1
|
Albers DPJ, Novikova S, Vieyto-Nuñez J, Almeida-Hernández Y, Pastorio C, Klassen F, Weiss D, von Maltitz P, Jaikishan J, Datta M, Jumaa H, Jebaraj BMC, Stilgenbauer S, Kumar M, Maity PC, Buske C, Stifel U, Zinngrebe J, Fischer-Posovszky P, Chevigné A, Kirchhoff F, Sanchez-Garcia E, Münch J, Harms M. GPR15LG binds CXCR4 and synergistically modulates CXCL12-induced cell signaling and migration. Cell Commun Signal 2025; 23:234. [PMID: 40394646 PMCID: PMC12093852 DOI: 10.1186/s12964-025-02231-x] [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: 02/11/2025] [Accepted: 05/04/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND GPR15LG, a chemokine-like ligand for the G-protein coupled receptor 15 (GPR15), is abundantly expressed in the gastrointestinal mucosa and inflamed skin. Emerging evidence suggests its involvement in inflammatory disorders and cancers. C-X-C chemokine receptor type 4 (CXCR4) plays a critical role in immune cell trafficking and cancer metastasis. Recent evidence suggests a connection between GPR15LG and CXCR4 signaling, which has not been investigated so far. METHODS We investigated the effects of GPR15LG on CXCR4 signaling and downstream functions. Binding assays and computational modeling were performed to assess the interaction between GPR15LG and CXCR4. Functional assays, including wound healing and cell migration assays, were conducted across various cell types, including CD4⁺ T cells and cancer cells, to evaluate the impact of GPR15LG on CXCL12-mediated CXCR4 signaling. RESULTS The results demonstrate that GPR15LG binds to the orthosteric site of CXCR4, modulating downstream signaling in a context-dependent manner. Specifically, GPR15LG enhances CXCL12-mediated CXCR4 signaling synergistically, promoting wound healing and cell migration across various cell types, including CD4 + T cells and cancer cells. CONCLUSIONS These findings underscore the role of GPR15LG in inflammation and metastasis, offering potential therapeutic avenues for CXCR4-mediated diseases.
Collapse
Affiliation(s)
| | - Sofya Novikova
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Julio Vieyto-Nuñez
- Chair of Computational Bioengineering, Department of Biochemical and Chemical Engineering, Technical University Dortmund, 44227, Dortmund, Germany
| | - Yasser Almeida-Hernández
- Chair of Computational Bioengineering, Department of Biochemical and Chemical Engineering, Technical University Dortmund, 44227, Dortmund, Germany
| | - Chiara Pastorio
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Florian Klassen
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Dana Weiss
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Pascal von Maltitz
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Janeni Jaikishan
- Institute of Immunology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Moumita Datta
- Institute of Immunology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Hassan Jumaa
- Institute of Immunology, Ulm University Medical Center, 89081, Ulm, Germany
| | | | - Stephan Stilgenbauer
- Division of CLL, Department of Internal Medicine III, Ulm University Medical Center, 89081, Ulm, Germany
| | - Manish Kumar
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081, Ulm, Germany
| | - Palash Chandra Maity
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081, Ulm, Germany
| | - Christian Buske
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081, Ulm, Germany
| | - Ulrich Stifel
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Julia Zinngrebe
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site Ulm, Ulm, Germany
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Elsa Sanchez-Garcia
- Chair of Computational Bioengineering, Department of Biochemical and Chemical Engineering, Technical University Dortmund, 44227, Dortmund, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
2
|
Franchini L, Porter JJ, Lueck JD, Orlandi C. G zESTY as an optimized cell-based assay for initial steps in GPCR deorphanization. Nat Commun 2025; 16:4521. [PMID: 40374633 PMCID: PMC12081699 DOI: 10.1038/s41467-025-59850-8] [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: 07/26/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are key pharmacological targets, yet many remain underutilized due to unknown activation mechanisms and ligands. Orphan GPCRs, lacking identified natural ligands, are a high priority for research, as identifying their ligands will aid in understanding their functions and potential as drug targets. Most GPCRs, including orphans, couple to Gi/o/z family members, however current assays to detect their activation are limited, hindering ligand identification efforts. We introduce GzESTY, a sensitive, cell-based assay developed in an easily deliverable format designed to study the pharmacology of Gi/o/z-coupled GPCRs and assist in deorphanization. We optimized assay conditions and developed an all-in-one vector employing cloning methods to ensure the correct expression ratio of GzESTY components. GzESTY successfully assessed activation of a library of ligand-activated GPCRs, detecting both full and partial agonism, and responses from endogenous GPCRs. Notably, with GzESTY we established the presence of endogenous ligands for GPR176 and GPR37 in brain extracts, validating its use in deorphanization efforts. This assay enhances the ability to find ligands for orphan GPCRs, expanding the toolkit for GPCR pharmacologists.
Collapse
Grants
- R01DC022104 U.S. Department of Health & Human Services | NIH | National Institute on Deafness and Other Communication Disorders (NIDCD)
- R01HL153988 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL153988 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Institute on Deafness and Other Communication Disorders (NIDCD)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- This work was supported by start-up funding from the Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry to C.O.; Ernest J. Del Monte Institute for Neuroscience Pilot Program, University of Rochester, to C.O.; University Research Award, University of Rochester to C.O; NIDCD/NIH grant R01DC022104 to C.O.; R01HL153988 to J.D.L.; this work was aided by the GCE4All Biomedical Technology Optimization and Dissemination Center supported by National Institute of General Medical Science grant RM1-GM144227; The Foundation Blanceflor Boncompagni Ludovisi-née Bildt fellowship to L.F.
Collapse
Affiliation(s)
- Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Joseph J Porter
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - John D Lueck
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
3
|
Larasati YA, Solis GP, Koval A, François‐Heude M, Piarroux J, Roubertie A, Yang R, Zhang Y, Cao D, Korff CM, Katanaev VL. Novel Mutation at Cys225 in GNAO1-Associated Developmental and Epileptic Encephalopathies: Clinical, Molecular, and Pharmacological Profiling of Case Studies. MedComm (Beijing) 2025; 6:e70196. [PMID: 40337144 PMCID: PMC12056497 DOI: 10.1002/mco2.70196] [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: 05/05/2024] [Revised: 02/15/2025] [Accepted: 03/03/2025] [Indexed: 05/09/2025] Open
Abstract
GNAO1-associated disorders have a large spectrum of neurological symptoms, from early-onset developmental and epileptic encephalopathies (DEE) to late-onset movement disorders. First reported in 2013 and now identified in around 400 cases worldwide, this disease is caused by dominant, mostly de novo missense mutations in GNAO1, the gene encoding the major neuronal G protein Gαo. Being the immediate transducer of a number of neuronal G protein-coupled receptors, Gαo plays crucial functions in brain development and physiology. Here, we discover a novel mutation site in GNAO1, Cys225 mutated to Tyr or Arg in pediatric individuals from France and China (p.(Cys225Tyr) and p.(Cys225Arg), respectively), leading to severe early-onset DEE. Molecular investigations characterize the novel pathogenic variants as deficient in the interactions with guanine nucleotides and physiological cellular partners of Gαo, with reduced stability and plasma membrane localization and a strong neomorphic interaction with the chaperone Ric8A. Salts of zinc, emerging as a promising targeted therapy for GNAO1-associated disorders, impose a previously unseen effect on the mutant Gαo, accelerating the loss of its ability to interact with guanine nucleotides. Our study, combining clinical, cellular, molecular, and modeling approaches, describes deep insights into molecular etiology and treatment perspectives of the novel form of GNAO1-associated disorders.
Collapse
Affiliation(s)
- Yonika A. Larasati
- Translational Research Center in OncohaematologyDepartment of Cell Physiology and MetabolismFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Gonzalo P. Solis
- Translational Research Center in OncohaematologyDepartment of Cell Physiology and MetabolismFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Alexey Koval
- Translational Research Center in OncohaematologyDepartment of Cell Physiology and MetabolismFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | | | - Julie Piarroux
- Service de Neuropédiatrie, Hôpital Gui de ChauliacMontpellierFrance
| | - Agathe Roubertie
- Service de Neuropédiatrie, Hôpital Gui de ChauliacMontpellierFrance
- Institut des Neurosciences de Montpellier, INSERM U 1298MontpellierFrance
| | - Ruihan Yang
- Department of NeurologyShenzhen Children's HospitalShenzhenChina
- Medical CollegeShantou UniversityShantouChina
| | - Ying Zhang
- Department of Pediatric Neurologythe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Dezhi Cao
- Department of NeurologyShenzhen Children's HospitalShenzhenChina
- Medical CollegeShenzhen UniversityShenzhenChina
| | | | - Vladimir L. Katanaev
- Translational Research Center in OncohaematologyDepartment of Cell Physiology and MetabolismFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
4
|
Bagger SM, Schihada H, Walser ALS, Drzazga AK, Grätz L, Palmisano T, Kuhn CK, Mavri M, Mølleskov-Jensen AS, Tall GG, Schöneberg T, Mathiasen SJ, Javitch JA, Schulte G, Spiess K, Rosenkilde MM. Complex G-protein signaling of the adhesion GPCR, ADGRA3. J Biol Chem 2025; 301:108441. [PMID: 40127866 PMCID: PMC12059339 DOI: 10.1016/j.jbc.2025.108441] [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: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025] Open
Abstract
ADGRA3 (GPR125) is an orphan adhesion G protein-coupled receptor (aGPCR) involved in planar cell polarity, primarily through recruitment of the signaling components disheveled (DVL) during vertebrate gastrulation and discs large homolog 1, implicated in cancer. Limited knowledge exists of the canonical G protein-coupled receptor pathways downstream of ADGRA3. Here, we employed a series of human cell line-based signaling assays to gain insight into the G protein-mediated signaling of ADGRA3. We designed ADGRA3 constructs based on transcript variant analysis in publicly available human liver and brain RNA-seq datasets. Cleavage in the GPCR autoproteolysis site (GPS) is an aGPCR hallmark; thus, we generated a truncated ADGRA3 (C-terminal fragment, CTF) corresponding to a potential cleavage at the GPS. We found low-level activation of Gi and Gs by ADGRA3 and slightly more by its CTF. As the N terminus of the CTF constitutes a class-defined tethered agonist (so-called stachel peptide), we removed the initial three amino acids of the CTF. This resulted in abrogated G protein-mediated signaling, as observed for other aGPCRs. Due to the central role of ADGRA3 in planar cell polarity signaling through DVL recruitment, we investigated the G-protein signaling in the absence of DVL1-3 and found it sustained. No transcriptional activation was observed in an assay of downstream β-catenin activity. Collectively, this establishes classical G protein-mediated signaling for ADGRA3.
Collapse
Affiliation(s)
- Sofie M Bagger
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hannes Schihada
- Department of Physiology and Pharmacology, Section of Receptor Biology and Signaling, Karolinska Institutet, Stockholm, Sweden
| | - Anna L S Walser
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna K Drzazga
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lukas Grätz
- Department of Physiology and Pharmacology, Section of Receptor Biology and Signaling, Karolinska Institutet, Stockholm, Sweden
| | - Tiago Palmisano
- Departments of Psychiatry and Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Christina K Kuhn
- Molecular Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig, Germany
| | - Maša Mavri
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Sophie Mølleskov-Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gregory G Tall
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Torsten Schöneberg
- Molecular Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig, Germany
| | - Signe J Mathiasen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Departments of Psychiatry and Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Jonathan A Javitch
- Departments of Psychiatry and Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Section of Receptor Biology and Signaling, Karolinska Institutet, Stockholm, Sweden
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
5
|
Borrega-Roman L, Hoare BL, Kosar M, Sarott RC, Patej KJ, Bouma J, Scott-Dennis M, Koers EJ, Gazzi T, Mach L, Barrondo S, Sallés J, Guba W, Kusznir E, Nazaré M, Rufer AC, Grether U, Heitman LH, Carreira EM, Sykes DA, Veprintsev DB. A universal cannabinoid CB1 and CB2 receptor TR-FRET kinetic ligand-binding assay. Front Pharmacol 2025; 16:1469986. [PMID: 40271066 PMCID: PMC12015242 DOI: 10.3389/fphar.2025.1469986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/11/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction The kinetics of ligand binding to G protein-coupled receptors (GPCRs) is an important optimization parameter in drug discovery. Traditional radioligand assays are labor-intensive, preventing their application at the early stages of drug discovery. Fluorescence-based assays offer several advantages, including a possibility to develop a homogeneous format, continuous data collection, and higher throughput. This study sought to develop a fluorescence-based binding assay to investigate ligand-binding kinetics at human cannabinoid type 1 and 2 receptors (CB1R and CB2R). Methods We synthesized D77, a novel tracer derived from the non-selective cannabinoid Δ8-THC. Using time-resolved Förster resonance energy transfer (TR-FRET), we developed an assay to study ligand-binding kinetics at physiological temperatures. For CB1R, we truncated the first 90 amino acids of its flexible N-terminal domain to reduce the FRET distance between the terbium cryptate (donor) and the fluorescent ligand (acceptor). The full-length CB2R construct was functional without modification due to its shorter N-terminus. The Motulsky-Mahan competition binding model was used to analyze the binding kinetics of the endocannabinoids and several other non-fluorescent ligands. Results The D77 tracer showed nanomolar-range affinity for truncated CB1R (CB1R91-472) and full-length CB2R (CB2R1-360), displaying competitive binding with orthosteric ligands. D77 exhibited rapid dissociation kinetics from both CB1R and CB2R, which were similar to the fastest dissociating reference compounds. This was critical for accurately determining the on- and off-rates of the fastest dissociating compounds. Using D77, we measured the kinetic binding properties of various CB1R and CB2R agonists and antagonists at physiological temperature and sodium ion concentration. Discussion The k on values for molecules binding to CB1R varied by three orders of magnitude, from the slowest (HU308) to the fastest (rimonabant). A strong correlation between k on and affinity was observed for compounds binding to CB1R, indicating that the association rate primarily determines their affinity for CB1R. Unlike CB1R, a stronger correlation was found between the dissociation rate constant k off and the affinity for CB2R, suggesting that both k on and k off dictate the overall affinity for CB2R. Exploring the kinetic parameters of cannabinoid drug candidates could help drug development programs targeting these receptors.
