1
|
Yu S, You Y, Liu L, Cai X, Huang C. Modulation of biomaterial-induced foreign body response by regulating the differentiation and migration of Treg cells through the CXCL12-CXCR4/7 axis. Biomater Sci 2025; 13:1529-1542. [PMID: 39932368 DOI: 10.1039/d4bm01474j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Tissue exposure to implanted biomaterials triggers a foreign body response (FBR), which is a stepwise immunological process involving innate immune cells and tissue repair cells. Although the regulatory T (Treg) cells play a crucial role in inflammation and tissue repair, their function in the process of FBR has not been well investigated. In this study, as titanium (Ti) exhibits better biocompatibility and induces milder FBR than polymethyl methacrylate (PMMA), we analyzed the characteristics of Treg cells during FBR caused by the two types of biomaterials. In a rat femur implantation model, we found that the number of Treg cells around titanium implants was much more than that in the PMMA-implanted group. Meanwhile, the expression of CXCR4 in tissues around Ti implants was significantly higher, and the expression of CXCR7 was lower. When co-cultured with biomaterials and macrophages, the differentiation and migration of Treg cells in the Ti-implanted group were promoted, and this effect could be modulated by CXCR4/7 inhibitors. Moreover, targeting CXCR4/7 influenced the amount of Treg cells in vivo and then reversed the FBR induced by PMMA or Ti implants. In summary, our findings revealed the role of CXCR4/CXCR7 in regulating the migration and differentiation of Treg cells during FBR and suggested that the CXCL12-CXCR4/CXCR7 axis may serve as a potential therapeutic target for immunomodulating foreign body response.
Collapse
Affiliation(s)
- Siyi Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Yuan You
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Lan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Xinjie Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Ma Y, Patterson B, Zhu L. Biased signaling in GPCRs: Structural insights and implications for drug development. Pharmacol Ther 2025; 266:108786. [PMID: 39719175 DOI: 10.1016/j.pharmthera.2024.108786] [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: 09/03/2024] [Revised: 11/18/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors in humans, playing a crucial role in regulating diverse cellular processes and serving as primary drug targets. Traditional drug design has primarily focused on ligands that uniformly activate or inhibit GPCRs. However, the concept of biased agonism-where ligands selectively stabilize distinct receptor conformations, leading to unique signaling outcomes-has introduced a paradigm shift in therapeutic development. Despite the promise of biased agonists to enhance drug efficacy and minimize side effects, a comprehensive understanding of the structural and biophysical mechanisms underlying biased signaling is essential. Recent advancements in GPCR structural biology have provided unprecedented insights into ligand binding, conformational dynamics, and the molecular basis of biased signaling. These insights, combined with improved techniques for characterizing ligand efficacy, have driven the development of biased ligands for several GPCRs, including opioid, angiotensin, and adrenergic receptors. This review synthesizes these developments, from mechanisms to drug discovery in biased signaling, emphasizing the role of structural insights in the rational design of next-generation biased agonists with superior therapeutic profiles. Ultimately, these advances hold the potential to revolutionize GPCR-targeted drug discovery, paving the way for more precise and effective treatments.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Brandon Patterson
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Lan Zhu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States.
| |
Collapse
|
3
|
Karl K, Del Piccolo N, Light T, Roy T, Dudeja P, Ursachi VC, Fafilek B, Krejci P, Hristova K. Ligand bias underlies differential signaling of multiple FGFs via FGFR1. eLife 2024; 12:RP88144. [PMID: 38568193 PMCID: PMC10990489 DOI: 10.7554/elife.88144] [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: 04/05/2024] Open
Abstract
The differential signaling of multiple FGF ligands through a single fibroblast growth factor (FGF) receptor (FGFR) plays an important role in embryonic development. Here, we use quantitative biophysical tools to uncover the mechanism behind differences in FGFR1c signaling in response to FGF4, FGF8, and FGF9, a process which is relevant for limb bud outgrowth. We find that FGF8 preferentially induces FRS2 phosphorylation and extracellular matrix loss, while FGF4 and FGF9 preferentially induce FGFR1c phosphorylation and cell growth arrest. Thus, we demonstrate that FGF8 is a biased FGFR1c ligand, as compared to FGF4 and FGF9. Förster resonance energy transfer experiments reveal a correlation between biased signaling and the conformation of the FGFR1c transmembrane domain dimer. Our findings expand the mechanistic understanding of FGF signaling during development and bring the poorly understood concept of receptor tyrosine kinase ligand bias into the spotlight.
