone metabolism, and memory. Consequently, the Y2R represents a potential drug target, e.g., for the treatment of epilepsy and mood disorders. Until now, the determination of the Y2R binding affinities of Y2R ligands has primarily been performed using 125I-labeled derivatives of the endogenous Y2R agonists NPY and peptide YY (PYY). A tritium-labeled NPY derivative has also been used; however, its suitability for binding assays in sodium-containing buffer is doubtful. We synthesized a tritium-labeled PYY derivative by [3H]propionylation at Lys4 ([3H]2). The radioligand was characterized by saturation binding, association, and dissociation kinetics and was applied in competition binding assays. Specific binding of [3H]2 at intact Chinese hamster ovary cells expressing the hY2R was saturable in both sodium-free buffer (apparent K d = 0.016-0.067 nM) and sodium-containing buffer (175 mM Na+, apparent K d = 0.16-0.18 nM). Competition binding experiments with Y2R reference ligands yielded K i values, which are in good agreement with the reported Y2R binding affinities, showing that [3H]2 represents a useful tritiated tool compound for the determination of Y2R binding affinities also in buffers containing sodium at physiological concentrations.
Collapse
Affiliation(s)
- Franziska Schettler
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| | - Albert O Gattor
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| | - Pierre Koch
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| |
Collapse
|
10
|
Varga BR, Bernhard SM, El Daibani A, Zaidi SA, Lam JH, Aguilar J, Appourchaux K, Nazarova AL, Kouvelis A, Shinouchi R, Hammond HR, Eans SO, Weinreb V, Margolis EB, Fay JF, Huang XP, Pradhan A, Katritch V, McLaughlin JP, Majumdar S, Che T. Structure-guided design of partial agonists at an opioid receptor. Nat Commun 2025; 16:2518. [PMID: 40082451 PMCID: PMC11906898 DOI: 10.1038/s41467-025-57734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Chronic pain and opioid overdose deaths highlight the need for non-addictive analgesics with novel mechanisms. The δ opioid receptor (δOR) is a promising target, as it lacks the respiratory depression associated with µ opioid receptor (µOR) agonists. However, early δOR full agonists caused seizures, limiting their clinical use. Partial δOR agonists may offer more controlled receptor activation than full agonists, but their development has been hindered by uncertainty regarding the molecular mechanism of partial agonism. Here we show that C6-Quino, a bitopic ligand developed through structure-based design, acts as a selective δOR partial agonist. Functional studies reveal that C6-Quino shows differential activity at G-protein and arrestin pathways and interacts with the sodium binding pocket, confirmed through cryo-EM analysis. C6-Quino demonstrates oral activity, analgesic activity in chronic pain models without causing δOR-related seizures and µOR-related adverse effects which have limited opioid usage in recent times. This discovery outlines a new strategy for developing δOR-targeted analgesics and provides a framework for optimizing signaling profiles of other Class A GPCRs.
Collapse
MESH Headings
- Animals
- Humans
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, delta/chemistry
- Drug Design
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/chemistry
- Cryoelectron Microscopy
- Mice
- Chronic Pain/drug therapy
- HEK293 Cells
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Male
- Ligands
- Drug Partial Agonism
Collapse
Affiliation(s)
- Balazs R Varga
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah M Bernhard
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Amal El Daibani
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Saheem A Zaidi
- Department of Quantitative & Computational Biology and Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Jordy H Lam
- Department of Quantitative & Computational Biology and Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Jhoan Aguilar
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin Appourchaux
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Antonina L Nazarova
- Department of Quantitative & Computational Biology and Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Alexa Kouvelis
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryosuke Shinouchi
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Haylee R Hammond
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Shainnel O Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Violetta Weinreb
- Department of Pharmacology School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Elyssa B Margolis
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jonathan F Fay
- Department of Biochemistry and Molecular Biology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Xi-Ping Huang
- Department of Pharmacology School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Amynah Pradhan
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vsevolod Katritch
- Department of Quantitative & Computational Biology and Department of Chemistry, University of Southern California, Los Angeles, CA, USA.
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA.
| | - Susruta Majumdar
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Tao Che
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Gong Z, Zhang X, Liu M, Jin C, Hu Y. Visualizing agonist-induced M2 receptor activation regulated by aromatic ring dynamics. Proc Natl Acad Sci U S A 2025; 122:e2418559122. [PMID: 40053366 PMCID: PMC11912407 DOI: 10.1073/pnas.2418559122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Despite the growing number of G protein-coupled receptor (GPCR) structures being resolved, the dynamic process of how GPCRs transit from the inactive toward the active state remains unclear. In this study, comprehensive molecular dynamics simulations were performed to explore how ligand binding modulates the conformational dynamics of the M2 muscarinic acetylcholine receptor (M2R). We observed a sequential occurrence of structural changes in the inactive-to-active transition of M2R induced by a superagonist iperoxo, which includes the orthosteric binding site contraction, the TM6 opening into an intermediate conformation, and a further structural change toward full activation upon binding to G protein or a G protein mimetic nanobody. Two activation intermediates were identified, which show structural features different from those reported for apo-GPCRs. Moreover, our results suggest that stabilization of a specific W4006.48 conformation and enhanced F3966.44 dynamics are crucial for activation, whereas distinct side-chain rotamer equilibriums of Y2065.58 in the cytoplasmic cavity are correlated with agonist efficacies. Our work provides atomic-level structural insights into the agonist-induced M2R activation pathway and highlights a mechanism by which ligand efficacy can be encoded and transduced in the form of aromatic ring dynamics.
Collapse
Affiliation(s)
- Zhou Gong
- State Key Laboratory of Magnetic Resonance Spectroscopy and Imaging, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan430071, China
| | - Xu Zhang
- State Key Laboratory of Magnetic Resonance Spectroscopy and Imaging, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan430071, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance Spectroscopy and Imaging, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan430071, China
| | - Changwen Jin
- Beijing NMR Center, College of Chemistry and Molecular Engineering and Beijing National Laboratory for Molecular Sciences, Peking University, Beijing100871, China
| | - Yunfei Hu
- State Key Laboratory of Magnetic Resonance Spectroscopy and Imaging, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan430071, China
| |
Collapse
|
12
|
Bansal PD, Kinnebrew M, Rohatgi R, Shukla D. Multiple modes of cholesterol translocation in the human Smoothened receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.25.625241. [PMID: 39651171 PMCID: PMC11623640 DOI: 10.1101/2024.11.25.625241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Smoothened (SMO), a member of the G Protein-Coupled Receptor superfamily, mediates Hedgehog signaling and is linked to cancer and birth defects. SMO responds to accessible cholesterol in the ciliary membrane, translocating it via a longitudinal tunnel to its extracellular domain. Reaching a complete mechanistic understanding of the cholesterol translocation process would help in the development of cancer therapies. Experimental data suggests two modes of translocation to support entry of cholesterol from outer and inner membrane leaflets, but the exact mechanism of translocation remains unclear. Using atomistic molecular dynamics simulations (~2 millisecond simulations) and biochemical assays of SMO mutants, we assess the energetic feasibilities of the two modes. We show that the highest energetic barrier for cholesterol translocation from the outer leaflet is lower than that from the inner leaflet. Mutagenesis experiments and complementary simulations of SMO mutants validate the role of critical amino acid residues along the translocation pathways. Our data suggests that cholesterol can take either pathway to enter SMO, thus explaining experimental observations in literature. Thus, our results illuminate the energetics and provide a first molecular description of cholesterol translocation in SMO.
Collapse
Affiliation(s)
- Prateek D Bansal
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Maia Kinnebrew
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Rajat Rohatgi
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
| |
Collapse
|
13
|
Aranda-García D, Stepniewski TM, Torrens-Fontanals M, García-Recio A, Lopez-Balastegui M, Medel-Lacruz B, Morales-Pastor A, Peralta-García A, Dieguez-Eceolaza M, Sotillo-Nuñez D, Ding T, Drabek M, Jacquemard C, Jakowiecki J, Jespers W, Jiménez-Rosés M, Jun-Yu-Lim V, Nicoli A, Orzel U, Shahraki A, Tiemann JKS, Ledesma-Martin V, Nerín-Fonz F, Suárez-Dou S, Canal O, Pándy-Szekeres G, Mao J, Gloriam DE, Kellenberger E, Latek D, Guixà-González R, Gutiérrez-de-Terán H, Tikhonova IG, Hildebrand PW, Filizola M, Babu MM, Di Pizio A, Filipek S, Kolb P, Cordomi A, Giorgino T, Marti-Solano M, Selent J. Large scale investigation of GPCR molecular dynamics data uncovers allosteric sites and lateral gateways. Nat Commun 2025; 16:2020. [PMID: 40016203 PMCID: PMC11868581 DOI: 10.1038/s41467-025-57034-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025] Open
Abstract
G protein-coupled receptors (GPCRs) constitute a functionally diverse protein family and are targets for a broad spectrum of pharmaceuticals. Technological progress in X-ray crystallography and cryogenic electron microscopy has enabled extensive, high-resolution structural characterisation of GPCRs in different conformational states. However, as highly dynamic events underlie GPCR signalling, a complete understanding of GPCR functionality requires insights into their conformational dynamics. Here, we present a large dataset of molecular dynamics simulations covering 60% of currently available GPCR structures. Our analysis reveals extensive local "breathing" motions of the receptor on a nano- to microsecond timescale and provides access to numerous previously unexplored receptor conformational states. Furthermore, we reveal that receptor flexibility impacts the shape of allosteric drug binding sites, which frequently adopt partially or completely closed states in the absence of a molecular modulator. We demonstrate that exploring membrane lipid dynamics and their interaction with GPCRs is an efficient approach to expose such hidden allosteric sites and even lateral ligand entrance gateways. The obtained insights and generated dataset on conformations, allosteric sites and lateral entrance gates in GPCRs allows us to better understand the functionality of these receptors and opens new therapeutic avenues for drug-targeting strategies.