Collapse
Affiliation(s)
- Leire Borrega-Roman
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
| | - Bradley L. Hoare
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Miroslav Kosar
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Roman C. Sarott
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Kacper J. Patej
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Jara Bouma
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University and Oncode Institute, Leiden, Netherlands
| | - Morgan Scott-Dennis
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Eline J. Koers
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Campus BerlinBuch, Berlin, Germany
| | - Leonard Mach
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Campus BerlinBuch, Berlin, Germany
| | - Sergio Barrondo
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Joan Sallés
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Wolfgang Guba
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Eric Kusznir
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Campus BerlinBuch, Berlin, Germany
| | - Arne C. Rufer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Laura H. Heitman
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University and Oncode Institute, Leiden, Netherlands
| | - Erick M. Carreira
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - David A. Sykes
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Dmitry B. Veprintsev
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| |
Collapse
|
6
|
Gratio V, Dragan P, Garcia L, Saveanu L, Nicole P, Voisin T, Latek D, Couvineau A. Pharmacodynamics of the orexin type 1 (OX 1) receptor in colon cancer cell models: A two-sided nature of antagonistic ligands resulting from partial dissociation of Gq. Br J Pharmacol 2025; 182:1528-1545. [PMID: 39675769 DOI: 10.1111/bph.17422] [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: 01/23/2024] [Revised: 10/24/2024] [Accepted: 11/10/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Orexins have important biological effects on the central and peripheral nervous systems. Their primary ability is to regulate the sleep-wake cycle. Orexins and their antagonists, via OX1 receptor have been shown to have proapoptotic and antitumor effects on various digestive cancers cell models. We investigated, (1) the ability of orexin-A and its antagonists to regulate OX1 receptor expression at the cell surface and (2), how OX1 antagonists induced proapoptotic effect in cancer cells models. EXPERIMENTAL APPROACH The OX1 receptor internalisation is determined by imaging flow cytometry in colon cancer cell models and the OX1 receptor coupling to G proteins via bioluminescence resonance energy transfer and molecular dynamic simulation. KEY RESULTS Orexin-A induced rapid receptor internalisation within 15 min via β-arrestin 2 recruitment, whereas antagonists had no effect. Furthermore, Gq is critical for receptor internalisation and signalling pathways, and no other G proteins appear to be recruited. Surprisingly, antagonists induced recruitment and conformational changes in Gq protein. Simulated molecular dynamics of agonists/orexin receptor/Gq complexes show that antagonists exhibits a similar binding mode, stable at the binding site and show conformational changes of ECL2, similar to that of the agonists. CONCLUSION AND IMPLICATIONS OX1 receptor activation induced orexin/β-arrestin-dependent internalisation, which was independent of the apoptotic pathway induced by orexins and antagonists. In addition, antagonists activate the Gq protein, suggesting its putative partial dissociation. These results suggest that the development of OX1 receptor targeting molecules, including orexin antagonists with antitumor properties, may pave the way for innovative cancer therapies.
Collapse
Affiliation(s)
- Valérie Gratio
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
- INSERM UMR1149/Inflammation Research Center (CRI), Flow Cytometry Platform (CytoCRI), DHU UNITY, Université Paris Cité, Paris, France
| | - Paulina Dragan
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Laurine Garcia
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| | - Loredana Saveanu
- INSERM UMR1149/Inflammation Research Center (CRI), Team "Antigen Presentation by Dendritic Cells to T cells (APreT)", DHU UNITY, Université Paris Cité, Paris, France
| | - Pascal Nicole
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| | - Thierry Voisin
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Alain Couvineau
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| |
Collapse
|
7
|
Perry-Hauser NA, Du Rand JR, Lee KH, Shi L, Javitch JA. N-terminal fragment shedding contributes to signaling of the full-length adhesion receptor ADGRL3. J Biol Chem 2025; 301:108174. [PMID: 39798870 PMCID: PMC11849108 DOI: 10.1016/j.jbc.2025.108174] [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: 09/24/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/15/2025] Open
Abstract
Most adhesion G protein-coupled receptors (GPCRs) undergo autoproteolytic cleavage during receptor biosynthesis, resulting in noncovalently bound N-terminal fragments (NTFs) and C-terminal fragments (CTFs) that remain associated during receptor trafficking to the plasma membrane. While substantial evidence supports increased G protein signaling when just the CTF is expressed, there is an ongoing debate about whether NTF removal is required to initiate signaling in the context of the WT receptor. Here, we use adhesion GPCR latrophilin-3 (ADGRL3) as a model receptor to investigate tethered agonist (TA)-mediated activation. First, we show that extending the N terminus of the TA in ADGRL3 CTF disrupts G protein signaling. This suggests that if the TA is not fully exposed, it is unlikely to interact with the orthosteric pocket in an optimal manner for G protein activation. Second, we show that when full-length ADGRL3 is expressed in heterologous cells, approximately 5% of the receptor population spontaneously sheds its NTF. We hypothesized that the signaling activity observed for full-length ADGRL3 is largely because of this shedding, which exposes the native TA. To test this hypothesis, we used a full-length cleavage-deficient ADGRL3 mutant. Compared with WT receptor, this mutant lost ∼80% of its signaling through Gα13 and showed a much lower level of spontaneous NTF shedding, approximately 20% of that observed for WT receptor. This loss of spontaneous NTF shedding likely explains its diminished signaling activity. These findings suggest that TA-mediated signal transduction by full-length ADGRL3 requires removal of its NTF.
Collapse
Affiliation(s)
- Nicole A Perry-Hauser
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Jonathan R Du Rand
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Kuo Hao Lee
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA.
| |
Collapse
|
8
|
Zhang Y, Zheng Q, Warshel A, Bai C. Key Interaction Changes Determine the Activation Process of Human Parathyroid Hormone Type 1 Receptor. J Am Chem Soc 2025; 147:3539-3552. [PMID: 39804793 DOI: 10.1021/jacs.4c15025] [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: 01/16/2025]
Abstract
The parathyroid hormone type 1 receptor (PTH1R) plays a crucial role in modulating various physiological functions and is considered an effective therapeutic target for osteoporosis. However, a lack of detailed molecular and energetic information about PTH1R limits our comprehensive understanding of its activation process. In this study, we performed computational simulations to explore key events in the activation process, such as conformational changes in PTH1R, Gs protein coupling, and the release of guanosine diphosphate (GDP). Our analysis identified kinetic information, including the rate-determining step, transition state, and energy barriers. Free-energy and structural analyses revealed that GDP could be released from the Gs protein when the binding cavity is partially open. Additionally, we predicted important residues, including potential pathogenic mutations, and verified their significance through site-directed mutations. These findings enhance our understanding of class B GPCR activation mechanisms. Furthermore, the methodology employed in this study can be applied to other biophysical systems.
Collapse
Affiliation(s)
- Yue Zhang
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun 130012, China
- Warshel Institute for Computational Biology, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
| | - Qingchuan Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Chen Bai
- Warshel Institute for Computational Biology, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
- Chenzhu (MoMeD) Biotechnology Co., Ltd., Hangzhou 310005, China
| |
Collapse
|
9
|
Duan Y, Xu Z, Hao B, Zhang A, Guo C, He Y. Molecular mechanism of ligand recognition and activation of lysophosphatidic acid receptor LPAR6. Proc Natl Acad Sci U S A 2025; 122:e2415426122. [PMID: 39847322 PMCID: PMC11789011 DOI: 10.1073/pnas.2415426122] [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: 07/31/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
Lysophosphatidic acid (LPA) exerts its physiological roles through the endothelialdifferentiation gene (EDG) family LPA receptors (LPAR1-3) or the non-EDG family LPA receptors (LPAR4-6). LPAR6 plays crucial roles in hair loss and cancer progression, yet its structural information is very limited. Here, we report the cryoelectron microscopy structure of LPA-bound human LPAR6 in complex with a mini G13 or Gq protein. These structures reveal a distinct ligand binding and recognition mode that differs significantly from that of LPAR1. Specifically, LPA uses its charged head to form an extensive polar interaction network with key polar residues on the extracellular side of transmembrane helix 5-6 and the extracellular loop 2. Structural comparisons and homology analysis suggest that the EDG and non-EDG families use two distinct modes for LPA binding. The structural observations are validated through functional mutagenesis studies. We further uncover the mechanisms of LPAR6 activation and principles of G-protein coupling. The structural information revealed by our study lays the groundwork for understanding LPAR6 signaling and provides a rational basis for designing compounds targeting LPAR6.
Collapse
Affiliation(s)
- Yaning Duan
- Faculty of Life Sciences and Medicine, Harbin Institute of Technology Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin150001, China
| | - Zhenmei Xu
- Faculty of Life Sciences and Medicine, Harbin Institute of Technology Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin150001, China
| | - Boyu Hao
- Faculty of Life Sciences and Medicine, Harbin Institute of Technology Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin150001, China
| | - Anqi Zhang
- Faculty of Life Sciences and Medicine, Harbin Institute of Technology Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin150001, China
| | - Changyou Guo
- Faculty of Life Sciences and Medicine, Harbin Institute of Technology Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin150001, China
| | - Yuanzheng He
- Faculty of Life Sciences and Medicine, Harbin Institute of Technology Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin150001, China
- Frontiers Science Center for Matter Behave in Space Environment, Harbin Institute of Technology, Harbin150001, China
| |
Collapse
|
10
|
Boon K, Vanalken N, Szpakowska M, Chevigné A, Schols D, Van Loy T. High-affinity ELR+ chemokine ligands show G protein bias over β-arrestin recruitment and receptor internalization in CXCR1 signaling. J Biol Chem 2025; 301:108044. [PMID: 39615686 PMCID: PMC11732455 DOI: 10.1016/j.jbc.2024.108044] [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: 06/26/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
The human CXC chemokine receptor 1 (CXCR1), a G protein-coupled receptor (GPCR), plays significant roles in inflammatory diseases and cancer. While CXCL8 is a well-established high-affinity ligand for CXCR1, there is no consensus regarding the binding ability of the other ELR+ chemokines (CXCL1-3 and CXCL5-8). Since research has predominantly focused on CXCL8-mediated CXCR1 signaling, insight into potential signaling bias induced by different CXCR1 ligands is lacking. Therefore, in this study we first compared and clarified the binding ability of all ELR+ chemokines using a competition binding assay. In this assay CXCL1-3 and CXCL5 behaved as low-affinity ligands while CXCL6-8 were high affinity ligands. We further investigated potential ligand bias within the CXCR1 signaling system. Using NanoBRET-based assays heterotrimeric G protein dissociation, β-arrestin recruitment and receptor internalization induced by chemokine binding to CXCR1 were investigated. A quantitative and qualitative investigation of ligand bias showed that the high-affinity ELR+ chemokines were biased towards G protein activation over β-arrestin recruitment and receptor internalization, when CXCL8 was used as a reference ligand.