Collapse
Affiliation(s)
- Kelly Karl
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins UniversityBaltimoreUnited States
| | - Nuala Del Piccolo
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins UniversityBaltimoreUnited States
| | - Taylor Light
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins UniversityBaltimoreUnited States
| | - Tanaya Roy
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins UniversityBaltimoreUnited States
| | - Pooja Dudeja
- Department of Biology, Faculty of Medicine, Masaryk UniversityBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Vlad-Constantin Ursachi
- Department of Biology, Faculty of Medicine, Masaryk UniversityBrnoCzech Republic
- International Clinical Research Center, St. Anne's University HospitalBrnoCzech Republic
| | - Bohumil Fafilek
- Department of Biology, Faculty of Medicine, Masaryk UniversityBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
- International Clinical Research Center, St. Anne's University HospitalBrnoCzech Republic
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk UniversityBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
- International Clinical Research Center, St. Anne's University HospitalBrnoCzech Republic
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
4
|
Karl K, Rajagopal S, Hristova K. Quantitative assessment of ligand bias from bias plots: The bias coefficient "kappa". Biochim Biophys Acta Gen Subj 2023; 1867:130428. [PMID: 37488010 PMCID: PMC10528940 DOI: 10.1016/j.bbagen.2023.130428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/15/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
The current methods for quantifying ligand bias involve the construction of bias plots and the calculations of bias coefficients that can be compared using statistical methods. However, widely used bias coefficients can diverge in their abilities to identify ligand bias and can give false positives. As the empirical bias plots are considered the most reliable tools in bias identification, here we develop an analytical description of bias plot trajectories and introduce a bias coefficient, kappa, which is calculated from these trajectories. The new bias coefficient complements the tool-set in ligand bias identification in cell signaling research.
Collapse
Affiliation(s)
- Kelly Karl
- Institute for NanoBioTechnology, Department of Materials Science and Engineering, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218, United States of America
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States of America
| | - Kalina Hristova
- Institute for NanoBioTechnology, Department of Materials Science and Engineering, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218, United States of America.
| |
Collapse
|
5
|
Intranuovo F, Brunetti L, DelRe P, Mangiatordi GF, Stefanachi A, Laghezza A, Niso M, Leonetti F, Loiodice F, Ligresti A, Kostrzewa M, Brea J, Loza MI, Sotelo E, Saviano M, Colabufo NA, Riganti C, Abate C, Contino M. Development of N-(1-Adamantyl)benzamides as Novel Anti-Inflammatory Multitarget Agents Acting as Dual Modulators of the Cannabinoid CB2 Receptor and Fatty Acid Amide Hydrolase. J Med Chem 2023; 66:235-250. [PMID: 36542836 DOI: 10.1021/acs.jmedchem.2c01084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cannabinoid type 2 receptor (CB2R), belonging to the endocannabinoid system, is overexpressed in pathologies characterized by inflammation, and its activation counteracts inflammatory states. Fatty acid amide hydrolase (FAAH) is an enzyme responsible for the degradation of the main endocannabinoid anandamide; thus, the simultaneous CB2R activation and FAAH inhibition may be a synergistic anti-inflammatory strategy. Encouraged by principal component analysis (PCA) data identifying a wide chemical space shared by CB2R and FAAH ligands, we designed a small library of adamantyl-benzamides, as potential dual agents, CB2R agonists, and FAAH inhibitors. The new compounds were tested for their CB2R affinity/selectivity and CB2R and FAAH activity. Derivatives 13, 26, and 27, displaying the best pharmacodynamic profile as CB2R full agonists and FAAH inhibitors, decreased pro-inflammatory and increased anti-inflammatory cytokines production. Molecular docking simulations complemented the experimental findings by providing a molecular rationale behind the observed activities. These multitarget ligands constitute promising anti-inflammatory agents.