Collapse
Affiliation(s)
- David Aranda-García
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- InterAx Biotech AG, Villigen, Switzerland
| | - Mariona Torrens-Fontanals
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Acellera Labs, Barcelona, Spain
| | - Adrian García-Recio
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Marta Lopez-Balastegui
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Brian Medel-Lacruz
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Adrián Morales-Pastor
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | | | - Miguel Dieguez-Eceolaza
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - David Sotillo-Nuñez
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Tianyi Ding
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Matthäus Drabek
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Célien Jacquemard
- Laboratoire d'Innovation Thérapeutique, University of Strasbourg, Strasbourg, France
| | - Jakub Jakowiecki
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
- Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Willem Jespers
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mireia Jiménez-Rosés
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Birmingham and Nottingham, Midlands, UK
- Sygnature Discovery Ltd., Nottingham, UK
| | - Víctor Jun-Yu-Lim
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Urszula Orzel
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
- Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Aida Shahraki
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Johanna K S Tiemann
- Medizinische Fakultät, Institut für Medizinische Physik und Biophysik, Universität Leipzig, Leipzig, Germany
| | - Vicente Ledesma-Martin
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Francho Nerín-Fonz
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Sergio Suárez-Dou
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Oriol Canal
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Gáspár Pándy-Szekeres
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Medicinal Chemistry Research Group, HUN-REN Research Center for Natural Sciences, Budapest, Hungary
| | - Jiafei Mao
- Beijing National Laboratory for Molecular Sciences (BNLMS) and Center for Physicochemical Analysis and Measurement, Institute of Chemistry Chinese Academy of Science (ICCAS), Beijing, China
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Esther Kellenberger
- Laboratoire d'Innovation Thérapeutique, University of Strasbourg, Strasbourg, France
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Ramon Guixà-González
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Uppsala, Sweden
- Research Center in Nanomaterials and Nanotechnology (CINN/CSIC) and Health Institute of Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Irina G Tikhonova
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Medical University Leipzig, Leipzig, Sachsen, Germany
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Madan Babu
- Department of Structural Biology and Center of Excellence for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | | | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Arnau Cordomi
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Toni Giorgino
- Institute of Biophysics (IBF-CNR), National Research Council of Italy, Milano, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | | | - Jana Selent
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain.
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| |
Collapse
|
14
|
Ramos-Gonzalez N, Varga BR, Majumdar S. Advances in the structural understanding of opioid allostery. Trends Pharmacol Sci 2025; 46:98-101. [PMID: 39827063 PMCID: PMC12001378 DOI: 10.1016/j.tips.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
Activation of the μ opioid receptor (MOR) can give analgesia, but also has dangerous side effects. Drugs that target MOR through an allosteric site, meaning they bind outside of the usual pocket, present a novel mode of receptor activation with different pharmacology relative to orthosteric drugs. Recent structural studies give valuable new information on how allosteric modulators interact with MOR.
Collapse
Affiliation(s)
- Nokomis Ramos-Gonzalez
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St Louis, MO, USA; Center for Clinical Pharmacology, Washington University School of Medicine, St Louis, MO, USA
| | - Balazs R Varga
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St Louis, MO, USA; Center for Clinical Pharmacology, Washington University School of Medicine, St Louis, MO, USA
| | - Susruta Majumdar
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St Louis, MO, USA; Center for Clinical Pharmacology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
15
|
Pettersen JM, McCracken O, Robinson AS. Ligand binding kinetics to evaluate the function and stability of A 2AR in nanodiscs. Biophys J 2025; 124:440-457. [PMID: 39690743 PMCID: PMC11788476 DOI: 10.1016/j.bpj.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/15/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024] Open
Abstract
G-protein-coupled receptors (GPCRs) represent one of the largest classes of therapeutic targets. However, developing successful therapeutics to target GPCRs is a challenging endeavor, with many molecules failing during in vivo clinical trials due to a lack of efficacy. The in vitro identification of drug-target residence time (1/koff) has been suggested to improve predictions of in vivo success. Here, a ligand binding assay using fluorescence anisotropy was implemented to successfully determine on rates (kon) and off rates (koff) of labeled and unlabeled ligands binding to the adenosine A2A receptor (A2AR) purified into nanodiscs (A2AR-NDs). The kinetic assay was used to determine the optimal storage conditions of A2AR-NDs, where they were found to be stable for more than 6 months at -80°C. The binding assay was implemented to further understand receptor function by determining the effects of charged lipids on agonist binding kinetics, how sodium levels allosterically modulate A2AR function, and how A2AR protonation affects agonist binding.
Collapse
Affiliation(s)
- John M Pettersen
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Olivia McCracken
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Anne Skaja Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
16
|
Pușcașu C, Andrei C, Olaru OT, Zanfirescu A. Metabolite-Sensing Receptors: Emerging Targets for Modulating Chronic Pain Pathways. Curr Issues Mol Biol 2025; 47:63. [PMID: 39852178 PMCID: PMC11763455 DOI: 10.3390/cimb47010063] [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: 12/24/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Chronic pain is a debilitating condition affecting millions worldwide, often resulting from complex interactions between the nervous and immune systems. Recent advances highlight the critical role of metabolite-sensing G protein-coupled receptors (GPCRs) in various chronic pain types. These receptors link metabolic changes with cellular responses, influencing inflammatory and degenerative processes. Receptors such as free fatty acid receptor 1 (FFAR1/GPR40), free fatty acid receptor 4 (FFAR4/GPR120), free fatty acid receptor 2 (FFAR2/GPR43), and Takeda G protein-coupled receptor 5 (TGR5/GPR131/GPBAR1) are key modulators of nociceptive signaling. GPR40, activated by long-chain fatty acids, exhibits strong anti-inflammatory effects by reducing cytokine expression. Butyrate-activated GPR43 inhibits inflammatory mediators like nitric oxide synthase-2 and cyclooxygenase-2, mitigating inflammation. TGR5, activated by bile acids, regulates inflammation and cellular senescence through pathways like NF-κB and p38. These receptors are promising therapeutic targets in chronic pain, addressing the metabolic and inflammatory factors underlying nociceptive sensitization and tissue degeneration. This review explores the molecular mechanisms of metabolite-sensing receptors in chronic pain, their therapeutic potential, and challenges in clinical application. By uncovering these mechanisms, metabolite-sensing receptors could lead to safer, more effective pain management strategies.
Collapse
Affiliation(s)
| | - Corina Andrei
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (O.T.O.); (A.Z.)
| | | | | |
Collapse
|
17
|
Ma Y, Wang Y, Tang M, Weng Y, Chen Y, Xu Y, An S, Wu Y, Zhao S, Xu H, Li D, Liu M, Lu W, Ru H, Song G. Cryo-EM structure of an activated GPR4-Gs signaling complex. Nat Commun 2025; 16:605. [PMID: 39799123 PMCID: PMC11724869 DOI: 10.1038/s41467-025-55901-2] [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: 06/05/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025] Open
Abstract
G protein-coupled receptor 4 (GPR4) belongs to the subfamily of proton-sensing GPCRs (psGPCRs), which detect pH changes in extracellular environment and regulate diverse physiological responses. GPR4 was found to be overactivated in acidic tumor microenvironment as well as inflammation sites, with a triad of acidic residues within the transmembrane domain identified as crucial for proton sensing. However, the 3D structure remains unknown, and the roles of other conserved residues within psGPCRs are not well understood. Here we report cryo-electron microscopy (cryo-EM) structures of active zebrafish GPR4 at both pH 6.5 and 8.5, each highlighting a distribution of histidine and acidic residues at the extracellular region. Cell-based assays show that these ionizable residues moderately influence the proton-sensing capacity of zebrafish GPR4, compared to the more significant effects of the triad residues. Furthermore, we reveal a cluster of aromatic residues within the orthosteric pocket that may propagate the signaling to the intercellular region via repacking the aromatic patch at the central region. This study provides a framework for future signaling and functional investigation of psGPCRs.
Collapse
Affiliation(s)
- Yitong Ma
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yijie Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mengyuan Tang
- Life Sciences Institute, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Weng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ying Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yueming Xu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Shuxiao An
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Huanhuan Xu
- College of Science, Yunnan Agricultural University, Kunming, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Weiqiang Lu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Heng Ru
- Life Sciences Institute, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Gaojie Song
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
18
|
Tyson AS, Khan S, Motiwala Z, Han GW, Zhang Z, Ranjbar M, Styrpejko D, Ramos-Gonzalez N, Woo S, Villers K, Landaker D, Kenakin T, Shenvi R, Majumdar S, Gati C. Molecular mechanisms of inverse agonism via κ-opioid receptor-G protein complexes. Nat Chem Biol 2025:10.1038/s41589-024-01812-0. [PMID: 39775170 DOI: 10.1038/s41589-024-01812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
Opioid receptors, a subfamily of G protein-coupled receptors (GPCRs), are key therapeutic targets. In the canonical GPCR activation model, agonist binding is required for receptor-G protein complex formation, while antagonists prevent G protein coupling. However, many GPCRs exhibit basal activity, allowing G protein association without an agonist. The pharmacological impact of agonist-free receptor-G protein complexes is poorly understood. Here we present biochemical evidence that certain κ-opioid receptor (KOR) inverse agonists can act via KOR-Gi protein complexes. To investigate this phenomenon, we determined cryo-EM structures of KOR-Gi protein complexes with three inverse agonists: JDTic, norBNI and GB18, corresponding to structures of inverse agonist-bound GPCR-G protein complexes. Remarkably, the orthosteric binding pocket resembles the G protein-free 'inactive' receptor conformation, while the receptor remains coupled to the G protein. In summary, our work challenges the canonical model of receptor antagonism and offers crucial insights into GPCR pharmacology.
Collapse
Affiliation(s)
- Aaliyah S Tyson
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Saif Khan
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Zenia Motiwala
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
- Amgen Inc., Thousand Oaks, CA, USA
| | - Gye Won Han
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Zixin Zhang
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mohsen Ranjbar
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Daniel Styrpejko
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Nokomis Ramos-Gonzalez
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, USA
| | - Stone Woo
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Kelly Villers
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Delainey Landaker
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Terry Kenakin
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Ryan Shenvi
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, USA
| | - Cornelius Gati
- The Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA.
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Thomson NJ, Zachariae U. Mechanism of negative μ-opioid receptor modulation by sodium ions. Structure 2025; 33:196-205.e2. [PMID: 39536757 DOI: 10.1016/j.str.2024.10.023] [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: 06/22/2023] [Revised: 08/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Negative allosteric modulation of G-protein coupled receptors (GPCRs) by Na+ ions was first described in the 1970s for opioid receptors (ORs) and has subsequently been detected for most class A GPCRs. In high-resolution structures of inactive-state class A GPCRs, a Na+ ion binds to a conserved pocket near residue D2.50, whereas active-state structures of GPCRs are incompatible with Na+ binding. Correspondingly, Na+ diminishes agonist affinity, stabilizes the receptors in the inactive state, and reduces basal signaling. We applied a mutual-information based analysis to μs-timescale biomolecular simulations of the μ-opioid receptor (μ-OR). Our results reveal that Na+ binding is coupled to a water wire linking the Na+ binding site with the agonist binding pocket and to rearrangements in polar networks propagating conformational changes to the agonist and G-protein binding sites. These findings provide a new mechanistic link between the presence of the ion, altered agonist affinity, receptor deactivation, and lowered basal signaling levels.