Collapse
Affiliation(s)
- Katrijn Boon
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium
| | - Nathan Vanalken
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium
| | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium.
| |
Collapse
|
11
|
Quon T, Lin LC, Ganguly A, Hudson BD, Tobin AB, Milligan G. Biased constitutive signaling of the G protein-coupled receptor GPR35 suppresses gut barrier permeability. J Biol Chem 2025; 301:108035. [PMID: 39615676 PMCID: PMC11732441 DOI: 10.1016/j.jbc.2024.108035] [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: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Agonist-independent, or constitutive, activity is an integral feature of G protein-coupled receptors, but its relevance in pathophysiological settings is generally poorly explored. GPR35 is a therapeutic target in inflammatory diseases of the lower gut. In colonic organoids from a human GPR35a-expressing transgenic mouse line, the GPR35 inverse agonist CID-2745687 increased barrier permeability substantially, indicating that constitutive receptor activity contributes to maintaining epithelial barrier integrity. High constitutive activity of GPR35 was also observed in both HT-29 and HEPG2 cells that express GPR35 endogenously. Mechanistic investigations in recombinant in vitro systems revealed that the constitutive activity of GPR35a was biased and not equivalent across signaling pathways. Hence, no constitutive interactions of the receptor with arrestin-adaptor proteins or activation of Gαo-containing G protein heterotrimers were detected while, even at low GPR35a expression levels, substantial constitutive activation of heterotrimers containing either Gα12 or Gα13 was observed. Similar biased constitutive activity was observed for the human GPR35b isoform. The extent of constitutive and agonist-mediated activity was dependent on receptor expression level. At high receptor levels, constitutive activation of Gα12 or Gα13 masked any agonist-induced effects while low expression levels with low constitutive activity allowed measurement of agonist-induced responses. These results highlight roles, selectivity, and the extent of constitutive activity of GPR35 in cells and tissues that express this receptor endogenously and highlight the contribution of its constitutive activity to maintaining the colonic epithelial barrier, potentially limiting the development of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Tezz Quon
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Li-Chiung Lin
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Amlan Ganguly
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Brian D Hudson
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom.
| |
Collapse
|
12
|
Saca VR, Burdette C, Sakmar TP. GPCR Biosensors to Study Conformational Dynamics and Signaling in Drug Discovery. Annu Rev Pharmacol Toxicol 2025; 65:7-28. [PMID: 39298797 DOI: 10.1146/annurev-pharmtox-061724-080836] [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] [Indexed: 09/22/2024]
Abstract
G protein-coupled receptors (GPCRs) are a superfamily of transmembrane signal transducers that facilitate the flow of chemical signals across membranes. GPCRs are a desirable class of drug targets, and the activation and deactivation dynamics of these receptors are widely studied. Multidisciplinary approaches for studying GPCRs, such as downstream biochemical signaling assays, cryo-electron microscopy structural determinations, and molecular dynamics simulations, have provided insights concerning conformational dynamics and signaling mechanisms. However, new approaches including biosensors that use luminescence- and fluorescence-based readouts have been developed to investigate GPCR-related protein interactions and dynamics directly in cellular environments. Luminescence- and fluorescence-based readout approaches have also included the development of GPCR biosensor platforms that utilize enabling technologies to facilitate multiplexing and miniaturization. General principles underlying the biosensor platforms and technologies include scalability, orthogonality, and kinetic resolution. Further application and development of GPCR biosensors could facilitate hit identification in drug discovery campaigns. The goals of this review are to summarize developments in the field of GPCR-related biosensors and to discuss the current available technologies.
Collapse
Affiliation(s)
- Victoria R Saca
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, NY, USA;
| | - Colin Burdette
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, NY, USA;
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, NY, USA;
| |
Collapse
|
13
|
Pearce A, Redfern-Nichols T, Wills E, Rosa M, Manulak I, Sisk C, Huang X, Atakpa-Adaji P, Prole DL, Ladds G. Quantitative approaches for studying G protein-coupled receptor signalling and pharmacology. J Cell Sci 2025; 138:JCS263434. [PMID: 39810711 PMCID: PMC11828474 DOI: 10.1242/jcs.263434] [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: 01/16/2025] Open
Abstract
G protein-coupled receptor (GPCR) signalling pathways underlie numerous physiological processes, are implicated in many diseases and are major targets for therapeutics. There are more than 800 GPCRs, which together transduce a vast array of extracellular stimuli into a variety of intracellular signals via heterotrimeric G protein activation and multiple downstream effectors. A key challenge in cell biology research and the pharmaceutical industry is developing tools that enable the quantitative investigation of GPCR signalling pathways to gain mechanistic insights into the varied cellular functions and pharmacology of GPCRs. Recent progress in this area has been rapid and extensive. In this Review, we provide a critical overview of these new, state-of-the-art approaches to investigate GPCR signalling pathways. These include novel sensors, Förster or bioluminescence resonance energy transfer assays, libraries of tagged G proteins and transcriptional reporters. These approaches enable improved quantitative studies of different stages of GPCR signalling, including GPCR activation, G protein activation, second messenger (cAMP and Ca2+) signalling, β-arrestin recruitment and the internalisation and intracellular trafficking of GPCRs.
Collapse
Affiliation(s)
- Abigail Pearce
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Theo Redfern-Nichols
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Edward Wills
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Matthew Rosa
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Iga Manulak
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Claudia Sisk
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Xianglin Huang
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Peace Atakpa-Adaji
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - David L. Prole
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| |
Collapse
|
14
|
Bekaddour N, Smith N, Caspar B, Grinberg S, Giorgiutti S, Rodeschini V, Dupuy S, Leboulanger N, Duffy D, Soulas-Sprauel P, Gies V, Korganow AS, Nisole S, Herbeuval JP. The histamine analogue clobenpropit modulates IRF7 phosphorylation and interferon production by targeting CXCR4 in systemic lupus erythematosus models. Front Immunol 2024; 15:1490593. [PMID: 39737176 PMCID: PMC11682962 DOI: 10.3389/fimmu.2024.1490593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by an overactive immune response, particularly involving excessive production of type I interferons. This overproduction is driven by the phosphorylation of IRF7, a crucial factor in interferon gene activation. Current treatments for SLE are often not very effective and can have serious side effects. Methods Our study introduces clobenpropit, a histamine analogue, as a potential new therapy targeting the CXCR4 receptor to reduce IRF7 phosphorylation and subsequent interferon production. We employed various laboratory techniques to investigate how clobenpropit interacts with CXCR4 and its effects on immune cells from healthy individuals and SLE patients. Results Clobenpropit binds effectively to CXCR4, significantly inhibiting IRF7 phosphorylation and reducing interferon production. Additionally, clobenpropit lowered levels of pro-inflammatory cytokines in a mouse model of lupus, demonstrating efficacy comparable to the standard treatment, prednisolone. Discussion These results suggest that clobenpropit could be a promising new treatment for SLE, offering a targeted approach with potential advantages over current therapies.
Collapse
Affiliation(s)
- Nassima Bekaddour
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR), Université Paris Cité, Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy (CBMIT), Paris, France
| | - Nikaïa Smith
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Birgit Caspar
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR), Université Paris Cité, Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy (CBMIT), Paris, France
| | - Severine Grinberg
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR), Université Paris Cité, Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy (CBMIT), Paris, France
| | - Stephane Giorgiutti
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) S1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantation Excellence Nouvelle Génération (Transplantex NG), Fédération Hospitalo-Universitaire OMICs for Care (OMICARE), Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (Centre de Référence des Maladies Auto-Immunes Rares de Strasbourg, (CNR RESO)), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | | | - Stephanie Dupuy
- Unité d’Appui et de Recherche (UAR) BioMedTech Facilities, Plateforme cyto2Bm, Université Paris Cité, Paris, France
| | - Nicolas Leboulanger
- Faculté de Médecine, Université Paris Cité, Paris, France
- Department of Paediatric Otolaryngology, Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Necker-Enfants Malades, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Pauline Soulas-Sprauel
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) S1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantation Excellence Nouvelle Génération (Transplantex NG), Fédération Hospitalo-Universitaire OMICs for Care (OMICARE), Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (Centre de Référence des Maladies Auto-Immunes Rares de Strasbourg, (CNR RESO)), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Vincent Gies
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) S1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantation Excellence Nouvelle Génération (Transplantex NG), Fédération Hospitalo-Universitaire OMICs for Care (OMICARE), Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (Centre de Référence des Maladies Auto-Immunes Rares de Strasbourg, (CNR RESO)), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- Université de Strasbourg, Faculty of Pharmacy, Strasbourg, France
| | - Anne-Sophie Korganow
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) S1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantation Excellence Nouvelle Génération (Transplantex NG), Fédération Hospitalo-Universitaire OMICs for Care (OMICARE), Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (Centre de Référence des Maladies Auto-Immunes Rares de Strasbourg, (CNR RESO)), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- Université de Strasbourg, Faculty of Medicine, Strasbourg, France
| | - Sébastien Nisole
- Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 9004, Montpellier, France
| | - Jean-Philippe Herbeuval
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR), Université Paris Cité, Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy (CBMIT), Paris, France
| |
Collapse
|
15
|
Endzhievskaya S, Chahal K, Resnick J, Khare E, Roy S, Handel TM, Kufareva I. Essential strategies for the detection of constitutive and ligand-dependent Gi-directed activity of 7TM receptors using bioluminescence resonance energy transfer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626681. [PMID: 39713355 PMCID: PMC11661105 DOI: 10.1101/2024.12.04.626681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The constitutive (ligand-independent) signaling of G protein-coupled receptors (GPCRs) is being increasingly appreciated as an integral aspect of their function; however, it can be technically hard to detect for poorly characterized, e.g. orphan, receptors of the cAMP-inhibitory Gi-coupled (GiPCR) family. In this study, we delineate the optimal strategies for the detection of such activity across several GiPCRs in two cell lines. As our study examples, we chose two canonical GiPCRs - the constitutively active Smoothened and the ligand-activated CXCR4, - and one atypical GPCRs, the chemokine receptor ACKR3. We verified the applicability of three Bioluminescence Resonance Energy Transfer (BRET)-based assays - one measuring changes in intracellular cAMP, another in Gβγ/GRK3ct association and third in Gαi-Gβγ dissociation, - for assessing both constitutive and ligand-modulated activity of these receptors. We also revealed the possible caveats and sources of false positives, and proposed optimization strategies. All three types of assays confirmed the ligand-dependent activity of CXCR4, the controversial G protein incompetence of ACKR3, the constitutive Gi-directed activity of SMO, and its modulation by PTCH1. We also demonstrated that PTCH1 promotes SMO localization to the cell surface, thus enhancing its responsiveness not only to agonists but also to antagonists, which is a novel mechanism of regulation of a Class F GiPCR Smoothened.
Collapse
Affiliation(s)
- Sofia Endzhievskaya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Kirti Chahal
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- LigronBio Inc., San Diego, CA, USA
| | - Julie Resnick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ekta Khare
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Suchismita Roy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Tracy M. Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
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
|
17
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [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: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
18
|
Batebi H, Pérez-Hernández G, Rahman SN, Lan B, Kamprad A, Shi M, Speck D, Tiemann JKS, Guixà-González R, Reinhardt F, Stadler PF, Papasergi-Scott MM, Skiniotis G, Scheerer P, Kobilka BK, Mathiesen JM, Liu X, Hildebrand PW. Mechanistic insights into G-protein coupling with an agonist-bound G-protein-coupled receptor. Nat Struct Mol Biol 2024; 31:1692-1701. [PMID: 38867113 DOI: 10.1038/s41594-024-01334-2] [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: 03/09/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024]
Abstract
G-protein-coupled receptors (GPCRs) activate heterotrimeric G proteins by promoting guanine nucleotide exchange. Here, we investigate the coupling of G proteins with GPCRs and describe the events that ultimately lead to the ejection of GDP from its binding pocket in the Gα subunit, the rate-limiting step during G-protein activation. Using molecular dynamics simulations, we investigate the temporal progression of structural rearrangements of GDP-bound Gs protein (Gs·GDP; hereafter GsGDP) upon coupling to the β2-adrenergic receptor (β2AR) in atomic detail. The binding of GsGDP to the β2AR is followed by long-range allosteric effects that significantly reduce the energy needed for GDP release: the opening of α1-αF helices, the displacement of the αG helix and the opening of the α-helical domain. Signal propagation to the Gs occurs through an extended receptor interface, including a lysine-rich motif at the intracellular end of a kinked transmembrane helix 6, which was confirmed by site-directed mutagenesis and functional assays. From this β2AR-GsGDP intermediate, Gs undergoes an in-plane rotation along the receptor axis to approach the β2AR-Gsempty state. The simulations shed light on how the structural elements at the receptor-G-protein interface may interact to transmit the signal over 30 Å to the nucleotide-binding site. Our analysis extends the current limited view of nucleotide-free snapshots to include additional states and structural features responsible for signaling and G-protein coupling specificity.