Collapse
Affiliation(s)
- Francesca Intranuovo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Leonardo Brunetti
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Pietro DelRe
- Institute of Crystallography, National Research Council of Italy, Via Amendola, 122/o, Bari 70126, Italy
| | | | - Angela Stefanachi
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Antonio Laghezza
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Alessia Ligresti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, Pozzuoli 80078, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, Pozzuoli 80078, Italy
| | - Jose Brea
- Innopharma Screening Platform, BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology. School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Maria Isabel Loza
- Innopharma Screening Platform, BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology. School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Eddy Sotelo
- ComBioMed Research Group, Centro de Química Biológica y Materiales Moleculares (CIQUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Michele Saviano
- Institute of Crystallography, National Research Council of Italy, Via Vivaldi, 43, Caserta 81100, Italy
| | - Nicola Antonio Colabufo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università Degli Studi di Torino, Torino 10126, Italy
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy.,Institute of Crystallography, National Research Council of Italy, Via Amendola, 122/o, Bari 70126, Italy
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| |
Collapse
|
6
|
McDonald JK, van der Westhuizen ET, Pham V, Thompson G, Felder CC, Paul SM, Thal DM, Christopoulos A, Valant C. Biased Profile of Xanomeline at the Recombinant Human M 4 Muscarinic Acetylcholine Receptor. ACS Chem Neurosci 2022; 13:1206-1218. [PMID: 35380782 DOI: 10.1021/acschemneuro.1c00827] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Many Food and Drug Administration (FDA)-approved drugs are structural analogues of the endogenous (natural) ligands of G protein-coupled receptors (GPCRs). However, it is becoming appreciated that chemically distinct ligands can bind to GPCRs in conformations that lead to different cellular signaling events, a phenomenon termed biased agonism. Despite this, the rigorous experimentation and analysis required to identify biased agonism are often not undertaken in most clinical candidates and go unrealized. Recently, xanomeline, a muscarinic acetylcholine receptor (mAChR) agonist, has entered phase III clinical trials for the treatment of schizophrenia. If successful, xanomeline will be the first novel FDA-approved antipsychotic drug in almost 50 years. Intriguingly, xanomeline's potential for biased agonism at the mAChRs and, in particular, the M4 mAChR, the most promising receptor target for schizophrenia, has not been assessed. Here, we quantify the biased agonism profile of xanomeline and three other mAChR agonists in Chinese hamster ovary cells recombinantly expressing the M4 mAChR. Agonist activity was examined across nine distinct signaling readouts, including the activation of five different G protein subtypes, ERK1/2 phosphorylation, β-arrestin recruitment, calcium mobilization, and cAMP regulation. Relative to acetylcholine (ACh), xanomeline was biased away from ERK1/2 phosphorylation and calcium mobilization compared to Gαi2 protein activation. These findings likely have important implications for our understanding of the therapeutic action of xanomeline and call for further investigation into the in vivo consequences of biased agonism in drugs targeting the M4 mAChR for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Jack K. McDonald
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Emma T. van der Westhuizen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Vi Pham
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Geoff Thompson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - Steven M. Paul
- Karuna Therapeutics, Boston, Massachusetts 02110, United States
| | - David M. Thal
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
7
|
Mao Q, Zhang B, Tian S, Qin W, Chen J, Huang XP, Xin Y, Yang H, Zhen XC, Shui W, Ye N. Structural optimizations and bioevaluation of N-H aporphine analogues as Gq-biased and selective serotonin 5-HT2C receptor agonists. Bioorg Chem 2022; 123:105795. [DOI: 10.1016/j.bioorg.2022.105795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
|
8
|