Collapse
Affiliation(s)
- Neil J Thomson
- Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Ulrich Zachariae
- Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK; Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK.
| |
Collapse
|
20
|
Glover H, Saßmannshausen T, Bertrand Q, Trabuco M, Slavov C, Bacchin A, Andres F, Kondo Y, Stipp R, Wranik M, Khusainov G, Carrillo M, Kekilli D, Nan J, Gonzalez A, Cheng R, Neidhart W, Weinert T, Leonarski F, Dworkowski F, Kepa M, Wachtveitl J, Hennig M, Standfuss J. Photoswitch dissociation from a G protein-coupled receptor resolved by time-resolved serial crystallography. Nat Commun 2024; 15:10837. [PMID: 39738009 PMCID: PMC11686364 DOI: 10.1038/s41467-024-55109-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors in humans. The binding and dissociation of ligands tunes the inherent conformational flexibility of these important drug targets towards distinct functional states. Here we show how to trigger and resolve protein-ligand interaction dynamics within the human adenosine A2A receptor. For this, we designed seven photochemical affinity switches derived from the anti-Parkinson's drug istradefylline. In a rational approach based on UV/Vis spectroscopy, time-resolved absorption spectroscopy, differential scanning fluorimetry and cryo-crystallography, we identified compounds suitable for time-resolved serial crystallography. Our analysis of millisecond-scale dynamics revealed how trans-to-cis isomerization shifts selected istradefylline derivatives within the binding pocket. Depending on the chemical nature of the ligand, interactions between extracellular loops 2 and 3, acting as a lid on the binding pocket, are disrupted and rearrangement of the orthosteric binding pocket is invoked upon ligand dissociation. This innovative approach provides insights into GPCR dynamics at the atomic level, offering potential for developing novel pharmaceuticals.
Collapse
Affiliation(s)
- Hannah Glover
- PSI Center for Life Sciences, Villigen PSI, Switzerland
| | - Torben Saßmannshausen
- Institute of Physical and Theoretical Chemistry, Goethe University, Frankfurt, Germany
| | | | | | - Chavdar Slavov
- Institute of Physical and Theoretical Chemistry, Goethe University, Frankfurt, Germany
- Department of Chemistry, University of South Florida, Tampa, USA
| | | | - Fabio Andres
- leadXpro AG, Park Innovaare, Villigen PSI, Switzerland
| | - Yasushi Kondo
- PSI Center for Life Sciences, Villigen PSI, Switzerland
| | - Robin Stipp
- PSI Center for Life Sciences, Villigen PSI, Switzerland
| | | | | | | | - Demet Kekilli
- PSI Center for Life Sciences, Villigen PSI, Switzerland
| | - Jie Nan
- MaxIV Laboratory, Lund University, Lund, Sweden
| | | | - Robert Cheng
- leadXpro AG, Park Innovaare, Villigen PSI, Switzerland
| | | | | | | | | | - Michal Kepa
- PSI Center for Life Sciences, Villigen PSI, Switzerland
| | - Josef Wachtveitl
- Institute of Physical and Theoretical Chemistry, Goethe University, Frankfurt, Germany
| | | | | |
Collapse
|
21
|
Rinne A, Bünemann M. Charge Movements and Conformational Changes: Biophysical Properties and Physiology of Voltage-Dependent GPCRs. Biomolecules 2024; 14:1652. [PMID: 39766359 PMCID: PMC11674552 DOI: 10.3390/biom14121652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
G protein-coupled receptors (GPCRs) regulate multiple cellular functions and represent important drug targets. More than 20 years ago, it was noted that GPCR activation (agonist binding) and signaling (G protein activation) are dependent on the membrane potential (VM). While it is now proven that many GPCRs display an intrinsic voltage dependence, the molecular mechanisms of how GPCRs sense depolarization of the plasma membrane are less well defined. This review summarizes the current knowledge of voltage-dependent signaling in GPCRs. We describe how voltage dependence was discovered in muscarinic receptors, present an overview of GPCRs that are regulated by voltage, and show how biophysical properties of GPCRs led to the discovery of voltage-sensing mechanisms in those receptors. Furthermore, we summarize physiological functions that have been shown to be regulated by voltage-dependent GPCR signaling of endogenous receptors in excitable tissues, such as the nervous system or the heart. Finally, we discuss challenges that remain in analyzing voltage-dependent signaling of GPCRs in vivo and present an outlook on experimental applications of the interesting concept of GPCR signaling.
Collapse
Affiliation(s)
- Andreas Rinne
- Department of Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | - Moritz Bünemann
- Institute of Pharmacology and Clinical Pharmacy, Biochemical Pharmaceutical Center (BPC) Marburg, University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
22
|
Seyedabadi M, Gurevich VV. Flavors of GPCR signaling bias. Neuropharmacology 2024; 261:110167. [PMID: 39306191 DOI: 10.1016/j.neuropharm.2024.110167] [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: 04/23/2024] [Revised: 08/06/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
GPCRs are inherently flexible molecules existing in an equilibrium of multiple conformations. Binding of GPCR agonists shifts this equilibrium. Certain agonists can increase the fraction of active-like conformations that predispose the receptor to coupling to a particular signal transducer or a select group of transducers. Such agonists are called biased, in contrast to balanced agonists that facilitate signaling via all transducers the receptor couples to. These biased agonists preferentially channel the signaling of a GPCR to particular G proteins, GRKs, or arrestins. Preferential activation of particular G protein or arrestin subtypes can be beneficial, as it would reduce unwanted on-target side effects, widening the therapeutic window. However, biasing GPCRs has two important limitations: a) complete bias is impossible due to inherent flexibility of GPCRs; b) receptor-independent functions of signal transducer proteins cannot be directly affected by GPCR ligands or differential receptor barcoding by GRK phosphorylation. This article is part of the Special Issue on "Ligand Bias".
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave South, PRB, Rm. 417D, Nashville, TN, 37232, USA.
| |
Collapse
|
23
|
Barekatain M, Johansson LC, Lam JH, Chang H, Sadybekov AV, Han GW, Russo J, Bliesath J, Brice N, Carlton MBL, Saikatendu KS, Sun H, Murphy ST, Monenschein H, Schiffer HH, Popov P, Lutomski CA, Robinson CV, Liu ZJ, Hua T, Katritch V, Cherezov V. Structural insights into the high basal activity and inverse agonism of the orphan receptor GPR6 implicated in Parkinson's disease. Sci Signal 2024; 17:eado8741. [PMID: 39626010 PMCID: PMC11850111 DOI: 10.1126/scisignal.ado8741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 11/07/2024] [Indexed: 02/06/2025]
Abstract
GPR6 is an orphan G protein-coupled receptor with high constitutive activity found in D2-type dopamine receptor-expressing medium spiny neurons of the striatopallidal pathway, which is aberrantly hyperactivated in Parkinson's disease. Here, we solved crystal structures of GPR6 without the addition of a ligand (a pseudo-apo state) and in complex with two inverse agonists, including CVN424, which improved motor symptoms in patients with Parkinson's disease in clinical trials. In addition, we obtained a cryo-electron microscopy structure of the signaling complex between GPR6 and its cognate Gs heterotrimer. The pseudo-apo structure revealed a strong density in the orthosteric pocket of GPR6 corresponding to a lipid-like endogenous ligand. A combination of site-directed mutagenesis, native mass spectrometry, and computer modeling suggested potential mechanisms for high constitutive activity and inverse agonism in GPR6 and identified a series of lipids and ions bound to the receptor. The structures and results obtained in this study could guide the rational design of drugs that modulate GPR6 signaling.
Collapse
Affiliation(s)
- Mahta Barekatain
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Linda C. Johansson
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Jordy H. Lam
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Hao Chang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Anastasiia V. Sadybekov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Gye Won Han
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Joseph Russo
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | - Joshua Bliesath
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | | | | | | | - Hukai Sun
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | - Sean T. Murphy
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | | | - Hans H. Schiffer
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | - Petr Popov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Corinne A. Lutomski
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford OX1 3QU, UK
| | - Carol V. Robinson
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford OX1 3QU, UK
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Vsevolod Katritch
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Vadim Cherezov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
24
|
Huang YH, Lin SY, Ou LC, Huang WC, Chao PK, Chang YC, Chang HF, Lee PT, Yeh TK, Kuo YH, Tien YW, Xi JH, Tao PL, Chen PY, Chuang JY, Shih C, Chen CT, Tung CW, Loh HH, Ueng SH, Yeh SH. Discovery of a mu-opioid receptor modulator that in combination with morphinan antagonists induces analgesia. Cell Chem Biol 2024; 31:1885-1898.e10. [PMID: 39025070 DOI: 10.1016/j.chembiol.2024.06.013] [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: 10/02/2023] [Revised: 04/09/2024] [Accepted: 06/22/2024] [Indexed: 07/20/2024]
Abstract
Morphinan antagonists, which block opioid effects at mu-opioid receptors, have been studied for their analgesic potential. Previous studies have suggested that these antagonists elicit analgesia with fewer adverse effects in the presence of the mutant mu-opioid receptor (MOR; S196A). However, introducing a mutant receptor for medical applications represents significant challenges. We hypothesize that binding a chemical compound to the MOR may elicit a comparable effect to the S196A mutation. Through high-throughput screening and structure-activity relationship studies, we identified a modulator, 4-(2-(4-fluorophenyl)-4-oxothiazolidin-3-yl)-3-methylbenzoic acid (BPRMU191), which confers agonistic properties to small-molecule morphinan antagonists, which induce G protein-dependent MOR activation. Co-application of BPRMU191 and morphinan antagonists resulted in MOR-dependent analgesia with diminished side effects, including gastrointestinal dysfunction, antinociceptive tolerance, and physical and psychological dependence. Combining BPRMU191 and morphinan antagonists could serve as a potential therapeutic strategy for severe pain with reduced adverse effects and provide an avenue for studying G protein-coupled receptor modulation.