Collapse
Affiliation(s)
- Hossein Batebi
- Universität Leipzig, Medizinische Fakultät, Institut für Medizinische Physik und Biophysik, Leipzig, Germany
- Freie Universität Berlin, Fachbereich Physik, Berlin, Germany
| | - Guillermo Pérez-Hernández
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany
| | - Sabrina N Rahman
- University of Copenhagen, Department of Drug Design and Pharmacology, Copenhagen, Denmark
| | - Baoliang Lan
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Antje Kamprad
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Mingyu Shi
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - David Speck
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Johanna K S Tiemann
- Universität Leipzig, Medizinische Fakultät, Institut für Medizinische Physik und Biophysik, Leipzig, Germany
- Novozymes A/S, Lyngby, Denmark
| | - Ramon Guixà-González
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Franziska Reinhardt
- Universität Leipzig, Department of Computer Science, Bioinformatics, Leipzig, Germany
| | - Peter F Stadler
- Universität Leipzig, Department of Computer Science, Bioinformatics, Leipzig, Germany
| | - Makaía M Papasergi-Scott
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Patrick Scheerer
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jesper M Mathiesen
- University of Copenhagen, Department of Drug Design and Pharmacology, Copenhagen, Denmark
| | - Xiangyu Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Peter W Hildebrand
- Universität Leipzig, Medizinische Fakultät, Institut für Medizinische Physik und Biophysik, Leipzig, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany.
| |
Collapse
|
19
|
Kurz M, Ulrich M, Kirchhofer SB, Bittner A, Daude M, Diederich WE, Pauck K, Garn H, Bünemann M. Arachidonic Acid Directly Activates the Human DP2 Receptor. Mol Pharmacol 2024; 106:216-224. [PMID: 39284672 DOI: 10.1124/molpharm.124.000884] [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: 01/23/2024] [Revised: 05/30/2024] [Accepted: 06/25/2024] [Indexed: 10/05/2024] Open
Abstract
Aberrant type 2 inflammatory responses are the underlying cause of the pathophysiology of allergic asthma, allergic rhinitis, and other atopic diseases, with an alarming prevalence in relevant parts of the Western world. A bulk of evidence points out the important role of the DP2 receptor in these inflammation processes. A screening of different polyunsaturated fatty acids at a fluorescence resonance energy transfer-based DP2 receptor conformation sensor expressed in human embryonic kidney (HEK) cells revealed an agonistic effect of the prostaglandin (PG)-D2 precursor arachidonic acid on DP2 receptor activity of about 80% of the effect induced by PGD2 In a combination of experiments at the conformation sensor and using a bioluminescence resonance energy transfer-based G protein activation sensor expressed together with DP2 receptor wild type in HEK cells, we found that arachidonic acid acts as a direct activator of the DP2 receptor, but not the DP1 receptor, in a concentration range considered physiologically relevant. Pharmacological inhibition of cyclooxygenases and lipoxygenases as well as cytochrome P450 did not lead to a diminished arachidonic acid response on the DP2 receptor, confirming a direct action of arachidonic acid on the receptor. SIGNIFICANCE STATEMENT: This study identified the prostaglandin precursor arachidonic acid to directly activate the DP2 receptor, a G protein-coupled receptor that is known to play an important role in type 2 inflammation.
Collapse
Affiliation(s)
- Michael Kurz
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Michaela Ulrich
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Sina B Kirchhofer
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Alwina Bittner
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Michael Daude
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Wibke E Diederich
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Kim Pauck
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Holger Garn
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| | - Moritz Bünemann
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.K., M.U., S.B.K., A.B., M.B.); Institute for Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany (M.D., W.E.D.); and Translational Inflammation Research Division and Core Facility for Single-Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany (K.P., H.G.)
| |
Collapse
|
20
|
Schulte G. International Union of Basic and Clinical Pharmacology CXV: The Class F of G Protein-Coupled Receptors. Pharmacol Rev 2024; 76:1009-1037. [PMID: 38955509 DOI: 10.1124/pharmrev.124.001062] [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: 01/03/2024] [Revised: 04/10/2024] [Accepted: 05/17/2024] [Indexed: 07/04/2024] Open
Abstract
The class F of G protein-coupled receptors (GPCRs) consists of 10 Frizzleds (FZD1-10) and Smoothened (SMO). FZDs bind and are activated by secreted lipoglycoproteins of the Wingless/Int-1 (WNT) family, and SMO is indirectly activated by the Hedgehog (Hh) family of morphogens acting on the transmembrane protein Patched. The advance of our understanding of FZDs and SMO as dynamic transmembrane receptors and molecular machines, which emerged during the past 14 years since the first-class F GPCR IUPHAR nomenclature report, justifies an update. This article focuses on the advances in molecular pharmacology and structural biology providing new mechanistic insight into ligand recognition, receptor activation mechanisms, signal initiation, and signal specification. Furthermore, class F GPCRs continue to develop as drug targets, and novel technologies and tools such as genetically encoded biosensors and CRISP/Cas9 edited cell systems have contributed to refined functional analysis of these receptors. Also, advances in crystal structure analysis and cryogenic electron microscopy contribute to the rapid development of our knowledge about structure-function relationships, providing a great starting point for drug development. Despite the progress, questions and challenges remain to fully understand the complexity of the WNT/FZD and Hh/SMO signaling systems. SIGNIFICANCE STATEMENT: The recent years of research have brought about substantial functional and structural insight into mechanisms of activation of Frizzleds and Smoothened. While the advance furthers our mechanistic understanding of ligand recognition, receptor activation, signal specification, and initiation, broader opportunities emerge that allow targeting class F GPCRs for therapy and regenerative medicine employing both biologics and small molecule compounds.
Collapse
Affiliation(s)
- Gunnar Schulte
- Karolinska Institutet, Department of Physiology & Pharmacology, Receptor Biology & Signaling, Biomedicum, Stockholm, Sweden
| |
Collapse
|
21
|
Hoang HTM, George K, Ahmad M. Application of bioluminescence resonance energy transfer assays in primary mouse neuronal cultures. STAR Protoc 2024; 5:103228. [PMID: 39068655 PMCID: PMC11339249 DOI: 10.1016/j.xpro.2024.103228] [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: 04/25/2024] [Revised: 05/28/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is widely employed for real-time monitoring of G protein-coupled receptor activity, interactions, and trafficking in heterologous cell lines, yet its use in neuronal systems remains limited. Here, we present a protocol to apply BRET assays to primary neuronal cultures from mouse embryos. We describe steps and key concepts for generating plasmid constructs and lentivirus preparations, plating and lentiviral transduction of primary cultured neurons in 96-well plates, and BRET data collection and analysis. For complete details on the use and execution of this protocol, please refer to George et al.1.
Collapse
Affiliation(s)
- Hanh T M Hoang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kiran George
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mohiuddin Ahmad
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
22
|
Brands J, Bravo S, Jürgenliemke L, Grätz L, Schihada H, Frechen F, Alenfelder J, Pfeil C, Ohse PG, Hiratsuka S, Kawakami K, Schmacke LC, Heycke N, Inoue A, König G, Pfeifer A, Wachten D, Schulte G, Steinmetzer T, Watts VJ, Gomeza J, Simon K, Kostenis E. A molecular mechanism to diversify Ca 2+ signaling downstream of Gs protein-coupled receptors. Nat Commun 2024; 15:7684. [PMID: 39227390 PMCID: PMC11372221 DOI: 10.1038/s41467-024-51991-6] [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: 01/30/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
A long-held tenet in inositol-lipid signaling is that cleavage of membrane phosphoinositides by phospholipase Cβ (PLCβ) isozymes to increase cytosolic Ca2+ in living cells is exclusive to Gq- and Gi-sensitive G protein-coupled receptors (GPCRs). Here we extend this central tenet and show that Gs-GPCRs also partake in inositol-lipid signaling and thereby increase cytosolic Ca2+. By combining CRISPR/Cas9 genome editing to delete Gαs, the adenylyl cyclase isoforms 3 and 6, or the PLCβ1-4 isozymes, with pharmacological and genetic inhibition of Gq and G11, we pin down Gs-derived Gβγ as driver of a PLCβ2/3-mediated cytosolic Ca2+ release module. This module does not require but crosstalks with Gαs-dependent cAMP, demands Gαq to release PLCβ3 autoinhibition, but becomes Gq-independent with mutational disruption of the PLCβ3 autoinhibited state. Our findings uncover the key steps of a previously unappreciated mechanism utilized by mammalian cells to finetune their calcium signaling regulation through Gs-GPCRs.
Collapse
Affiliation(s)
- Julian Brands
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Sergi Bravo
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Lars Jürgenliemke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Research Training Group 2873, University of Bonn, Bonn, Germany
| | - Lukas Grätz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Fabian Frechen
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Cy Pfeil
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Research Training Group 1873, University of Bonn, Bonn, Germany
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Paul Georg Ohse
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Suzune Hiratsuka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, 153-8505, Japan
| | - Luna C Schmacke
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nina Heycke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Gabriele König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Dagmar Wachten
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Torsten Steinmetzer
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute of Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Jesús Gomeza
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
23
|
Sajkowska JJ, Tsang CH, Kozielewicz P. Application of FRET- and BRET-based live-cell biosensors in deorphanization and ligand discovery studies on orphan G protein-coupled receptors. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100174. [PMID: 39084335 DOI: 10.1016/j.slasd.2024.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Bioluminescence- and fluorescence-based resonance energy transfer assays have gained considerable attention in pharmacological research as high-throughput scalable tools applicable to drug discovery. To this end, G protein-coupled receptors represent the biggest target class for marketed drugs, and among them, orphan G protein-coupled receptors have the biggest untapped therapeutic potential. In this review, the cases where biophysical methods, BRET and FRET, were employed for deorphanization and ligand discovery studies on orphan G protein-coupled receptors are listed and discussed.
Collapse
Affiliation(s)
- Joanna J Sajkowska
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Choi Har Tsang
- Department of Physiology and Pharmacology, Molecular Pharmacology of GPCRs, Karolinska Institute, Stockholm, Sweden
| | - Paweł Kozielewicz
- Department of Physiology and Pharmacology, Molecular Pharmacology of GPCRs, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
24
|
Janicot R, Garcia-Marcos M. Get Ready to Sharpen Your Tools: A Short Guide to Heterotrimeric G Protein Activity Biosensors. Mol Pharmacol 2024; 106:129-144. [PMID: 38991745 PMCID: PMC11331509 DOI: 10.1124/molpharm.124.000949] [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: 05/21/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest class of transmembrane receptors encoded in the human genome, and they initiate cellular responses triggered by a plethora of extracellular stimuli ranging from neurotransmitters and hormones to photons. Upon stimulation, GPCRs activate heterotrimeric G proteins (Gαβγ) in the cytoplasm, which then convey signals to their effectors to elicit cellular responses. Given the broad biological and biomedical relevance of GPCRs and G proteins in physiology and disease, there is great interest in developing and optimizing approaches to measure their signaling activity with high accuracy and across experimental systems pertinent to their functions in cellular communication. This review provides a historical perspective on approaches to measure GPCR-G protein signaling, from quantification of second messengers and other indirect readouts of activity to biosensors that directly detect the activity of G proteins. The latter is the focus of a more detailed overview of the evolution of design principles for various optical biosensors of G protein activity with different experimental capabilities. We will highlight advantages and limitations of biosensors that detect different G protein activation hallmarks, like dissociation of Gα and Gβγ or nucleotide exchange on Gα, as well as their suitability to detect signaling mediated by endogenous versus exogenous signaling components or in physiologically relevant systems like primary cells. Overall, this review intends to provide an assessment of the state-of-the-art for biosensors that directly measure G protein activity to allow readers to make informed decisions on the selection and implementation of currently available tools. SIGNIFICANCE STATEMENT: G protein activity biosensors have become essential and widespread tools to assess GPCR signaling and pharmacology. Yet, investigators face the challenge of choosing from a growing list of G protein activity biosensors. This review provides an overview of the features and capabilities of different optical biosensor designs for the direct detection of G protein activity in cells, with the aim of facilitating the rational selection of systems that align with the specific scientific questions and needs of investigators.