Abi-Dargham A, Javitch JA, Slifstein M, Anticevic A, Calkins ME, Cho YT, Fonteneau C, Gil R, Girgis R, Gur RE, Gur RC, Grinband J, Kantrowitz J, Kohler C, Krystal J, Murray J, Ranganathan M, Santamauro N, Van Snellenberg J, Tamayo Z, Wolf D, Gray D, Lieberman J. Dopamine D1R Receptor Stimulation as a Mechanistic Pro-cognitive Target for Schizophrenia. Schizophr Bull 2021; 48:199-210. [PMID: 34423843 PMCID: PMC8781338 DOI: 10.1093/schbul/sbab095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Decades of research have highlighted the importance of optimal stimulation of cortical dopaminergic receptors, particularly the D1R receptor (D1R), for prefrontal-mediated cognition. This mechanism is particularly relevant to the cognitive deficits in schizophrenia, given the abnormalities in cortical dopamine (DA) neurotransmission and in the expression of D1R. Despite the critical need for D1R-based therapeutics, many factors have complicated their development and prevented this important therapeutic target from being adequately interrogated. Challenges include determination of the optimal level of D1R stimulation needed to improve cognitive performance, especially when D1R expression levels, affinity states, DA levels, and the resulting D1R occupancy by DA, are not clearly known in schizophrenia, and may display great interindividual and intraindividual variability related to cognitive states and other physiological variables. These directly affect the selection of the level of stimulation necessary to correct the underlying neurobiology. The optimal mechanism for stimulation is also unknown and could include partial or full agonism, biased agonism, or positive allosteric modulation. Furthermore, the development of D1R targeting drugs has been complicated by complexities in extrapolating from in vitro affinity determinations to in vivo use. Prior D1R-targeted drugs have been unsuccessful due to poor bioavailability, pharmacokinetics, and insufficient target engagement at tolerable doses. Newer drugs have recently become available, and these must be tested in the context of carefully designed paradigms that address methodological challenges. In this paper, we discuss how a better understanding of these challenges has shaped our proposed experimental design for testing a new D1R/D5R partial agonist, PF-06412562, renamed CVL-562.
Collapse
Affiliation(s)
- Anissa Abi-Dargham
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA,Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA,Department of Psychiatry, Yale University, New Haven, CT, USA,Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Cerevel Therapeutics Research and Development, Boston, MA, USA,To whom correspondence should be addressed; Tel: +(631) 885-0814; e-mail:
| | - Jonathan A Javitch
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Mark Slifstein
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA
| | - Alan Anticevic
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Monica E Calkins
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Youngsun T Cho
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Clara Fonteneau
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Roberto Gil
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA
| | - Ragy Girgis
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Raquel E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben C Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack Grinband
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Joshua Kantrowitz
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Christian Kohler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Krystal
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - John Murray
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | | | | | - Jared Van Snellenberg
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA
| | - Zailyn Tamayo
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Daniel Wolf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - David Gray
- Cerevel Therapeutics Research and Development, Boston, MA, USA
| | - Jeffrey Lieberman
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| |
Collapse
|
9
|
Mallo-Abreu A, Reyes-Resina I, Azuaje J, Franco R, García-Rey A, Majellaro M, Miranda-Pastoriza D, García-Mera X, Jespers W, Gutiérrez-de-Terán H, Navarro G, Sotelo E. Potent and Subtype-Selective Dopamine D 2 Receptor Biased Partial Agonists Discovered via an Ugi-Based Approach. J Med Chem 2021; 64:8710-8726. [PMID: 34110150 PMCID: PMC8552448 DOI: 10.1021/acs.jmedchem.1c00704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Using
a previously unexplored, efficient, and versatile multicomponent
method, we herein report the rapid generation of novel potent and
subtype-selective DRD2 biased partial agonists. This strategy
exemplifies the search for diverse and previously unexplored moieties
for the secondary/allosteric pharmacophore of the common phenyl-piperazine
scaffold. The pharmacological characterization of the new compound
series led to the identification of several ligands with excellent
DRD2 affinity and subtype selectivity and remarkable functional
selectivity for either the cAMP (22a and 24d) or the β-arrestin (27a and 29c)
signaling pathways. These results were further interpreted on the
basis of molecular models of these ligands in complex with the recent
DRD2 crystal structures, highlighting the critical role
of the secondary/allosteric pharmacophore in modulating the functional
selectivity profile.