Collapse
Affiliation(s)
- Yi-Han Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Research Center for Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Shu-Yu Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Li-Chin Ou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Wei-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Po-Kuan Chao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yung-Chiao Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hsiao-Fu Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Pin-Tse Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yu-Hsien Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Ya-Wen Tien
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Jing-Hua Xi
- Department of Pharmacology, Medical School University of Minnesota, Minneapolis, MN 55455-0217, USA
| | - Pao-Luh Tao
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Pin-Yuan Chen
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Chang Gung University, Keelung 20401, Taiwan
| | - Jian-Ying Chuang
- Research Center for Neuroscience, Taipei Medical University, Taipei 110, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Horace H Loh
- Department of Pharmacology, Medical School University of Minnesota, Minneapolis, MN 55455-0217, USA; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.
| | - Shau-Hua Ueng
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| | - Shiu-Hwa Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan.
| |
Collapse
|
25
|
den Hollander LS, Zweemer AJM, Béquignon OJM, Hammerl DM, Bleijs BTM, Veenhuizen M, Lantsheer WJF, Chau B, van Westen GJP, IJzerman AP, Heitman LH. CC chemokine receptor 2 is allosterically modulated by sodium ions and amiloride derivatives through a distinct sodium ion binding site. Biochem Pharmacol 2024; 229:116464. [PMID: 39111604 DOI: 10.1016/j.bcp.2024.116464] [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: 03/06/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024]
Abstract
CC chemokine receptor 2 and CCL2 are highly involved in cancer growth and metastasis, and immune escape. Raised sodium ion concentrations in solid tumours have also been correlated to metastasis and immune modulation. Sodium ions can modulate class A G protein-coupled receptors through the sodium ion binding site characterized by a highly conserved aspartic acid residue (D2.50), also present in CCR2. Hence, we further explored this binding site in CCR2 by radioligand binding studies and mutagenesis. Modulation of three distinctly binding radioligands by sodium ions and amiloride derivates was investigated. Sodium ions were observed to be relatively weak modulators of antagonist binding, but substantially increased 125I-CCL2 dissociation from CCR2. 6-Substituted Hexamethylene Amiloride (HMA) modulated all tested radioligands. Induced-fit docking of HMA in the presumed sodium ion binding site of CCR2 confirmed its binding site. Finally, investigation of (cancer-associated) mutations in the sodium ion binding site showed a markedly decreased expression compared to wild type. Only two mutants, G123A3.35 and G127K3.39, were able to be bound by [3H]INCB3344 and [3H]CCR2-RA-[R]. Thus, mutagenesis showed that the sodium ion binding site residues, which are distinct from other class A GPCRs and related to chemokine receptor evolution, are crucial for receptor integrity. Moreover, the tested mutations appeared to have no effect on modulation observed by HMA or a minor effect on sodium chloride modulation on the tested radioligands. All in all, these results invite further exploration of the CCR2 sodium ion binding site in (cancer) biology, and potentially as a third druggable binding site.
Collapse
Affiliation(s)
- Lisa S den Hollander
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Annelien J M Zweemer
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Olivier J M Béquignon
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Dora M Hammerl
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Bente T M Bleijs
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Margo Veenhuizen
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Wernard J F Lantsheer
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Bobby Chau
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Gerard J P van Westen
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Adriaan P IJzerman
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Laura H Heitman
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands; Oncode Institute, the Netherlands.
| |
Collapse
|
26
|
Hovah ME, Holzgrabe U. Bivalent and bitopic ligands of the opioid receptors: The prospects of a dual approach. Med Res Rev 2024; 44:2545-2599. [PMID: 38751227 DOI: 10.1002/med.22050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 10/05/2024]
Abstract
Opioid receptors belonging to the class A G-protein coupled receptors (GPCRs) are the targets of choice in the treatment of acute and chronic pain. However, their on-target side effects such as respiratory depression, tolerance and addiction have led to the advent of the 'opioid crisis'. In the search for safer analgesics, bivalent and more recently, bitopic ligands have emerged as valuable tool compounds to probe these receptors. The activity of bivalent and bitopic ligands rely greatly on the allosteric nature of the GPCRs. Bivalent ligands consist of two pharmacophores, each binding to the individual orthosteric binding site (OBS) of the monomers within a dimer. Bitopic or dualsteric ligands bridge the gap between the OBS and the spatially distinct, less conserved allosteric binding site (ABS) through the simultaneous occupation of these two sites. Bivalent and bitopic ligands stabilize distinct conformations of the receptors which ultimately translates into unique signalling and pharmacological profiles. Some of the interesting properties shown by these ligands include improved affinity and/or efficacy, subtype and/or functional selectivity and reduced side effects. This review aims at providing an overview of some of the bivalent and bitopic ligands of the opioid receptors and, their pharmacology in the hope of inspiring the design and discovery of the next generation of opioid analgesics.
Collapse
Affiliation(s)
- Marie Emilie Hovah
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| |
Collapse
|
27
|
Wu Z, Sun X, Su J, Zhang X, Hu J, Li C. Revealing the graded activation mechanism of neurotensin receptor 1. Int J Biol Macromol 2024; 278:134488. [PMID: 39111461 DOI: 10.1016/j.ijbiomac.2024.134488] [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: 04/17/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Graded activation contributes to the precise regulation of GPCR activity, presenting new opportunities for drug design. In this work, a total of 10 μs enhanced-sampling simulations are performed to provide molecular insights into the binding dynamics differences of the neurotensin receptor 1 (NTSR1) to the full agonist SRI-9829, partial agonist RTI-3a and inverse agonist SR48692. The possible graded activation mechanism of NTSR1 is revealed by an integrated analysis utilizing the reweighted potential of mean force (PMF), deep learning (DL) and transfer entropy (TE). Specifically, the orthosteric pocket is observed to undergo expansion and contraction, with the G-protein-binding site experiencing interconversions among the inactive, intermediate and active-like states. Detailed structural comparisons capture subtle conformational differences arising from ligand binding in allosteric signaling, which can well explain the graded activation. Critical microswitches that contribute to graded activation are efficiently identified with the DL model. TE calculations enable the visualization of allosteric communication networks within the receptor, elucidating the driver-responder relationships associated with signal transduction. Fortunately, the dissociation of the full agonist from the orthosteric pocket is observed. The current findings systematically reveal the mechanism of NTSR1 graded activation, and also provide implications for structure-based drug design.
Collapse
Affiliation(s)
- Zhixiang Wu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xiaohan Sun
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Jingjie Su
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xinyu Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Jianping Hu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China.
| | - Chunhua Li
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China.
| |
Collapse
|
28
|
Barreto CAV, Vitorino JNM, Reis PBPS, Machuqueiro M, Moreira IS. p Ka Calculations of GPCRs: Understanding Protonation States in Receptor Activation. J Chem Inf Model 2024; 64:6850-6856. [PMID: 39150719 PMCID: PMC11388449 DOI: 10.1021/acs.jcim.4c01125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The increase in the available G protein-coupled receptor (GPCR) structures has been pivotal in helping to understand their activation process. However, the role of protonation-conformation coupling in GPCR activation still needs to be clarified. We studied the protonation behavior of the highly conserved Asp2.50 residue in five different class A GPCRs (active and inactive conformations) using a linear response approximation (LRA) pKa calculation protocol. We observed consistent differences (1.3 pK units) for the macroscopic pKa values between the inactive and active states of the A2AR and B2AR receptors, indicating the protonation of Asp2.50 during GPCR activation. This process seems to be specific and not conserved, as no differences were observed in the pKa values of the remaining receptors (CB1R, NT1R, and GHSR).
Collapse
Affiliation(s)
- Carlos A V Barreto
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
- CNC─Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - João N M Vitorino
- BioSI─Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Pedro B P S Reis
- BioSI─Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Miguel Machuqueiro
- BioSI─Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Irina S Moreira
- CNC─Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
29
|
Ople R, Ramos-Gonzalez N, Li Q, Sobecks BL, Aydin D, Powers AS, Faouzi A, Polacco BJ, Bernhard SM, Appourchaux K, Sribhashyam S, Eans SO, Tsai BA, Dror RO, Varga BR, Wang H, Hüttenhain R, McLaughlin JP, Majumdar S. Signaling Modulation Mediated by Ligand Water Interactions with the Sodium Site at μOR. ACS CENTRAL SCIENCE 2024; 10:1490-1503. [PMID: 39220695 PMCID: PMC11363324 DOI: 10.1021/acscentsci.4c00525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 09/04/2024]
Abstract
The mu opioid receptor (μOR) is a target for clinically used analgesics. However, adverse effects, such as respiratory depression and physical dependence, necessitate the development of alternative treatments. Recently we reported a novel strategy to design functionally selective opioids by targeting the sodium binding allosteric site in μOR with a supraspinally active analgesic named C6guano. Presently, to improve systemic activity of this ligand, we used structure-based design, identifying a new ligand named RO76 where the flexible alkyl linker and polar guanidine guano group is swapped with a benzyl alcohol, and the sodium site is targeted indirectly through waters. A cryoEM structure of RO76 bound to the μOR-Gi complex confirmed that RO76 interacts with the sodium site residues through a water molecule, unlike C6guano which engages the sodium site directly. Signaling assays coupled with APEX based proximity labeling show binding in the sodium pocket modulates receptor efficacy and trafficking. In mice, RO76 was systemically active in tail withdrawal assays and showed reduced liabilities compared to those of morphine. In summary, we show that targeting water molecules in the sodium binding pocket may be an avenue to modulate signaling properties of opioids, and which may potentially be extended to other G-protein coupled receptors where this site is conserved.