Collapse
Affiliation(s)
- Remi Janicot
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine (R.J., M.G.-M.) and Department of Biology, College of Arts & Sciences (M.G.-M.), Boston University, Boston, Massachusetts
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine (R.J., M.G.-M.) and Department of Biology, College of Arts & Sciences (M.G.-M.), Boston University, Boston, Massachusetts
| |
Collapse
|
25
|
Franchini L, Porter JJ, Lueck JD, Orlandi C. Gz Enhanced Signal Transduction assaY (G ZESTY) for GPCR deorphanization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605282. [PMID: 39091869 PMCID: PMC11291178 DOI: 10.1101/2024.07.26.605282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
G protein-coupled receptors (GPCRs) are key pharmacological targets, yet many remain underutilized due to unknown activation mechanisms and ligands. Orphan GPCRs, lacking identified natural ligands, are a high priority for research, as identifying their ligands will aid in understanding their functions and potential as drug targets. Most GPCRs, including orphans, couple to Gi/o/z family members, however current assays to detect their activation are limited, hindering ligand identification efforts. We introduce GZESTY, a highly sensitive, cell-based assay developed in an easily deliverable format designed to study the pharmacology of Gi/o/z-coupled GPCRs and assist in deorphanization. We optimized assay conditions and developed an all-in-one vector employing novel cloning methods to ensure the correct expression ratio of GZESTY components. GZESTY successfully assessed activation of a library of ligand-activated GPCRs, detecting both full and partial agonism, as well as responses from endogenous GPCRs. Notably, with GZESTY we established the presence of endogenous ligands for GPR176 and GPR37 in brain extracts, validating its use in deorphanization efforts. This assay enhances the ability to find ligands for orphan GPCRs, expanding the toolkit for GPCR pharmacologists.
Collapse
Affiliation(s)
- Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Joseph J. Porter
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John D. Lueck
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
26
|
Gao M, Ooms JF, Leurs R, Vischer HF. Histamine H 3 Receptor Isoforms: Insights from Alternative Splicing to Functional Complexity. Biomolecules 2024; 14:761. [PMID: 39062475 PMCID: PMC11274711 DOI: 10.3390/biom14070761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Alternative splicing significantly enhances the diversity of the G protein-coupled receptor (GPCR) family, including the histamine H3 receptor (H3R). This post-transcriptional modification generates multiple H3R isoforms with potentially distinct pharmacological and physiological profiles. H3R is primarily involved in the presynaptic inhibition of neurotransmitter release in the central nervous system. Despite the approval of pitolisant for narcolepsy (Wakix®) and daytime sleepiness in adults with obstructive sleep apnea (Ozawade®) and ongoing clinical trials for other H3R antagonists/inverse agonists, the functional significance of the numerous H3R isoforms remains largely enigmatic. Recent publicly available RNA sequencing data have confirmed the expression of multiple H3R isoforms in the brain, with some isoforms exhibiting unique tissue-specific distribution patterns hinting at isoform-specific functions and interactions within neural circuits. In this review, we discuss the complexity of H3R isoforms with a focus on their potential roles in central nervous system (CNS) function. Comparative analysis across species highlights evolutionary conservation and divergence in H3R splicing, suggesting species-specific regulatory mechanisms. Understanding the functionality of H3R isoforms is crucial for the development of targeted therapeutics. This knowledge will inform the design of more precise pharmacological interventions, potentially enhancing therapeutic efficacy and reducing adverse effects in the treatment of neurological and psychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Henry F. Vischer
- Amsterdam Institute of Molecular and Life Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (M.G.); (J.F.O.); (R.L.)
| |
Collapse
|
27
|
George K, Hoang HT, Tibbs T, Nagaraja RY, Li G, Troyano-Rodriguez E, Ahmad M. Robust GRK2/3/6-dependent desensitization of oxytocin receptor in neurons. iScience 2024; 27:110047. [PMID: 38883814 PMCID: PMC11179071 DOI: 10.1016/j.isci.2024.110047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/22/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Oxytocin plays critical roles in the brain as a neuromodulator, regulating social and other affective behavior. However, the regulatory mechanisms controlling oxytocin receptor (OXTR) signaling in neurons remain unexplored. In this study, we have identified robust and rapid-onset desensitization of OXTR response in multiple regions of the mouse brain. Both cell autonomous spiking response and presynaptic activation undergo similar agonist-induced desensitization. G-protein-coupled receptor kinases (GRK) GRK2, GRK3, and GRK6 are recruited to the activated OXTR in neurons, followed by recruitment of β-arrestin-1 and -2. Neuronal OXTR desensitization was impaired by suppression of GRK2/3/6 kinase activity but remained unaltered with double knockout of β-arrestin-1 and -2. Additionally, we observed robust agonist-induced internalization of neuronal OXTR and its Rab5-dependent recruitment to early endosomes, which was impaired by GRK2/3/6 inhibition. This work defines distinctive aspects of the mechanisms governing OXTR desensitization and internalization in neurons compared to prior studies in heterologous cells.
Collapse
Affiliation(s)
- Kiran George
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hanh T.M. Hoang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Taryn Tibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Raghavendra Y. Nagaraja
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Guangpu Li
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eva Troyano-Rodriguez
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mohiuddin Ahmad
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
28
|
Solis GP, Koval A, Valnohova J, Kazemzadeh A, Savitsky M, Katanaev VL. Neomorphic Gαo mutations gain interaction with Ric8 proteins in GNAO1 encephalopathies. J Clin Invest 2024; 134:e172057. [PMID: 38874642 PMCID: PMC11291268 DOI: 10.1172/jci172057] [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: 05/15/2023] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
GNAO1 mutated in pediatric encephalopathies encodes the major neuronal G protein Gαo. Of the more than 80 pathogenic mutations, most are single amino acid substitutions spreading across the Gαo sequence. We performed extensive characterization of Gαo mutants, showing abnormal GTP uptake and hydrolysis and deficiencies in binding Gβγ and RGS19. Plasma membrane localization of Gαo was decreased for a subset of mutations that leads to epilepsy; dominant interactions with GPCRs also emerged for the more severe mutants. Pathogenic mutants massively gained interaction with Ric8A and, surprisingly, Ric8B proteins, relocalizing them from cytoplasm to Golgi. Of these 2 mandatory Gα-subunit chaperones, Ric8A is normally responsible for the Gαi/Gαo, Gαq, and Gα12/Gα13 subfamilies, and Ric8B solely responsible for Gαs/Gαolf. Ric8 mediates the disease dominance when engaging in neomorphic interactions with pathogenic Gαo through imbalance of the neuronal G protein signaling networks. As the strength of Gαo-Ric8B interactions correlates with disease severity, our study further identifies an efficient biomarker and predictor for clinical manifestations in GNAO1 encephalopathies. Our work uncovers the neomorphic molecular mechanism of mutations underlying pediatric encephalopathies and offers insights into other maladies caused by G protein malfunctioning and further genetic diseases.
Collapse
Affiliation(s)
- Gonzalo P. Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jana Valnohova
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arghavan Kazemzadeh
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mikhail Savitsky
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- School of Medicine and Life Sciences, Department of Pharmacy and Pharmacology, Far Eastern Federal University, Vladivostok, Russia
| |
Collapse
|
29
|
Schüß C, Behr V, Beck-Sickinger AG. Illuminating the neuropeptide Y 4 receptor and its ligand pancreatic polypeptide from a structural, functional, and therapeutic perspective. Neuropeptides 2024; 105:102416. [PMID: 38430725 DOI: 10.1016/j.npep.2024.102416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
The neuropeptide Y4 receptor (Y4R), a rhodopsin-like G protein-coupled receptor (GPCR) and the hormone pancreatic polypeptide (PP) are members of the neuropeptide Y family consisting of four receptors (Y1R, Y2R, Y4R, Y5R) and three highly homologous peptide ligands (neuropeptide Y, peptide YY, PP). In this family, the Y4R is of particular interest as it is the only subtype with high affinity to PP over NPY. The Y4R, as a mediator of PP signaling, has a pivotal role in appetite regulation and energy homeostasis, offering potential avenues for the treatment of metabolic disorders such as obesity. PP as anorexigenic peptide is released postprandial from the pancreas in response to food intake, induces satiety signals and contributes to hamper excessive food intake. Moreover, this system was also described to be associated with different types of cancer: overexpression of Y4R have been found in human adenocarcinoma cells, while elevated levels of PP are related to the development of pancreatic endocrine tumors. The pharmacological relevance of the Y4R advanced the search for potent and selective ligands for this receptor subtype, which will be significantly progressed through the elucidation of the active state PP-Y4R cryo-EM structure. This review summarizes the development of novel PP-derived ligands, like Obinepitide as dual Y2R/Y4R agonist in clinical trials or UR-AK86c as small hexapeptide agonist with picomolar affinity, as well as the first allosteric modulators that selectively target the Y4R, e.g. VU0506013 as potent Y4R positive allosteric modulator or (S)-VU0637120 as allosteric antagonist. Here, we provide valuable insights into the complex physiological functions of the Y4R and PP and the pharmacological relevance of the system in appetite regulation to open up new avenues for the development of tool compounds for targeted therapies with potential applications in metabolic disorders.
Collapse
Affiliation(s)
- Corinna Schüß
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Germany.
| | - Victoria Behr
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Germany
| | | |
Collapse
|
30
|
Lim VJY, Gerber HD, Schihada H, Trinh VT, Hilger D, Vázquez O, Kolb P. A virtual library of small molecules mimicking dipeptides. Arch Pharm (Weinheim) 2024; 357:e2300636. [PMID: 38332463 DOI: 10.1002/ardp.202300636] [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: 11/01/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Virtual combinatorial libraries are prevalent in drug discovery due to improvements in the prediction of synthetic reactions that can be performed. This has gone hand in hand with the development of virtual screening capabilities to effectively screen the large chemical spaces spanned by exhaustive enumeration of reaction products. In this study, we generated a small-molecule dipeptide mimic library to target proteins binding small peptides. The library was created based on the general idea of peptide synthesis, that is, amino acid mimics were reacted in silico to form the dipeptide mimics, yielding 2,036,819 unique compounds. After docking calculations, two compounds from the library were synthesized and tested against WD repeat-containing protein 5 (WDR5) and histamine receptors H1-H4 to evaluate whether these molecules are viable in assays. The compounds showed the highest potency at the histamine H3 receptor, with Ki values in the two-digit micromolar range.
Collapse
Affiliation(s)
- Victor Jun Yu Lim
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Hans-Dieter Gerber
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Hannes Schihada
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Van Tuan Trinh
- Chemical Biology, Department of Chemistry, University of Marburg, Marburg, Germany
| | - Daniel Hilger
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
| | - Olalla Vázquez
- Chemical Biology, Department of Chemistry, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), University of Marburg, Marburg, Germany
| | - Peter Kolb
- Pharmaceutical Chemistry, Department of Pharmacy, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), University of Marburg, Marburg, Germany
| |
Collapse
|
31
|
Ma S, Yin X, Pin JP, Rondard P, Yi P, Liu J. Absence of calcium-sensing receptor basal activity due to inter-subunit disulfide bridges. Commun Biol 2024; 7:501. [PMID: 38664468 PMCID: PMC11045811 DOI: 10.1038/s42003-024-06189-3] [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: 08/21/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
G protein-coupled receptors naturally oscillate between inactive and active states, often resulting in receptor constitutive activity with important physiological consequences. Among the class C G protein-coupled receptors that typically sense amino-acids and their derivatives, the calcium sensing receptor (CaSR) tightly controls blood calcium levels. Its constitutive activity has not yet been studied. Here, we demonstrate the importance of the inter-subunit disulfide bridges in maintaining the inactive state of CaSR, resulting in undetectable constitutive activity, unlike the other class C receptors. Deletion of these disulfide bridges results in strong constitutive activity that is abolished by mutations preventing amino acid binding. It shows that this inter-subunit disulfide link is necessary to limit the agonist effect of amino acids on CaSR. Furthermore, human genetic mutations deleting these bridges and associated with hypocalcemia result in elevated CaSR constitutive activity. These results highlight the physiological importance of fine tuning the constitutive activity of G protein-coupled receptors.
Collapse
Affiliation(s)
- Shumin Ma
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, and College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xueliang Yin
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, and College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, Cedex 5, France
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, Cedex 5, France.
| | - Ping Yi
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, and College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jianfeng Liu
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, and College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
32
|
Aydin A, Klenk C, Nemec K, Işbilir A, Martin LM, Zauber H, Rrustemi T, Toka HR, Schuster H, Gong M, Stricker S, Bock A, Bähring S, Selbach M, Lohse MJ, Luft FC. ADAM19 cleaves the PTH receptor and associates with brachydactyly type E. Life Sci Alliance 2024; 7:e202302400. [PMID: 38331475 PMCID: PMC10853454 DOI: 10.26508/lsa.202302400] [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: 09/27/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Brachydactyly type E (BDE), shortened metacarpals, metatarsals, cone-shaped epiphyses, and short stature commonly occurs as a sole phenotype. Parathyroid hormone-like protein (PTHrP) has been shown to be responsible in all forms to date, either directly or indirectly. We used linkage and then whole genome sequencing in a small pedigree, to elucidate BDE and identified a truncated disintegrin-and-metalloproteinase-19 (ADAM19) allele in all affected family members, but not in nonaffected persons. Since we had shown earlier that the extracellular domain of the parathyroid hormone receptor (PTHR1) is subject to an unidentified metalloproteinase cleavage, we tested the hypothesis that ADAM19 is a sheddase for PTHR1. WT ADAM19 cleaved PTHR1, while mutated ADAM-19 did not. We mapped the cleavage site that we verified with mass spectrometry between amino acids 64-65. ADAM-19 cleavage increased Gq and decreased Gs activation. Moreover, perturbed PTHR1 cleavage by ADAM19 increased ß-arrestin2 recruitment, while cAMP accumulation was not altered. We suggest that ADAM19 serves as a regulatory element for PTHR1 and could be responsible for BDE. This sheddase may affect other PTHrP or PTH-related functions.
Collapse
Affiliation(s)
- Atakan Aydin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Christoph Klenk
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Katarina Nemec
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Department of Structural Biology and Center of Excellence for Data-Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ali Işbilir
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Lisa M Martin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Henrik Zauber
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Trendelina Rrustemi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hakan R Toka
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Herbert Schuster
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Maolian Gong
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Andreas Bock
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sylvia Bähring
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Matthias Selbach
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Martin J Lohse
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- ISAR Bioscience Institute, Munich, Germany
| | - Friedrich C Luft
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
33
|
Xia R, Shi S, Xu Z, Vischer HF, Windhorst AD, Qian Y, Duan Y, Liang J, Chen K, Zhang A, Guo C, Leurs R, He Y. Structural basis of ligand recognition and design of antihistamines targeting histamine H 4 receptor. Nat Commun 2024; 15:2493. [PMID: 38509098 PMCID: PMC10954740 DOI: 10.1038/s41467-024-46840-5] [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: 09/18/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The histamine H4 receptor (H4R) plays key role in immune cell function and is a highly valued target for treating allergic and inflammatory diseases. However, structural information of H4R remains elusive. Here, we report four cryo-EM structures of H4R/Gi complexes, with either histamine or synthetic agonists clobenpropit, VUF6884 and clozapine bound. Combined with mutagenesis, ligand binding and functional assays, the structural data reveal a distinct ligand binding mode where D943.32 and a π-π network determine the orientation of the positively charged group of ligands, while E1825.46, located at the opposite end of the ligand binding pocket, plays a key role in regulating receptor activity. The structural insight into H4R ligand binding allows us to identify mutants at E1825.46 for which the agonist clobenpropit acts as an inverse agonist and to correctly predict inverse agonism of a closely related analog with nanomolar potency. Together with the findings regarding receptor activation and Gi engagement, we establish a framework for understanding H4R signaling and provide a rational basis for designing novel antihistamines targeting H4R.
Collapse
Affiliation(s)
- Ruixue Xia
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Shuang Shi
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
| | - Zhenmei Xu
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Henry F Vischer
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Yu Qian
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yaning Duan
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jiale Liang
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kai Chen
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Anqi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Changyou Guo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Rob Leurs
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands.
| | - Yuanzheng He
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China.
- Frontiers Science Center for Matter Behave in Space Environment, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
34
|
Mönnich D, Humphrys LJ, Höring C, Hoare BL, Forster L, Pockes S. Activation of Multiple G Protein Pathways to Characterize the Five Dopamine Receptor Subtypes Using Bioluminescence Technology. ACS Pharmacol Transl Sci 2024; 7:834-854. [PMID: 38481695 PMCID: PMC10928903 DOI: 10.1021/acsptsci.3c00339] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 11/01/2024]
Abstract
G protein-coupled receptors show preference for G protein subtypes but can recruit multiple G proteins with various downstream signaling cascades. This functional selection can guide drug design. Dopamine receptors are both stimulatory (D1-like) and inhibitory (D2-like) with diffuse expression across the central nervous system. Functional selectivity of G protein subunits may help with dopamine receptor targeting and their downstream effects. Three bioluminescence-based assays were used to characterize G protein coupling and function with the five dopamine receptors. Most proximal to ligand binding was the miniG protein assay with split luciferase technology used to measure recruitment. For endogenous and selective ligands, the G-CASE bioluminescence resonance energy transfer (BRET) assay measured G protein activation and receptor selectivity. Downstream, the BRET-based CAMYEN assay quantified cyclic adenosine monophosphate (cAMP) changes. Several dopamine receptor agonists and antagonists were characterized for their G protein recruitment and cAMP effects. G protein selectivity with dopamine revealed potential Gq coupling at all five receptors, as well as the ability to activate subtypes with the "opposite" effects to canonical signaling. D1-like receptor agonist (+)-SKF-81297 and D2-like receptor agonist pramipexole showed selectivity at all receptors toward Gs or Gi/o/z activation, respectively. The five dopamine receptors show a wide range of potentials for G protein coupling and activation, reflected in their downstream cAMP signaling. Targeting these interactions can be achieved through drug design. This opens the door to pharmacological treatment with more selectivity options for inducing the correct physiological events.
Collapse
Affiliation(s)
- Denise Mönnich
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Laura J. Humphrys
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Carina Höring
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Bradley L. Hoare
- Florey
Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lisa Forster
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Steffen Pockes
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| |
Collapse
|
35
|
Xu C, Zhou Y, Liu Y, Lin L, Liu P, Wang X, Xu Z, Pin JP, Rondard P, Liu J. Specific pharmacological and G i/o protein responses of some native GPCRs in neurons. Nat Commun 2024; 15:1990. [PMID: 38443355 PMCID: PMC10914727 DOI: 10.1038/s41467-024-46177-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/15/2024] [Indexed: 03/07/2024] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest family of membrane proteins and are important drug targets. The discovery of drugs targeting these receptors and their G protein signaling properties are based on assays mainly performed with modified receptors expressed in heterologous cells. However, GPCR responses may differ in their native environment. Here, by using highly sensitive Gi/o sensors, we reveal specific properties of Gi/o protein-mediated responses triggered by GABAB, α2 adrenergic and cannabinoid CB1 receptors in primary neurons, different from those in heterologous cells. These include different profiles in the Gi/o protein subtypes-mediated responses, and differences in the potencies of some ligands even at similar receptor expression levels. Altogether, our results show the importance of using biosensors compatible with primary cells for evaluating the activities of endogenous GPCRs in their native environment.
Collapse
Affiliation(s)
- Chanjuan Xu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005, Guangzhou, China
| | - Yiwei Zhou
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Kindstar Global Precision Medicine Institute, Wuhan, China
| | - Yuxuan Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Lin
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomei Wang
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhengyuan Xu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34094, Montpellier, France.
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34094, Montpellier, France.
| | - Jianfeng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005, Guangzhou, China.
| |
Collapse
|
36
|
Köck Z, Schnelle K, Persechino M, Umbach S, Schihada H, Januliene D, Parey K, Pockes S, Kolb P, Dötsch V, Möller A, Hilger D, Bernhard F. Cryo-EM structure of cell-free synthesized human histamine 2 receptor/G s complex in nanodisc environment. Nat Commun 2024; 15:1831. [PMID: 38418462 PMCID: PMC10901899 DOI: 10.1038/s41467-024-46096-z] [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: 07/13/2023] [Accepted: 02/14/2024] [Indexed: 03/01/2024] Open
Abstract
Here we describe the cryo-electron microscopy structure of the human histamine 2 receptor (H2R) in an active conformation with bound histamine and in complex with Gs heterotrimeric protein at an overall resolution of 3.4 Å. The complex was generated by cotranslational insertion of the receptor into preformed nanodisc membranes using cell-free synthesis in E. coli lysates. Structural comparison with the inactive conformation of H2R and the inactive and Gq-coupled active state of H1R together with structure-guided functional experiments reveal molecular insights into the specificity of ligand binding and G protein coupling for this receptor family. We demonstrate lipid-modulated folding of cell-free synthesized H2R, its agonist-dependent internalization and its interaction with endogenously synthesized H1R and H2R in HEK293 cells by applying a recently developed nanotransfer technique.
Collapse
Affiliation(s)
- Zoe Köck
- Centre for Biomolecular Magnetic Resonance, Institute for Biophysical Chemistry, Goethe-University of Frankfurt/Main, Frankfurt, Germany
| | - Kilian Schnelle
- Department of Biology/Chemistry, Structural Biology section, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | | | - Simon Umbach
- Centre for Biomolecular Magnetic Resonance, Institute for Biophysical Chemistry, Goethe-University of Frankfurt/Main, Frankfurt, Germany
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Dovile Januliene
- Department of Biology/Chemistry, Structural Biology section, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Kristian Parey
- Department of Biology/Chemistry, Structural Biology section, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Steffen Pockes
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Volker Dötsch
- Centre for Biomolecular Magnetic Resonance, Institute for Biophysical Chemistry, Goethe-University of Frankfurt/Main, Frankfurt, Germany
| | - Arne Möller
- Department of Biology/Chemistry, Structural Biology section, University of Osnabrück, Osnabrück, Germany.
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), University of Osnabrück, Osnabrück, Germany.
| | - Daniel Hilger
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany.
| | - Frank Bernhard
- Centre for Biomolecular Magnetic Resonance, Institute for Biophysical Chemistry, Goethe-University of Frankfurt/Main, Frankfurt, Germany.
| |
Collapse
|
37
|
Jones AJY, Harman TH, Harris M, Lewis OE, Ladds G, Nietlispach D. Binding kinetics drive G protein subtype selectivity at the β 1-adrenergic receptor. Nat Commun 2024; 15:1334. [PMID: 38351103 PMCID: PMC10864275 DOI: 10.1038/s41467-024-45680-7] [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: 08/25/2023] [Accepted: 02/01/2024] [Indexed: 02/16/2024] Open
Abstract
G protein-coupled receptors (GPCRs) bind to different G protein α-subtypes with varying degrees of selectivity. The mechanism by which GPCRs achieve this selectivity is still unclear. Using 13C methyl methionine and 19F NMR, we investigate the agonist-bound active state of β1AR and its ternary complexes with different G proteins in solution. We find the receptor in the ternary complexes adopts very similar conformations. In contrast, the full agonist-bound receptor active state assumes a conformation differing from previously characterised activation intermediates or from β1AR in ternary complexes. Assessing the kinetics of binding for the agonist-bound receptor with different G proteins, we find the increased affinity of β1AR for Gs results from its much faster association with the receptor. Consequently, we suggest a kinetic-driven selectivity gate between canonical and secondary coupling which arises from differential favourability of G protein binding to the agonist-bound receptor active state.
Collapse
Affiliation(s)
- Andrew J Y Jones
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Thomas H Harman
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Matthew Harris
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Oliver E Lewis
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Daniel Nietlispach
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK.
| |
Collapse
|
38
|
Abdul-Ridha A, de Zhang LA, Betrie AH, Deluigi M, Vaid TM, Whitehead A, Zhang Y, Davis B, Harris R, Simmonite H, Hubbard RE, Gooley PR, Plückthun A, Bathgate RA, Chalmers DK, Scott DJ. Identification of a Novel Subtype-Selective α 1B-Adrenoceptor Antagonist. ACS Chem Neurosci 2024; 15:671-684. [PMID: 38238043 PMCID: PMC10854767 DOI: 10.1021/acschemneuro.3c00767] [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: 11/29/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 02/08/2024] Open
Abstract
α1A-, α1B-, and α1D-adrenoceptors (α1-ARs) are members of the adrenoceptor G protein-coupled receptor family that are activated by adrenaline (epinephrine) and noradrenaline. α1-ARs are clinically targeted using antagonists that have minimal subtype selectivity, such as prazosin and tamsulosin, to treat hypertension and benign prostatic hyperplasia, respectively. Abundant expression of α1-ARs in the heart and central nervous system (CNS) makes these receptors potential targets for the treatment of cardiovascular and CNS disorders, such as heart failure, epilepsy, and Alzheimer's disease. Our understanding of the precise physiological roles of α1-ARs, however, and their involvement in disease has been hindered by the lack of sufficiently subtype-selective tool compounds, especially for α1B-AR. Here, we report the discovery of 4-[(2-hydroxyethyl)amino]-6-methyl-2H-chromen-2-one (Cpd1), as an α1B-AR antagonist that has 10-15-fold selectivity over α1A-AR and α1D-AR. Through computational and site-directed mutagenesis studies, we have identified the binding site of Cpd1 in α1B-AR and propose the molecular basis of α1B-AR selectivity, where the nonconserved V19745.52 residue plays a major role, with contributions from L3146.55 within the α1B-AR pocket. By exploring the structure-activity relationships of Cpd1 at α1B-AR, we have also identified 3-[(cyclohexylamino)methyl]-6-methylquinolin-2(1H)-one (Cpd24), which has a stronger binding affinity than Cpd1, albeit with reduced selectivity for α1B-AR. Cpd1 and Cpd24 represent potential leads for α1B-AR-selective drug discovery and novel tool molecules to further study the physiology of α1-ARs.