Collapse
Affiliation(s)
- Rohini
S. Ople
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Nokomis Ramos-Gonzalez
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Qiongyu Li
- Department
of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California 94158, United States
| | - Briana L. Sobecks
- Department of Computer Science, Stanford
University, Stanford, California 94305, United States
- Department
of Structural Biology, Stanford University
School of Medicine, Stanford, California 94305, United States
- Department
of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Deniz Aydin
- Department of Computer Science, Stanford
University, Stanford, California 94305, United States
- Department
of Structural Biology, Stanford University
School of Medicine, Stanford, California 94305, United States
- Department
of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Alexander S. Powers
- Department of Computer Science, Stanford
University, Stanford, California 94305, United States
- Department
of Structural Biology, Stanford University
School of Medicine, Stanford, California 94305, United States
- Department
of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Abdelfattah Faouzi
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Benjamin J. Polacco
- Department of Computer Science, Stanford
University, Stanford, California 94305, United States
- Department
of Structural Biology, Stanford University
School of Medicine, Stanford, California 94305, United States
| | - Sarah M. Bernhard
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Kevin Appourchaux
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Sashrik Sribhashyam
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shainnel O. Eans
- Department
of Pharmacodynamics, University of Florida, Gainesville, Florida 032610, United
States
| | - Bowen A. Tsai
- Department
of Pharmacodynamics, University of Florida, Gainesville, Florida 032610, United
States
| | - Ron O. Dror
- Department of Computer Science, Stanford
University, Stanford, California 94305, United States
- Department
of Structural Biology, Stanford University
School of Medicine, Stanford, California 94305, United States
- Department
of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Balazs R. Varga
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Haoqing Wang
- Department
of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Ruth Hüttenhain
- Department
of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jay P. McLaughlin
- Department
of Pharmacodynamics, University of Florida, Gainesville, Florida 032610, United
States
| | - Susruta Majumdar
- Center
for Clinical Pharmacology, University of
Health Sciences & Pharmacy at St. Louis and Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
30
|
Distler K, Maschauer S, Neu E, Hübner H, Einsiedel J, Prante O, Gmeiner P. Structure-guided discovery of orexin receptor-binding PET ligands. Bioorg Med Chem 2024; 110:117823. [PMID: 38964170 DOI: 10.1016/j.bmc.2024.117823] [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: 05/03/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Molecular imaging using positron emission tomography (PET) can serve as a promising tool for visualizing biological targets in the brain. Insights into the expression pattern and the in vivo imaging of the G protein-coupled orexin receptors OX1R and OX2R will further our understanding of the orexin system and its role in various physiological and pathophysiological processes. Guided by crystal structures of our lead compound JH112 and the approved hypnotic drug suvorexant bound to OX1R and OX2R, respectively, we herein describe the design and synthesis of two novel radioligands, [18F]KD23 and [18F]KD10. Key to the success of our structural modifications was a bioisosteric replacement of the triazole moiety with a fluorophenyl group. The 19F-substituted analog KD23 showed high affinity for the OX1R and selectivity over OX2R, while the high affinity ligand KD10 displayed similar Ki values for both subtypes. Radiolabeling starting from the respective pinacol ester precursors resulted in excellent radiochemical yields of 93% and 88% for [18F]KD23 and [18F]KD10, respectively, within 20 min. The new compounds will be useful in PET studies aimed at subtype-selective imaging of orexin receptors in brain tissue.
Collapse
Affiliation(s)
- Katharina Distler
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaulallee 12, 91054 Erlangen, Germany
| | - Eduard Neu
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Jürgen Einsiedel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Olaf Prante
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaulallee 12, 91054 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany.
| |
Collapse
|
31
|
Zmajkovicova K, Pawar S, Sharapova SO, Geier CB, Wiest I, Nguyen C, Monticelli H, Maier-Munsa S, Chen K, Sleasman JW, Aleshkevich S, Polyakova E, Sakovich I, Warnatz K, Grimbacher B, Proietti M, Sondheimer N, Ujhazi B, Gordon S, Ellison M, Yilmaz M, Walter JE, Badarau A, Taveras AG, Neff JL, Bledsoe JR, Tarrant TK. A novel transmembrane CXCR4 variant that expands the WHIM genotype-phenotype paradigm. Blood Adv 2024; 8:3754-3759. [PMID: 38768429 PMCID: PMC11296240 DOI: 10.1182/bloodadvances.2023011875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/07/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Affiliation(s)
| | - Sumit Pawar
- Formerly X4 Pharmaceuticals (Austria) GmbH, Vienna, Austria
| | - Svetlana O. Sharapova
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology, Minsk, Belarus
| | - Christoph B. Geier
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ivana Wiest
- Formerly X4 Pharmaceuticals (Austria) GmbH, Vienna, Austria
| | - Chi Nguyen
- X4 Pharmaceuticals (Austria) GmbH, Vienna, Austria
| | | | | | | | - John W. Sleasman
- Division of Allergy, Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Svetlana Aleshkevich
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology, Minsk, Belarus
| | - Ekaterina Polyakova
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology, Minsk, Belarus
| | - Inga Sakovich
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology, Minsk, Belarus
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michele Proietti
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, Hannover, Germany
- RESIST-Cluster of Excellence 2155, Hannover Medical School, Hannover, Germany
| | - Neal Sondheimer
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Boglarka Ujhazi
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Sumai Gordon
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Maryssa Ellison
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Melis Yilmaz
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, FL
- Division of Allergy & Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Jolan E. Walter
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, FL
- Division of Allergy & Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL
- Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA
| | | | | | - Jadee L. Neff
- Division of Hematopathology, Department of Pathology, Duke University, Durham, NC
| | | | - Teresa K. Tarrant
- Division of Rheumatology and Immunology, Department of Medicine, Duke University, Durham, NC
- Durham Veterans Affairs Medical Center, Durham, NC
| |
Collapse
|
32
|
Otun O, Aljamous C, Del Nero E, Arimont-Segura M, Bosma R, Zarzycka B, Girbau T, Leyrat C, de Graaf C, Leurs R, Durroux T, Granier S, Cong X, Bechara C. Conformational dynamics underlying atypical chemokine receptor 3 activation. Proc Natl Acad Sci U S A 2024; 121:e2404000121. [PMID: 39008676 PMCID: PMC11287255 DOI: 10.1073/pnas.2404000121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/28/2024] [Indexed: 07/17/2024] Open
Abstract
Atypical Chemokine Receptor 3 (ACKR3) belongs to the G protein-coupled receptor family but it does not signal through G proteins. The structural properties that govern the functional selectivity and the conformational dynamics of ACKR3 activation are poorly understood. Here, we combined hydrogen/deuterium exchange mass spectrometry, site-directed mutagenesis, and molecular dynamics simulations to examine the binding mode and mechanism of action of ACKR3 ligands of different efficacies. Our results show that activation or inhibition of ACKR3 is governed by intracellular conformational changes of helix 6, intracellular loop 2, and helix 7, while the DRY motif becomes protected during both processes. Moreover, we identified the binding sites and the allosteric modulation of ACKR3 upon β-arrestin 1 binding. In summary, this study highlights the structure-function relationship of small ligands, the binding mode of β-arrestin 1, the activation dynamics, and the atypical dynamic features in ACKR3 that may contribute to its inability to activate G proteins.
Collapse
Affiliation(s)
- Omolade Otun
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Christelle Aljamous
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Elise Del Nero
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Marta Arimont-Segura
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Reggie Bosma
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Barbara Zarzycka
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Tristan Girbau
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Cédric Leyrat
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Chris de Graaf
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Rob Leurs
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Xiaojing Cong
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Cherine Bechara
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
- Institut Universitaire de France, Paris75005, France
| |
Collapse
|
33
|
Che T, Varga B, Bernhard SM, El Daibani A, Zaidi S, Lam J, Aguilar J, Appourchaux K, Nazarova A, Kouvelis A, Eans S, Margolis E, Fay J, Pradhan A, Katritch V, McLaughlin J, Majumdar S. Structure-Guided Design of Partial Agonists at an Opioid Receptor. RESEARCH SQUARE 2024:rs.3.rs-4664764. [PMID: 39070616 PMCID: PMC11276012 DOI: 10.21203/rs.3.rs-4664764/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The persistence of chronic pain and continuing overdose deaths from pain-relieving opioids targeting μ opioid receptor (μOR) have fueled the need for reliable long-term analgesics which use different targets and mechanisms. The δ opioid receptor (δOR) is a potential alternative target for non-addictive analgesics to alleviate chronic pain, made more attractive by its lack of respiratory depression associated with μOR agonists. However, early δOR full agonists were found to induce seizures, precluding clinical use. Partial δOR agonists may offer more controlled activation of the receptor compared to full agonists, but the development of such ligands has been hindered by uncertainty over the molecular mechanism mediating partial agonism. Using a structure-based approach, we explored the engagement of the sodium binding pocket in δOR and developed a bitopic ligand, C6-Quino, predicted to be a selective δOR partial agonist. Functional studies of C6-Quino revealed that it displayed δOR partial agonist activity at both G-protein and arrestin pathways. Its interaction with the sodium pocket was confirmed and analyzed using a single particle cryo-EM. Additionally, C6-Quino demonstrated favorable chemical and physiological properties like oral activity, and analgesic activity in multiple chronic pain models. Notably, μOR-related hyperlocomotion and respiratory depression, and δOR-related convulsions, were not observed at analgesic doses of C6-Quino. This fundamentally new approach to designing δOR ligands provides a blueprint for the development of partial agonists as safe analgesics and acts as a generic method to optimize signaling profiles of other Class A GPCRs.
Collapse
Affiliation(s)
- Tao Che
- Washington University in St. Louis
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Iseppon F, Kanellopoulos AH, Tian N, Zhou J, Caan G, Chiozzi R, Thalassinos K, Çubuk C, Lewis MJ, Cox JJ, Zhao J, Woods CG, Wood JN. Sodium channels Na v1.7, Na v1.8 and pain; two distinct mechanisms for Na v1.7 null analgesia. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100168. [PMID: 39559752 PMCID: PMC11570969 DOI: 10.1016/j.ynpai.2024.100168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/20/2024]
Abstract
Genetic deletion and pharmacological inhibition are distinct approaches to unravelling pain mechanisms, identifying targets and developing new analgesics. Both approaches have been applied to the voltage-gated sodium channels Nav1.7 and Nav1.8. Genetic deletion of Nav1.8 in mice leads to a loss of pain and antagonists are effective analgesics. The situation with Nav1.7 is more complex. Complete embryonic loss of Nav1.7 in humans or in mouse sensory neurons leads to anosmia as well as profound analgesia as a result of diminished neurotransmitter release. This is mediated by enhanced endogenous opioid signaling in humans and mice. In contrast, anosmia is opioid-independent. Sensory neuron excitability and autonomic function appear to be normal. Adult deletion of Nav1.7 in sensory neurons also leads to analgesia, but through diminished sensory and autonomic neuron excitability. There is no opioid component of analgesia or anosmia as shown by a lack of effect of naloxone. Pharmacological inhibition of Nav1.7 in mice and humans leads both to analgesia and dramatic side-effects on the autonomic nervous system with no therapeutic window. These data demonstrate that specific Nav1.7 channel blockers will fail as analgesic drugs. The viability of embryonic null mutants suggests that there are compensatory changes to replace the lost Nav1.7 channel. Here we show that sensory neuron sodium channels Nav1.1, Nav1.2 and β4 subunits detected by Mass Spectrometry are upregulated in Nav1.7 embryonic null neurons and, together with other proteome changes, potentially compensate for the loss of Nav1.7. Interestingly, many of the upregulated proteins are known to interact with Nav1.7.