Collapse
Affiliation(s)
- Alaa Abdul-Ridha
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lazarus A. de Zhang
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | | | - Mattia Deluigi
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Tasneem M. Vaid
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21
Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alice Whitehead
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Yifan Zhang
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Ben Davis
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
| | - Richard Harris
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
| | | | - Roderick E. Hubbard
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - Paul R. Gooley
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21
Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Plückthun
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Ross A.D. Bathgate
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - David K. Chalmers
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Daniel J. Scott
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
39
|
Nagl M, Mönnich D, Rosier N, Schihada H, Sirbu A, Konar N, Reyes-Resina I, Navarro G, Franco R, Kolb P, Annibale P, Pockes S. Fluorescent Tools for the Imaging of Dopamine D 2 -Like Receptors. Chembiochem 2024; 25:e202300659. [PMID: 37942961 DOI: 10.1002/cbic.202300659] [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: 09/26/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/10/2023]
Abstract
The family of dopamine D2 -like receptors represents an interesting target for a variety of neurological diseases, e. g. Parkinson's disease (PD), addiction, or schizophrenia. In this study we describe the synthesis of a new set of fluorescent ligands as tools for visualization of dopamine D2 -like receptors. Pharmacological characterization in radioligand binding studies identified UR-MN212 (20) as a high-affinity ligand for D2 -like receptors (pKi (D2long R)=8.24, pKi (D3 R)=8.58, pKi (D4 R)=7.78) with decent selectivity towards D1 -like receptors. Compound 20 is a neutral antagonist in a Go1 activation assay at the D2long R, D3 R, and D4 R, which is an important feature for studies using whole cells. The neutral antagonist 20, equipped with a 5-TAMRA dye, displayed rapid association to the D2long R in binding studies using confocal microscopy demonstrating its suitability for fluorescence microscopy. Furthermore, in molecular brightness studies, the ligand's binding affinity could be determined in a single-digit nanomolar range that was in good agreement with radioligand binding data. Therefore, the fluorescent compound can be used for quantitative characterization of native D2 -like receptors in a broad variety of experimental setups.
Collapse
Affiliation(s)
- Martin Nagl
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Denise Mönnich
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Niklas Rosier
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Alexei Sirbu
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Nergis Konar
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Irene Reyes-Resina
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Franco
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
- School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, Scotland
| | - Steffen Pockes
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| |
Collapse
|
40
|
Rosier N, Mönnich D, Nagl M, Schihada H, Sirbu A, Konar N, Reyes-Resina I, Navarro G, Franco R, Kolb P, Annibale P, Pockes S. Shedding Light on the D 1 -Like Receptors: A Fluorescence-Based Toolbox for Visualization of the D 1 and D 5 Receptors. Chembiochem 2024; 25:e202300658. [PMID: 37983731 DOI: 10.1002/cbic.202300658] [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: 09/26/2023] [Revised: 11/08/2023] [Indexed: 11/22/2023]
Abstract
Dopamine D1 -like receptors are the most abundant type of dopamine receptors in the central nervous system and, even after decades of discovery, still highly interesting for the study of neurological diseases. We herein describe the synthesis of a new set of fluorescent ligands, structurally derived from D1 R antagonist SCH-23390 and labeled with two different fluorescent dyes, as tool compounds for the visualization of D1 -like receptors. Pharmacological characterization in radioligand binding studies identified UR-NR435 (25) as a high-affinity ligand for D1 -like receptors (pKi (D1 R)=8.34, pKi (D5 R)=7.62) with excellent selectivity towards D2 -like receptors. Compound 25 proved to be a neutral antagonist at the D1 R and D5 R in a Gs heterotrimer dissociation assay, an important feature to avoid receptor internalization and degradation when working with whole cells. The neutral antagonist 25 displayed rapid association and complete dissociation to the D1 R in kinetic binding studies using confocal microscopy verifying its applicability for fluorescence microscopy. Moreover, molecular brightness studies determined a single-digit nanomolar binding affinity of the ligand, which was in good agreement with radioligand binding data. For this reason, this fluorescent ligand is a useful tool for a sophisticated characterization of native D1 receptors in a variety of experimental setups.
Collapse
Affiliation(s)
- Niklas Rosier
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Denise Mönnich
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Martin Nagl
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Alexei Sirbu
- Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Nergis Konar
- Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Irene Reyes-Resina
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Franco
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- School of Physics and Astronomy, University of St. Andrews, North Haugh, St. Andrews, Scotland, UK
| | - Steffen Pockes
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN 55414, USA
| |
Collapse
|
41
|
Demby A, Zaccolo M. Investigating G-protein coupled receptor signalling with light-emitting biosensors. Front Physiol 2024; 14:1310197. [PMID: 38260094 PMCID: PMC10801095 DOI: 10.3389/fphys.2023.1310197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are the most frequent target of currently approved drugs and play a central role in both physiological and pathophysiological processes. Beyond the canonical understanding of GPCR signal transduction, the importance of receptor conformation, beta-arrestin (β-arr) biased signalling, and signalling from intracellular locations other than the plasma membrane is becoming more apparent, along with the tight spatiotemporal compartmentalisation of downstream signals. Fluorescent and bioluminescent biosensors have played a pivotal role in elucidating GPCR signalling events in live cells. To understand the mechanisms of action of the GPCR-targeted drugs currently available, and to develop new and better GPCR-targeted therapeutics, understanding these novel aspects of GPCR signalling is critical. In this review, we present some of the tools available to interrogate each of these features of GPCR signalling, we illustrate some of the key findings which have been made possible by these tools and we discuss their limitations and possible developments.
Collapse
Affiliation(s)
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Pillaiyar T, Wozniak M, Abboud D, Rasch A, Liebing AD, Poso A, Kronenberger T, Stäubert C, Laufer SA, Hanson J. Development of Ligands for the Super Conserved Orphan G Protein-Coupled Receptor GPR27 with Improved Efficacy and Potency. J Med Chem 2023; 66:17118-17137. [PMID: 38060818 DOI: 10.1021/acs.jmedchem.3c02030] [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: 12/29/2023]
Abstract
The orphan G protein-coupled receptor GPR27 appears to play a role in insulin production, secretion, lipid metabolism, neuronal plasticity, and l-lactate homeostasis. However, investigations on the function of GPR27 are impaired by the lack of potent and efficacious agonists. We describe herein the development of di- and trisubstituted benzamide derivatives 4a-e, 7a-z, and 7aa-ai, which display GPR27-specific activity in a β-arrestin 2 recruitment-based assay. Highlighted compounds are PT-91 (7p: pEC50 6.15; Emax 100%) and 7ab (pEC50 6.56; Emax 99%). A putative binding mode was revealed by the docking studies of 7p and 7ab with a GPR27 homology model. The novel active compounds exhibited no GPR27-mediated activation of G proteins, indicating that the receptor may possess an atypical profile. Compound 7p displays high metabolic stability and brain exposure in mice. Thus, 7p represents a novel tool to investigate the elusive pharmacology of GPR27 and assess its potential as a drug target.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Monika Wozniak
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liège, B-4000 Liège, Belgium
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liège, B-4000 Liège, Belgium
| | - Alexander Rasch
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Aenne-Dorothea Liebing
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Antti Poso
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided & Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided & Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Claudia Stäubert
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Stefan A Laufer
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided & Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
| | - Julien Hanson
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liège, B-4000 Liège, Belgium
- Laboratory of Medicinal Chemistry, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, B-4000 Liège, Belgium
| |
Collapse
|
43
|
Voss JH. Recommended Tool Compounds: Application of YM-254890 and FR900359 to Interrogate Gα q/11-Mediated Signaling Pathways. ACS Pharmacol Transl Sci 2023; 6:1790-1800. [PMID: 38093837 PMCID: PMC10714435 DOI: 10.1021/acsptsci.3c00214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2024]
Abstract
The macrocyclic depsipeptides YM-254890 (YM) and FR900359 (FR) are natural products, which inhibit heterotrimeric Gαq/11 proteins with high potency and outstanding selectivity. Historically, pharmacological modulation of Gα proteins was only achieved by treatment with pertussis toxin and cholera toxin, whose application can be tedious and is restricted to the inhibition of Gαi/o proteins and activation of Gαs proteins, respectively. The breakthrough discovery and characterization of YM and FR rendered the closely related Gαq, Gα11, and Gα14 proteins amenable to pharmacological inhibition, and since then, both compounds have become widely used in molecular pharmacology and were also proven to be efficacious in animal models of disease. In the past years, both YM and FR were thoroughly characterized and have substantially contributed to an improved understanding of Gαq/11 signaling on a molecular and cellular level. Yet, the possibilities to interrogate Gαq/11 signaling in complex systems have only been exploited in a very limited number of studies, whose promising initial results warrant further application of YM and FR in basic and translational research. As both compounds have become commercially available as of late, this review focuses on their application in cell-based assays and in vivo systems, highlighting their qualities as tool compounds and providing instructions for their use.
Collapse
Affiliation(s)
- Jan Hendrik Voss
- Department of Physiology and Pharmacology,
Section of Receptor Biology and Signaling, Karolinska Institutet, S-171 65 Stockholm, Sweden
| |
Collapse
|
44
|
Wang N, Qian Y, Xia R, Zhu X, Xiong Y, Zhang A, Guo C, He Y. Structural basis of CD97 activation and G-protein coupling. Cell Chem Biol 2023; 30:1343-1353.e5. [PMID: 37673067 DOI: 10.1016/j.chembiol.2023.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
CD97 (ADGRE5) is an adhesion G protein-coupled receptor (aGPCR) which plays crucial roles in immune system and cancer. However, the mechanism of CD97 activation and the determinant of G13 coupling selectivity remain unknown. Here, we present the cryo-electron microscopy structures of human CD97 in complex with G13, Gq, and Gs. Our structures reveal the stalk peptide recognition mode of CD97, adding missing information of the current tethered-peptide activation model of aGPCRs. For instance, a revised "FXφφφ" motif and a framework of conserved aromatic residues in the ligand-binding pocket. Importantly, structural comparisons of G13, Gq, and Gs engagements of CD97 reveal key determinants of G13 coupling selectivity, where a deep insertion of the α helix 5 and a closer contact with the transmembrane helix 6, 5, and 3 dictate coupling preferences. Taken together, our structural study of CD97 provides a framework for understanding CD97 signaling and the G13 coupling selectivity.
Collapse
Affiliation(s)
- Na Wang
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yu Qian
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ruixue Xia
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xinyan Zhu
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yangjie Xiong
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Anqi Zhang
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Changyou Guo
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yuanzheng He
- Laboratory of Receptor Structure and Signaling, HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|
45
|
Bresinsky M, Shahraki A, Kolb P, Pockes S, Schihada H. Development of Fluorescent AF64394 Analogues Enables Real-Time Binding Studies for the Orphan Class A GPCR GPR3. J Med Chem 2023; 66:15025-15041. [PMID: 37907069 PMCID: PMC10641823 DOI: 10.1021/acs.jmedchem.3c01707] [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: 09/14/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023]
Abstract
The orphan G protein-coupled receptor (oGPCR) GPR3 represents a potential drug target for the treatment of Alzheimer's disease and metabolic disorders. However, the limited toolbox of pharmacological assays hampers the development of advanced ligands. Here, we developed a signaling pathway-independent readout of compound-GPR3 interaction. Starting from computational binding pose predictions of the most potent GPR3 ligand, we designed a series of fluorescent AF64394 analogues and assessed their suitability for BRET-based binding studies. The most potent ligand, 45 (UR-MB-355), bound to GPR3 and closely related receptors, GPR6 and GPR12, with similar submicromolar affinities. Furthermore, we found that 45 engages GPR3 in a distinct mode compared to AF64394, and coincubation studies with the GPR3 agonist diphenyleneiodonium chloride revealed allosteric modulation of 45 binding. These insights provide new cues for the pharmacological manipulation of GPR3 activity. This novel binding assay will foster the development of future drugs acting through these pharmacologically attractive oGPCRs.