Collapse
Affiliation(s)
- Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Alexandros H. Kanellopoulos
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Naxi Tian
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Jun Zhou
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Gozde Caan
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Riccardo Chiozzi
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
- University College London Mass Spectrometry Science Technology Platform, Division of Biosciences, University College London, London, UK
| | - Konstantinos Thalassinos
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
- University College London Mass Spectrometry Science Technology Platform, Division of Biosciences, University College London, London, UK
| | - Cankut Çubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Myles J. Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - James J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Christopher G. Woods
- Cambridge Institute for Medical Research, Keith Peters Building, Biomedical Campus, Hills Rd, Cambridge CB2 0XY, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
35
|
Patel K, Smith NJ. Primary cilia, A-kinase anchoring proteins and constitutive activity at the orphan G protein-coupled receptor GPR161: A tale about a tail. Br J Pharmacol 2024; 181:2182-2196. [PMID: 36772847 DOI: 10.1111/bph.16053] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/22/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023] Open
Abstract
Primary cilia are non-motile antennae-like structures responsible for sensing environmental changes in most mammalian cells. Ciliary signalling is largely mediated by the Sonic Hedgehog (Shh) pathway, which acts as a master regulator of ciliary protein transit and is essential for normal embryonic development. One particularly important player in primary cilia is the orphan G protein-coupled receptor, GPR161. In this review, we introduce GPR161 in the context of Shh signalling and describe the unique features on its C-terminus such as PKA phosphorylation sites and an A-kinase anchoring protein motif, which may influence the function of the receptor, cAMP compartmentalisation and/or trafficking within primary cilia. We discuss the recent putative pairing of GPR161 and spexin-1, highlighting the additional steps needed before GPR161 could be considered 'deorphanised'. Finally, we speculate that the marked constitutive activity and unconventional regulation of GPR161 may indicate that the receptor may not require an endogenous ligand. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Kinjal Patel
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, New South Wales, Australia
| | - Nicola J Smith
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, New South Wales, Australia
| |
Collapse
|
36
|
Tauber M, Ben-Chaim Y. Voltage Sensors Embedded in G Protein-Coupled Receptors. Int J Mol Sci 2024; 25:5295. [PMID: 38791333 PMCID: PMC11120775 DOI: 10.3390/ijms25105295] [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: 04/18/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Some signaling processes mediated by G protein-coupled receptors (GPCRs) are modulated by membrane potential. In recent years, increasing evidence that GPCRs are intrinsically voltage-dependent has accumulated. A recent publication challenged the view that voltage sensors are embedded in muscarinic receptors. Herein, we briefly discuss the evidence that supports the notion that GPCRs themselves are voltage-sensitive proteins and an alternative mechanism that suggests that voltage-gated sodium channels are the voltage-sensing molecules involved in such processes.
Collapse
Affiliation(s)
| | - Yair Ben-Chaim
- Department of Natural Sciences, The Open University of Israel, Ra’anana 4353701, Israel
| |
Collapse
|
37
|
Pirona L, Ballabio F, Alfonso-Prieto M, Capelli R. Calcium-Driven In Silico Inactivation of a Human Olfactory Receptor. J Chem Inf Model 2024; 64:2971-2978. [PMID: 38523266 DOI: 10.1021/acs.jcim.4c00249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Conformational changes as well as molecular determinants related to the activation and inactivation of olfactory receptors are still poorly understood due to the intrinsic difficulties in the structural determination of this GPCR family. Here, we perform, for the first time, the in silico inactivation of human olfactory receptor OR51E2, highlighting the possible role of calcium in this receptor state transition. Using molecular dynamics simulations, we show that a divalent ion in the ion binding site, coordinated by two acidic residues at positions 2.50 and 3.39 conserved across most ORs, stabilizes the receptor in its inactive state. In contrast, protonation of the same two acidic residues is not sufficient to drive inactivation within the microsecond timescale of our simulations. Our findings suggest a novel molecular mechanism for OR inactivation, potentially guiding experimental validation and offering insights into the possible broader role of divalent ions in GPCR signaling.
Collapse
Affiliation(s)
- Lorenza Pirona
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, I-20133 Milano, Italy
| | - Federico Ballabio
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, I-20133 Milano, Italy
| | - Mercedes Alfonso-Prieto
- Computational Biomedicine, Institute for Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, D-54248 Jülich, Germany
| | - Riccardo Capelli
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, I-20133 Milano, Italy
| |
Collapse
|
38
|
Bertalan É, Rodrigues MJ, Schertler GFX, Bondar AN. Graph-based algorithms to dissect long-distance water-mediated H-bond networks for conformational couplings in GPCRs. Br J Pharmacol 2024. [PMID: 38636539 DOI: 10.1111/bph.16387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/03/2024] [Accepted: 03/02/2024] [Indexed: 04/20/2024] Open
Abstract
Changes in structure and dynamics elicited by agonist ligand binding at the extracellular side of G protein coupled receptors (GPCRs) must be relayed to the cytoplasmic G protein binding side of the receptors. To decipher the role of water-mediated hydrogen-bond networks in this relay mechanism, we have developed graph-based algorithms and analysis methodologies applicable to datasets of static structures of distinct GPCRs. For a reference dataset of static structures of bovine rhodopsin solved at the same resolution, we show that graph analyses capture the internal protein-water hydrogen-bond network. The extended analyses of static structures of rhodopsins and opioid receptors suggest a relay mechanism whereby inactive receptors have in place much of the internal core hydrogen-bond network required for long-distance relay of structural change, with extensive local H-bond clusters observed in structures solved at high resolution and with internal water molecules.
Collapse
Affiliation(s)
- Éva Bertalan
- Physikzentrum, RWTH-Aachen University, Aachen, Germany
| | | | | | - Ana-Nicoleta Bondar
- Forschungszentrum Jülich, Institute of Computational Biomedicine, Jülich, Germany
- Faculty of Physics, University of Bucharest, Măgurele, Romania
| |
Collapse
|
39
|
Gleixner J, Kopanchuk S, Grätz L, Tahk MJ, Laasfeld T, Veikšina S, Höring C, Gattor AO, Humphrys LJ, Müller C, Archipowa N, Köckenberger J, Heinrich MR, Kutta RJ, Rinken A, Keller M. Illuminating Neuropeptide Y Y 4 Receptor Binding: Fluorescent Cyclic Peptides with Subnanomolar Binding Affinity as Novel Molecular Tools. ACS Pharmacol Transl Sci 2024; 7:1142-1168. [PMID: 38633582 PMCID: PMC11019746 DOI: 10.1021/acsptsci.4c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 04/19/2024]
Abstract
The neuropeptide Y (NPY) Y4 receptor (Y4R), a member of the family of NPY receptors, is physiologically activated by the linear 36-amino acid peptide pancreatic polypeptide (PP). The Y4R is involved in the regulation of various biological processes, most importantly pancreatic secretion, gastrointestinal motility, and regulation of food intake. So far, Y4R binding affinities have been mostly studied in radiochemical binding assays. Except for a few fluorescently labeled PP derivatives, fluorescence-tagged Y4R ligands with high affinity have not been reported. Here, we introduce differently fluorescence-labeled (Sulfo-Cy5, Cy3B, Py-1, Py-5) Y4R ligands derived from recently reported cyclic hexapeptides showing picomolar Y4R binding affinity. With pKi values of 9.22-9.71 (radioligand competition binding assay), all fluorescent ligands (16-19) showed excellent Y4R affinity. Y4R saturation binding, binding kinetics, and competition binding with reference ligands were studied using different fluorescence-based methods: flow cytometry (Sulfo-Cy5, Cy3B, and Py-1 label), fluorescence anisotropy (Cy3B label), and NanoBRET (Cy3B label) binding assays. These experiments confirmed the high binding affinity to Y4R (equilibrium pKd: 9.02-9.9) and proved the applicability of the probes for fluorescence-based Y4R competition binding studies and imaging techniques such as single-receptor molecule tracking.
Collapse
Affiliation(s)
- Jakob Gleixner
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Sergei Kopanchuk
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Lukas Grätz
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Maris-Johanna Tahk
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Tõnis Laasfeld
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Santa Veikšina
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Carina Höring
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Albert O. Gattor
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Laura J. Humphrys
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Christoph Müller
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Nataliya Archipowa
- Institute
of Biophysics and Physical Biochemistry, Faculty of Biology and Preclinical
Medicine, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| | - Johannes Köckenberger
- Department
of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Markus R. Heinrich
- Department
of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Roger Jan Kutta
- Institute
of Physical and Theoretical Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Ago Rinken
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Max Keller
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, Germany
| |
Collapse
|
40
|
Zhang M, Chen T, Lu X, Lan X, Chen Z, Lu S. G protein-coupled receptors (GPCRs): advances in structures, mechanisms, and drug discovery. Signal Transduct Target Ther 2024; 9:88. [PMID: 38594257 PMCID: PMC11004190 DOI: 10.1038/s41392-024-01803-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of human membrane proteins and an important class of drug targets, play a role in maintaining numerous physiological processes. Agonist or antagonist, orthosteric effects or allosteric effects, and biased signaling or balanced signaling, characterize the complexity of GPCR dynamic features. In this study, we first review the structural advancements, activation mechanisms, and functional diversity of GPCRs. We then focus on GPCR drug discovery by revealing the detailed drug-target interactions and the underlying mechanisms of orthosteric drugs approved by the US Food and Drug Administration in the past five years. Particularly, an up-to-date analysis is performed on available GPCR structures complexed with synthetic small-molecule allosteric modulators to elucidate key receptor-ligand interactions and allosteric mechanisms. Finally, we highlight how the widespread GPCR-druggable allosteric sites can guide structure- or mechanism-based drug design and propose prospects of designing bitopic ligands for the future therapeutic potential of targeting this receptor family.
Collapse
Affiliation(s)
- Mingyang Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xun Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaobing Lan
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziqiang Chen
- Department of Orthopedics, Changhai Hospital, Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Shaoyong Lu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
41
|
Chen J, Gou Q, Chen X, Song Y, Zhang F, Pu X. Exploring biased activation characteristics by molecular dynamics simulation and machine learning for the μ-opioid receptor. Phys Chem Chem Phys 2024; 26:10698-10710. [PMID: 38512140 DOI: 10.1039/d3cp05050e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Biased ligands selectively activating specific downstream signaling pathways (termed as biased activation) exhibit significant therapeutic potential. However, the conformational characteristics revealed are very limited for the biased activation, which is not conducive to biased drug development. Motivated by the issue, we combine extensive accelerated molecular dynamics simulations and an interpretable deep learning model to probe the biased activation features for two complex systems constructed by the inactive μOR and two different biased agonists (G-protein-biased agonist TRV130 and β-arrestin-biased agonist endomorphin2). The results indicate that TRV130 binds deeper into the receptor core compared to endomorphin2, located between W2936.48 and D1142.50, and forms hydrogen bonding with D1142.50, while endomorphin2 binds above W2936.48. The G protein-biased agonist induces greater outward movements of the TM6 intracellular end, forming a typical active conformation, while the β-arrestin-biased agonist leads to a smaller extent of outward movements of TM6. Compared with TRV130, endomorphin2 causes more pronounced inward movements of the TM7 intracellular end and more complex conformational changes of H8 and ICL1. In addition, important residues determining the two different biased activation states were further identified by using an interpretable deep learning classification model, including some common biased activation residues across Class A GPCRs like some key residues on the TM2 extracellular end, ECL2, TM5 intracellular end, TM6 intracellular end, and TM7 intracellular end, and some specific important residues of ICL3 for μOR. The observations will provide valuable information for understanding the biased activation mechanism for GPCRs.
Collapse
Affiliation(s)
- Jianfang Chen
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Qiaoling Gou
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Xin Chen
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Yuanpeng Song
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Fuhui Zhang
- Graduate School, Sichuan University, Chengdu 610064, China
| | - Xuemei Pu
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
42
|
Imamura K, Akagi KI, Miyanoiri Y, Tsujimoto H, Hirokawa T, Ashida H, Murakami K, Inoue A, Suno R, Ikegami T, Sekiyama N, Iwata S, Kobayashi T, Tochio H. Interaction modes of human orexin 2 receptor with selective and nonselective antagonists studied by NMR spectroscopy. Structure 2024; 32:352-361.e5. [PMID: 38194963 DOI: 10.1016/j.str.2023.12.008] [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: 02/24/2023] [Revised: 08/17/2023] [Accepted: 12/13/2023] [Indexed: 01/11/2024]
Abstract
Orexin neuropeptides have many physiological roles in the sleep-wake cycle, feeding behavior, reward demands, and stress responses by activating cognitive receptors, the orexin receptors (OX1R and OX2R), distributed in the brain. There are only subtle differences between OX1R and OX2R in the orthosteric site, which has hindered the rational development of subtype-selective antagonists. In this study, we utilized solution-state NMR to capture the structural plasticity of OX2R labeled with 13CH3-ε-methionine in complex with antagonists. Mutations in the orthosteric site allosterically affected the intracellular tip of TM6. Ligand exchange experiments with the subtype-selective EMPA and the nonselective suvorexant identified three methionine residues that were substantially perturbed. The NMR spectra suggested that the suvorexant-bound state exhibited more structural plasticity than the EMPA-bound state, which has not been foreseen from the close similarity of their crystal structures, providing insights into dynamic features to be considered in understanding the ligand recognition mode.
Collapse
Affiliation(s)
- Kayo Imamura
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Ken-Ichi Akagi
- Section of Laboratory Equipment, National Institute of Biomedical Innovation, Health, and Nutrition, Osaka 567-0085, Japan
| | - Yohei Miyanoiri
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hirokazu Tsujimoto
- Department of Cell Biology and Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Takatsugu Hirokawa
- Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Hideo Ashida
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Kaori Murakami
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Ryoji Suno
- Department of Medical Chemistry, Kansai Medical University, Hirakata 573-1010, Japan
| | - Takahisa Ikegami
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro, Yokohama, Kanagawa 230-0045, Japan
| | - Naotaka Sekiyama
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - So Iwata
- Department of Cell Biology and Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Kobayashi
- Department of Medical Chemistry, Kansai Medical University, Hirakata 573-1010, Japan
| | - Hidehito Tochio
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan.
| |
Collapse
|
43
|
Wu C, Xu M, Dong J, Cui W, Yuan S. The structure and function of olfactory receptors. Trends Pharmacol Sci 2024; 45:268-280. [PMID: 38296675 DOI: 10.1016/j.tips.2024.01.004] [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: 11/09/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024]
Abstract
Olfactory receptors (ORs) form the most important chemosensory receptor family responsible for our sense of smell in the nasal olfactory epithelium. This receptor family belongs to the class A G protein-coupled receptors (GPCRs). Recent research has indicated that ORs are involved in many nonolfactory physiological processes in extranasal tissue, such as the brain, pancreas, and testes, and implies the possible role of their dysregulation in various diseases. The recently released structures of OR51E2 and consensus OR52 have also unveiled the uniqueness of ORs from other class A GPCR members. In this review, we discuss these recent developments and computational modeling efforts toward understanding the structural properties of unresolved ORs, which could guide potential future OR-targeted drug discovery.
Collapse
Affiliation(s)
- Chenyang Wu
- The AlphaMol-SIAT Joint Laboratory, Shenzhen 518055, China; The Research Center for Computer-aided Drug Discovery, The Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Marc Xu
- The AlphaMol-SIAT Joint Laboratory, Shenzhen 518055, China; The Research Center for Computer-aided Drug Discovery, The Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junlin Dong
- The AlphaMol-SIAT Joint Laboratory, Shenzhen 518055, China; The Research Center for Computer-aided Drug Discovery, The Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenqiang Cui
- The AlphaMol-SIAT Joint Laboratory, Shenzhen 518055, China
| | - Shuguang Yuan
- The AlphaMol-SIAT Joint Laboratory, Shenzhen 518055, China; AlphaMol Science Ltd, Shenzhen 518055, China.
| |
Collapse
|
44
|
Peña KA, Savransky S, Lewis B. Endosomal signaling via cAMP in parathyroid hormone (PTH) type 1 receptor biology. Mol Cell Endocrinol 2024; 581:112107. [PMID: 37981188 DOI: 10.1016/j.mce.2023.112107] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Compartmentalization of GPCR signaling is an emerging topic that highlights the physiological relevance of spatial bias in signaling. The parathyroid hormone (PTH) type 1 receptor (PTH1R) was the first GPCR described to signal via heterotrimeric G-protein and cAMP from endosomes after β-arrestin mediated internalization, challenging the canonical GPCR signaling model which established that signaling is terminated by receptor internalization. More than a decade later, many other GPCRs have been shown to signal from endosomes via cAMP, and recent studies have proposed that location of cAMP generation impacts physiological outcomes of GPCR signaling. Here, we review the extensive literature regarding PTH1R endosomal signaling via cAMP, the mechanisms that regulate endosomal generation of cAMP, and the implications of spatial bias in PTH1R physiological functions.
Collapse
Affiliation(s)
- Karina A Peña
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Sofya Savransky
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Breanna Lewis
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Helfinger L, Tate CG. Structures of Adrenoceptors. Handb Exp Pharmacol 2024; 285:13-26. [PMID: 37460660 DOI: 10.1007/164_2023_674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The first structure of an adrenoceptor (AR), the human β2-adrenoceptor (hβ2AR) was published in 2007 and since then a total of 78 structures (up to June 2022) have been determined by X-ray crystallography and electron cryo-microscopy (cryo-EM) of all three βARs (β1, β2 and β3) and four out of six αARs (α1B, α2A, α2B, α2C). The structures are in a number of different conformational states, including the inactive state bound to an antagonist, an intermediate state bound to agonist and active states bound to agonist and an intracellular transducer (G protein or arrestin) or transducer mimetic (nanobody). The structures identify molecular details of how ligands bind in the orthosteric binding pocket (OBP; 19 antagonists, 18 agonists) and also how three different small molecule allosteric modulators bind. The structures have been used to define the molecular details of receptor activation and also the molecular determinants for transducer coupling. This chapter will give a brief overview of the structures, receptor activation, a comparison across the different subfamilies and commonalities of ligand-receptor interactions.
Collapse
|
46
|
Heydenreich FM, Marti-Solano M, Sandhu M, Kobilka BK, Bouvier M, Babu MM. Molecular determinants of ligand efficacy and potency in GPCR signaling. Science 2023; 382:eadh1859. [PMID: 38127743 PMCID: PMC7615523 DOI: 10.1126/science.adh1859] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/07/2023] [Indexed: 12/23/2023]
Abstract
Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) bind to extracellular ligands and drugs and modulate intracellular responses through conformational changes. Despite their importance as drug targets, the molecular origins of pharmacological properties such as efficacy (maximum signaling response) and potency (the ligand concentration at half-maximal response) remain poorly understood for any ligand-receptor-signaling system. We used the prototypical adrenaline-β2 adrenergic receptor-G protein system to reveal how specific receptor residues decode and translate the information encoded in a ligand to mediate a signaling response. We present a data science framework to integrate pharmacological and structural data to uncover structural changes and allosteric networks relevant for ligand pharmacology. These methods can be tailored to study any ligand-receptor-signaling system, and the principles open possibilities for designing orthosteric and allosteric compounds with defined signaling properties.
Collapse
Affiliation(s)
- Franziska M. Heydenreich
- Department of Molecular and Cellular Physiology, Stanford University
School of Medicine, Stanford, CA, USA
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Biochemistry and Molecular Medicine, Institute for
Research in Immunology and Cancer, Université de Montréal, Montreal,
QC, Canada
| | - Maria Marti-Solano
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Pharmacology, University of Cambridge, Cambridge,
UK
| | - Manbir Sandhu
- Department of Pharmacology, University of Cambridge, Cambridge,
UK
| | - Brian K. Kobilka
- Department of Molecular and Cellular Physiology, Stanford University
School of Medicine, Stanford, CA, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for
Research in Immunology and Cancer, Université de Montréal, Montreal,
QC, Canada
| | - M. Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Structural Biology and Center of Excellence for
Data-Driven Discovery, St. Jude Children’s Research Hospital, Memphis, TN,
USA
| |
Collapse
|
47
|
Giraldo J, Madsen JJ, Wang X, Wang L, Zhang C, Ye L. A 19F-qNMR-Guided Mathematical Model for G Protein-Coupled Receptor Signaling. Mol Pharmacol 2023; 105:54-62. [PMID: 37907352 PMCID: PMC10739436 DOI: 10.1124/molpharm.123.000754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/13/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023] Open
Abstract
G protein-coupled receptors (GPCRs) exhibit a wide range of pharmacological efficacies, yet the molecular mechanisms responsible for the differential efficacies in response to various ligands remain poorly understood. This lack of understanding has hindered the development of a solid foundation for establishing a mathematical model for signaling efficacy. However, recent progress has been made in delineating and quantifying receptor conformational states and associating function with these conformations. This progress has allowed us to construct a mathematical model for GPCR signaling efficacy that goes beyond the traditional ON/OFF binary switch model. In this study, we present a quantitative conformation-based mathematical model for GPCR signaling efficacy using the adenosine A2A receptor (A2AR) as a model system, under the guide of 19F quantitative nuclear magnetic resonance experiments. This model encompasses two signaling states, a fully activated state and a partially activated state, defined as being able to regulate the cognate Gα s nucleotide exchange with respective G protein recognition capacity. By quantifying the population distribution of each state, we can now in turn examine GPCR signaling efficacy. This advance provides a foundation for assessing GPCR signaling efficacy using a conformation-based mathematical model in response to ligand binding. SIGNIFICANCE STATEMENT: Mathematical models to describe signaling efficacy of GPCRs mostly suffer from considering only two states (ON/OFF). However, research indicates that a GPCR possesses multiple active-(like) states that can interact with Gαβγ independently, regulating varied nucleotide exchanges. With the guide of 19F-qNMR, the transitions among these states are quantified as a function of ligand and Gαβγ, serving as a foundation for a novel conformation-based mathematical signaling model.