Collapse
Affiliation(s)
- Merlin Bresinsky
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Aida Shahraki
- Department
of Pharmaceutical Chemistry, University
of Marburg, Marbacher Weg 8, 35032 Marburg, Germany
| | - Peter Kolb
- Department
of Pharmaceutical Chemistry, University
of Marburg, Marbacher Weg 8, 35032 Marburg, Germany
| | - Steffen Pockes
- Institute
of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
- Department
of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Hannes Schihada
- Department
of Pharmaceutical Chemistry, University
of Marburg, Marbacher Weg 8, 35032 Marburg, Germany
| |
Collapse
|
46
|
Kaczmarek I, Wower I, Ettig K, Kuhn CK, Kraft R, Landgraf K, Körner A, Schöneberg T, Horn S, Thor D. Identifying G protein-coupled receptors involved in adipose tissue function using the innovative RNA-seq database FATTLAS. iScience 2023; 26:107841. [PMID: 37766984 PMCID: PMC10520334 DOI: 10.1016/j.isci.2023.107841] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/26/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
G protein-coupled receptors (GPCRs) modulate the function of adipose tissue (AT) in general and of adipocytes, specifically. Although it is well-established that GPCRs are widely expressed in AT, their repertoire as well as their regulation and function in (patho)physiological conditions (e.g., obesity) is not fully resolved. Here, we established FATTLAS, an interactive public database, for improved access and analysis of RNA-seq data of mouse and human AT. After extracting the GPCRome of non-obese and obese individuals, highly expressed and differentially regulated GPCRs were identified. Exemplarily, we describe four receptors (GPR146, MRGPRF, FZD5, PTGER2) and analyzed their functions in a (pre)adipocyte cell model. Besides all receptors being involved in adipogenesis, MRGPRF is essential for adipocyte viability and regulates cAMP levels, while GPR146 modulates adipocyte lipolysis via constitutive activation of Gi proteins. Taken together, by implementing and using FATTLAS we describe four hitherto unrecognized GPCRs associated with AT function and adipogenesis.
Collapse
Affiliation(s)
- Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Isabel Wower
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Katja Ettig
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Christina Katharina Kuhn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Robert Kraft
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- School of Medicine, University of Global Health Equity (UGHE), Kigali, Rwanda
| | - Susanne Horn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, 45122 Essen, Germany
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
47
|
Larasati YA, Solis GP, Koval A, Griffiths ST, Berentsen R, Aukrust I, Lesca G, Chatron N, Ville D, Korff CM, Katanaev VL. Clinical Cases and the Molecular Profiling of a Novel Childhood Encephalopathy-Causing GNAO1 Mutation P170R. Cells 2023; 12:2469. [PMID: 37887313 PMCID: PMC10605901 DOI: 10.3390/cells12202469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
De novo mutations in GNAO1, the gene encoding the major neuronal G protein Gαo, cause a spectrum of pediatric encephalopathies with seizures, motor dysfunction, and developmental delay. Of the >80 distinct missense pathogenic variants, many appear to uniformly destabilize the guanine nucleotide handling of the mutant protein, speeding up GTP uptake and deactivating GTP hydrolysis. Zinc supplementation emerges as a promising treatment option for this disease, as Zn2+ ions reactivate the GTP hydrolysis on the mutant Gαo and restore cellular interactions for some of the mutants studied earlier. The molecular etiology of GNAO1 encephalopathies needs further elucidation as a prerequisite for the development of efficient therapeutic approaches. In this work, we combine clinical and medical genetics analysis of a novel GNAO1 mutation with an in-depth molecular dissection of the resultant protein variant. We identify two unrelated patients from Norway and France with a previously unknown mutation in GNAO1, c.509C>G that results in the production of the Pro170Arg mutant Gαo, leading to severe developmental and epileptic encephalopathy. Molecular investigations of Pro170Arg identify this mutant as a unique representative of the pathogenic variants. Its 100-fold-accelerated GTP uptake is not accompanied by a loss in GTP hydrolysis; Zn2+ ions induce a previously unseen effect on the mutant, forcing it to lose the bound GTP. Our work combining clinical and molecular analyses discovers a novel, biochemically distinct pathogenic missense variant of GNAO1 laying the ground for personalized treatment development.
Collapse
Affiliation(s)
- Yonika A. Larasati
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
| | - Gonzalo P. Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
| | - Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
| | - Silja T. Griffiths
- Department of Pediatrics, Haukeland University Hospital, 5009 Bergen, Norway
| | - Ragnhild Berentsen
- Department of Medical Genetics, Haukeland University Hospital, 5009 Bergen, Norway; (R.B.)
| | - Ingvild Aukrust
- Department of Medical Genetics, Haukeland University Hospital, 5009 Bergen, Norway; (R.B.)
- Department of Clinical Science, University of Bergen, 5008 Bergen, Norway
| | - Gaetan Lesca
- Department of Medical Genetics, University Hospital of Lyon, 69002 Lyon, France; (G.L.); (N.C.)
| | - Nicolas Chatron
- Department of Medical Genetics, University Hospital of Lyon, 69002 Lyon, France; (G.L.); (N.C.)
| | - Dorothée Ville
- Pediatric Neurology Department, University Hospital of Lyon, 69002 Lyon, France;
| | - Christian M. Korff
- Pediatric Neurology Unit, University Hospitals of Geneva, CH-1211 Geneva, Switzerland;
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690090, Russia
| |
Collapse
|
48
|
Scott-Dennis M, Rafani FA, Yi Y, Perera T, Harwood CR, Guba W, Rufer AC, Grether U, Veprintsev DB, Sykes DA. Development of a membrane-based Gi-CASE biosensor assay for profiling compounds at cannabinoid receptors. Front Pharmacol 2023; 14:1158091. [PMID: 37637423 PMCID: PMC10450933 DOI: 10.3389/fphar.2023.1158091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/05/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction: The cannabinoid receptor (CBR) subtypes 1 (CB1R) and 2 (CB2R) are key components of the endocannabinoid system (ECS), playing a central role in the control of peripheral pain, inflammation and the immune response, with further roles in the endocrine regulation of food intake and energy balance. So far, few medicines targeting these receptors have reached the clinic, suggesting that a better understanding of the receptor signalling properties of existing tool compounds and clinical candidates may open the door to the development of more effective and safer treatments. Both CB1R and CB2R are Gαi protein-coupled receptors but detecting Gαi protein signalling activity reliably and reproducibly is challenging. This is due to the inherent variability in live cell-based assays and restrictions around the use of radioactive [35S]-GTPγS, a favoured technology for developing higher-throughput membrane-based Gαi protein activity assays. Methods: Here, we describe the development of a membrane-based Gαi signalling system, produced from membrane preparations of HEK293TR cells, stably overexpressing CB1R or CB2R, and components of the Gαi-CASE biosensor. This BRET-based system allows direct detection of Gαi signalling in both cells and membranes by monitoring bioluminescence resonance energy transfer (BRET) between the α and the βγ subunits. Cells and membranes were subject to increasing concentrations of reference cannabinoid compounds, with 10 μM furimazine added to generate RET signals, which were detected on a PHERAstar FSX plate reader, then processed using MARS software and analysed in GraphPad PRISM 9.2. Results: In membranes expressing the Gi-CASE biosensor, the cannabinoid ligands profiled were found to show agonist and inverse agonist activity. Agonist activity elicited a decrease in the BRET signal, indicative of receptor activation and G protein dissociation. Inverse agonist activity caused an increase in BRET signal, indicative of receptor inactivation, and the accumulation of inactive G protein. Our membrane-based Gi-CASE NanoBRET system successfully characterised the potency (pEC50) and efficacy (Emax) of CBR agonists and inverse agonists in a 384-well screening format. Values obtained were in-line with whole-cell Gi-CASE assays and consistent with literature values obtained in the GTPγS screening format. Discussion: This novel, membrane-based Gαi protein activation assay is applicable to other Gαi-coupled GPCRs, including orphan receptors, allowing real-time higher-throughput measurements of receptor activation.
Collapse
Affiliation(s)
- Morgan Scott-Dennis
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
| | - Fikri A. Rafani
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
| | - Yicheng Yi
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
| | - Themiya Perera
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
| | - Clare R. Harwood
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
| | - Wolfgang Guba
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Arne C. Rufer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Dmitry B. Veprintsev
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
- Z7 Biotech Limited, London, United Kingdom
| | - David A. Sykes
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Midlands, United Kingdom
- Z7 Biotech Limited, London, United Kingdom
| |
Collapse
|
49
|
Grätz L, Kowalski-Jahn M, Scharf MM, Kozielewicz P, Jahn M, Bous J, Lambert NA, Gloriam DE, Schulte G. Pathway selectivity in Frizzleds is achieved by conserved micro-switches defining pathway-determining, active conformations. Nat Commun 2023; 14:4573. [PMID: 37516754 PMCID: PMC10387068 DOI: 10.1038/s41467-023-40213-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 07/12/2023] [Indexed: 07/31/2023] Open
Abstract
The class Frizzled of G protein-coupled receptors (GPCRs), consisting of ten Frizzled (FZD1-10) paralogs and Smoothened, remains one of the most enigmatic GPCR families. This class mediates signaling predominantly through Disheveled (DVL) or heterotrimeric G proteins. However, the mechanisms underlying pathway selection are elusive. Here we employ a structure-driven mutagenesis approach in combination with an extensive panel of functional signaling readouts to investigate the importance of conserved state-stabilizing residues in FZD5 for signal specification. Similar data were obtained for FZD4 and FZD10 suggesting that our findings can be extrapolated to other members of the FZD family. Comparative molecular dynamics simulations of wild type and selected FZD5 mutants further support the concept that distinct conformational changes in FZDs specify the signal outcome. In conclusion, we find that FZD5 and FZDs in general prefer coupling to DVL rather than heterotrimeric G proteins and that distinct active state micro-switches in the receptor are essential for pathway selection arguing for conformational changes in the receptor protein defining transducer selectivity.
Collapse
Affiliation(s)
- Lukas Grätz
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Biomedicum, S-17165, Stockholm, Sweden
| | - Maria Kowalski-Jahn
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Biomedicum, S-17165, Stockholm, Sweden
| | - Magdalena M Scharf
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Biomedicum, S-17165, Stockholm, Sweden
| | - Pawel Kozielewicz
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Biomedicum, S-17165, Stockholm, Sweden
| | - Michael Jahn
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, KTH - Royal Institute of Technology, S-17121, Solna, Sweden
- Max Planck Unit for the Science of Pathogens, Bioinformatics platform, Charitéplatz 1, D-10117, Berlin, Germany
| | - Julien Bous
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Biomedicum, S-17165, Stockholm, Sweden
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Gunnar Schulte
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Biomedicum, S-17165, Stockholm, Sweden.
| |
Collapse
|
50
|
Boon K, Vanalken N, Meyen E, Schols D, Van Loy T. REGA-SIGN: Development of a Novel Set of NanoBRET-Based G Protein Biosensors. BIOSENSORS 2023; 13:767. [PMID: 37622853 PMCID: PMC10452170 DOI: 10.3390/bios13080767] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023]
Abstract
Despite G protein-coupled receptors (GPCRs) being important theapeutic targets, the signaling properties of many GPCRs remain poorly characterized. GPCR activation primarily initiates heterotrimeric G protein signaling. To detect ligand-induced G protein activation, Bioluminescence Resonance Energy Transfer (BRET)-based biosensors were previously developed. Here, we designed a novel set of Nanoluciferase (NLuc) BRET-based biosensors (REGA-SIGN) that covers all Gα protein families (i.e., Gαi/o, GαSs/L, Gα12/13 and Gαq/15). REGA-SIGN uses NLuc as a bioluminescent donor and LSS-mKATE2, a red-shifted fluorophore, as an acceptor. Due to the enhanced spectral separation between donor and acceptor emission and the availability of a stable substrate for NLuc, this donor-acceptor pair enables sensitive kinetic assessment of G protein activity. After optimization, the NLuc integration sites into the Gα subunit largely corresponded with previously reported integration sites, except for GαSs/L for which we describe an alternative NLuc insertion site. G protein rescue experiments validated the biological activity of these Gα donor proteins. Direct comparison between EGFP and LSS-mKATE2 as acceptor fluorophores revealed improved sensitivity for nearly all G protein subtypes when using the latter one. Hence, REGA-SIGN can be used as a panel of kinetic G protein biosensors with high sensitivity.
Collapse
Affiliation(s)
| | | | | | | | - Tom Van Loy
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49, P.O. Box 1030, 3000 Leuven, Belgium; (K.B.); (N.V.); (E.M.); (D.S.)
| |
Collapse
|