Collapse
Affiliation(s)
- Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (J.G.), CIBERSAM, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Spain; Global and Planetary Health, College of Public Health (J.J.M.), Center for Global Health and Infectious Diseases Research, College of Public Health (J.J.M.), Department of Molecular Medicine, Morsani College of Medicine (J.J.M.), Department of Molecular Biosciences (X.W., L.Y.), University of South Florida, Tampa, Florida; Department of Pharmacology and Chemical Biology, University of PittsburghSchool of Medicine (L.W., C.Z.), University of Pittsburgh, Pittsburgh, Pennsylvania; and Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (L.Y.)
| | - Jesper J Madsen
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (J.G.), CIBERSAM, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Spain; Global and Planetary Health, College of Public Health (J.J.M.), Center for Global Health and Infectious Diseases Research, College of Public Health (J.J.M.), Department of Molecular Medicine, Morsani College of Medicine (J.J.M.), Department of Molecular Biosciences (X.W., L.Y.), University of South Florida, Tampa, Florida; Department of Pharmacology and Chemical Biology, University of PittsburghSchool of Medicine (L.W., C.Z.), University of Pittsburgh, Pittsburgh, Pennsylvania; and Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (L.Y.)
| | - Xudong Wang
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (J.G.), CIBERSAM, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Spain; Global and Planetary Health, College of Public Health (J.J.M.), Center for Global Health and Infectious Diseases Research, College of Public Health (J.J.M.), Department of Molecular Medicine, Morsani College of Medicine (J.J.M.), Department of Molecular Biosciences (X.W., L.Y.), University of South Florida, Tampa, Florida; Department of Pharmacology and Chemical Biology, University of PittsburghSchool of Medicine (L.W., C.Z.), University of Pittsburgh, Pittsburgh, Pennsylvania; and Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (L.Y.)
| | - Lei Wang
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (J.G.), CIBERSAM, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Spain; Global and Planetary Health, College of Public Health (J.J.M.), Center for Global Health and Infectious Diseases Research, College of Public Health (J.J.M.), Department of Molecular Medicine, Morsani College of Medicine (J.J.M.), Department of Molecular Biosciences (X.W., L.Y.), University of South Florida, Tampa, Florida; Department of Pharmacology and Chemical Biology, University of PittsburghSchool of Medicine (L.W., C.Z.), University of Pittsburgh, Pittsburgh, Pennsylvania; and Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (L.Y.)
| | - Cheng Zhang
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (J.G.), CIBERSAM, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Spain; Global and Planetary Health, College of Public Health (J.J.M.), Center for Global Health and Infectious Diseases Research, College of Public Health (J.J.M.), Department of Molecular Medicine, Morsani College of Medicine (J.J.M.), Department of Molecular Biosciences (X.W., L.Y.), University of South Florida, Tampa, Florida; Department of Pharmacology and Chemical Biology, University of PittsburghSchool of Medicine (L.W., C.Z.), University of Pittsburgh, Pittsburgh, Pennsylvania; and Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (L.Y.)
| | - Libin Ye
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (J.G.), CIBERSAM, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona (J.G.), Spain; Global and Planetary Health, College of Public Health (J.J.M.), Center for Global Health and Infectious Diseases Research, College of Public Health (J.J.M.), Department of Molecular Medicine, Morsani College of Medicine (J.J.M.), Department of Molecular Biosciences (X.W., L.Y.), University of South Florida, Tampa, Florida; Department of Pharmacology and Chemical Biology, University of PittsburghSchool of Medicine (L.W., C.Z.), University of Pittsburgh, Pittsburgh, Pennsylvania; and Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (L.Y.)
| |
Collapse
|
48
|
Xu Z, Guo L, Yu J, Shen S, Wu C, Zhang W, Zhao C, Deng Y, Tian X, Feng Y, Hou H, Su L, Wang H, Guo S, Wang H, Wang K, Chen P, Zhao J, Zhang X, Yong X, Cheng L, Liu L, Yang S, Yang F, Wang X, Yu X, Xu Y, Sun JP, Yan W, Shao Z. Ligand recognition and G-protein coupling of trace amine receptor TAAR1. Nature 2023; 624:672-681. [PMID: 37935376 DOI: 10.1038/s41586-023-06804-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023]
Abstract
Trace-amine-associated receptors (TAARs), a group of biogenic amine receptors, have essential roles in neurological and metabolic homeostasis1. They recognize diverse endogenous trace amines and subsequently activate a range of G-protein-subtype signalling pathways2,3. Notably, TAAR1 has emerged as a promising therapeutic target for treating psychiatric disorders4,5. However, the molecular mechanisms underlying its ability to recognize different ligands remain largely unclear. Here we present nine cryo-electron microscopy structures, with eight showing human and mouse TAAR1 in a complex with an array of ligands, including the endogenous 3-iodothyronamine, two antipsychotic agents, the psychoactive drug amphetamine and two identified catecholamine agonists, and one showing 5-HT1AR in a complex with an antipsychotic agent. These structures reveal a rigid consensus binding motif in TAAR1 that binds to endogenous trace amine stimuli and two extended binding pockets that accommodate diverse chemotypes. Combined with mutational analysis, functional assays and molecular dynamic simulations, we elucidate the structural basis of drug polypharmacology and identify the species-specific differences between human and mouse TAAR1. Our study provides insights into the mechanism of ligand recognition and G-protein selectivity by TAAR1, which may help in the discovery of ligands or therapeutic strategies for neurological and metabolic disorders.
Collapse
Affiliation(s)
- Zheng Xu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Lulu Guo
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Yu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Shen
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Wu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weifeng Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Chang Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Deng
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuying Feng
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hanlin Hou
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lantian Su
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Shuo Guo
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Heli Wang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kexin Wang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Peipei Chen
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Xiaoyu Zhang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Cheng
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shengyong Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Yang
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
- Beijing National Laboratory for Molecular Sciences, Beijing, China
| | - Xiao Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Jin-Peng Sun
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
49
|
Wei S, Pour NG, Tiruvadi-Krishnan S, Ray AP, Thakur N, Eddy MT, Lamichhane R. Single-molecule visualization of human A 2A adenosine receptor activation by a G protein and constitutively activating mutations. Commun Biol 2023; 6:1218. [PMID: 38036689 PMCID: PMC10689853 DOI: 10.1038/s42003-023-05603-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Mutations that constitutively activate G protein-coupled receptors (GPCRs), known as constitutively activating mutations (CAMs), modify cell signaling and interfere with drugs, resulting in diseases with limited treatment options. We utilize fluorescence imaging at the single-molecule level to visualize the dynamic process of CAM-mediated activation of the human A2A adenosine receptor (A2AAR) in real time. We observe an active-state population for all CAMs without agonist stimulation. Importantly, activating mutations significantly increase the population of an intermediate state crucial for receptor activation, notably distinct from the addition of a partner G protein. Activation kinetics show that while CAMs increase the frequency of transitions to the intermediate state, mutations altering sodium sensitivity increase transitions away from it. These findings indicate changes in GPCR function caused by mutations may be predicted based on whether they favor or disfavor formation of an intermediate state, providing a framework for designing receptors with altered functions or therapies that target intermediate states.
Collapse
Affiliation(s)
- Shushu Wei
- Department of Biochemistry & Cellular and Molecular Biology, College of Arts and Sciences, University of Tennessee, Knoxville, TN, USA
| | - Niloofar Gopal Pour
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Sriram Tiruvadi-Krishnan
- Department of Biochemistry & Cellular and Molecular Biology, College of Arts and Sciences, University of Tennessee, Knoxville, TN, USA
| | - Arka Prabha Ray
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Naveen Thakur
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Matthew T Eddy
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA.
| | - Rajan Lamichhane
- Department of Biochemistry & Cellular and Molecular Biology, College of Arts and Sciences, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
50
|
Papadourakis M, Sinenka H, Matricon P, Hénin J, Brannigan G, Pérez-Benito L, Pande V, van Vlijmen H, de Graaf C, Deflorian F, Tresadern G, Cecchini M, Cournia Z. Alchemical Free Energy Calculations on Membrane-Associated Proteins. J Chem Theory Comput 2023; 19:7437-7458. [PMID: 37902715 PMCID: PMC11017255 DOI: 10.1021/acs.jctc.3c00365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 10/31/2023]
Abstract
Membrane proteins have diverse functions within cells and are well-established drug targets. The advances in membrane protein structural biology have revealed drug and lipid binding sites on membrane proteins, while computational methods such as molecular simulations can resolve the thermodynamic basis of these interactions. Particularly, alchemical free energy calculations have shown promise in the calculation of reliable and reproducible binding free energies of protein-ligand and protein-lipid complexes in membrane-associated systems. In this review, we present an overview of representative alchemical free energy studies on G-protein-coupled receptors, ion channels, transporters as well as protein-lipid interactions, with emphasis on best practices and critical aspects of running these simulations. Additionally, we analyze challenges and successes when running alchemical free energy calculations on membrane-associated proteins. Finally, we highlight the value of alchemical free energy calculations calculations in drug discovery and their applicability in the pharmaceutical industry.
Collapse
Affiliation(s)
- Michail Papadourakis
- Biomedical
Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| | - Hryhory Sinenka
- Institut
de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| | - Pierre Matricon
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Jérôme Hénin
- Laboratoire
de Biochimie Théorique UPR 9080, CNRS and Université Paris Cité, 75005 Paris, France
| | - Grace Brannigan
- Center
for Computational and Integrative Biology, Rutgers University−Camden, Camden, New Jersey 08103, United States of America
- Department
of Physics, Rutgers University−Camden, Camden, New Jersey 08102, United States
of America
| | - Laura Pérez-Benito
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Vineet Pande
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Herman van Vlijmen
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Chris de Graaf
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Francesca Deflorian
- Sosei
Heptares, Steinmetz Building,
Granta Park, Great Abington, Cambridge CB21 6DG, United
Kingdom
| | - Gary Tresadern
- CADD,
In Silico Discovery, Janssen Research &
Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marco Cecchini
- Institut
de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| | - Zoe Cournia
- Biomedical
Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527 Athens, Greece
| |
Collapse
|