Collapse
Affiliation(s)
- Ana Mallo-Abreu
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Jhonny Azuaje
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Rafael Franco
- Faculty of Chemistry, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Aitor García-Rey
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria Majellaro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Darío Miranda-Pastoriza
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Xerardo García-Mera
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Willem Jespers
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE-75124, Sweden
| | | | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
10
|
The biophysical basis of receptor tyrosine kinase ligand functional selectivity: Trk-B case study. Biochem J 2021; 477:4515-4526. [PMID: 33094812 DOI: 10.1042/bcj20200671] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/08/2023]
Abstract
Tropomyosin receptor kinase B (Trk-B) belongs to the second largest family of membrane receptors, Receptor Tyrosine Kinases (RTKs). Trk-B is known to interact with three different neurotrophins: Brain-Derived Neurotrophic Factor (BDNF), Neurotrophin-4 (NT-4), and Neurotrophin-3 (NT-3). All three neurotrophins are involved in survival and proliferation of neuronal cells, but each induces distinct signaling through Trk-B. We hypothesize that the different biological effects correlate with differences in the interactions between the Trk-B receptors, when bound to different ligands, in the plasma membrane. To test this hypothesis, we use quantitative FRET to characterize Trk-B dimerization in response to NT-3 and NT-4 in live cells, and compare it to the previously published data for Trk-B in the absence and presence of BDNF. Our study reveals that the distinct Trk-B signaling outcomes are underpinned by both different configurations and different stabilities of the three ligand-bound Trk-B dimers in the plasma membrane.
Collapse
|
11
|
McNeill SM, Baltos JA, White PJ, May LT. Biased agonism at adenosine receptors. Cell Signal 2021; 82:109954. [PMID: 33610717 DOI: 10.1016/j.cellsig.2021.109954] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 01/14/2023]
Abstract
Adenosine modulates many aspects of human physiology and pathophysiology through binding to the adenosine family of G protein-coupled receptors, which are comprised of four subtypes, the A1R, A2AR, A2BR and A3R. Modulation of adenosine receptor function by exogenous agonists, antagonists and allosteric modulators can be beneficial for a number of conditions including cardiovascular disease, Parkinson's disease, and cancer. Unfortunately, many preclinical drug candidates targeting adenosine receptors have failed in clinical trials due to limited efficacy and/or severe on-target undesired effects. To overcome the key barriers typically encountered when transitioning adenosine receptor ligands into the clinic, research efforts have focussed on exploiting the phenomenon of biased agonism. Biased agonism provides the opportunity to develop ligands that favour therapeutic signalling pathways, whilst avoiding signalling associated with on-target undesired effects. Recent studies have begun to define the structure-function relationships that underpin adenosine receptor biased agonism and establish how this phenomenon can be harnessed therapeutically. In this review we describe the recent advancements made towards achieving therapeutically relevant biased agonism at adenosine receptors.
Collapse
Affiliation(s)
- Samantha M McNeill
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia; Department of Pharmacology, Monash University, Melbourne, VIC, Australia.
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia; Department of Pharmacology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
12
|
Sasaki T, Watanabe Y, Kuboyama A, Oikawa A, Shimizu M, Yamauchi Y, Sato R. Muscle-specific TGR5 overexpression improves glucose clearance in glucose-intolerant mice. J Biol Chem 2021; 296:100131. [PMID: 33262218 PMCID: PMC7949087 DOI: 10.1074/jbc.ra120.016203] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/09/2020] [Accepted: 12/01/2020] [Indexed: 01/05/2023] Open
Abstract
TGR5, a G protein-coupled bile acid receptor, is expressed in various tissues and regulates several physiological processes. In the skeletal muscle, TGR5 activation is known to induce muscle hypertrophy; however, the effects on glucose and lipid metabolism are not well understood, despite the fact that the skeletal muscle plays a major role in energy metabolism. Here, we demonstrate that skeletal muscle-specific TGR5 transgenic (Tg) mice exhibit increased glucose utilization, without altering the expression of major genes related to glucose and lipid metabolism. Metabolite profiling analysis by capillary electrophoresis time-of-flight mass spectrometry showed that glycolytic flux was activated in the skeletal muscle of Tg mice, leading to an increase in glucose utilization. Upon long-term, high-fat diet challenge, blood glucose clearance was improved in Tg mice without an accompanying increase in insulin sensitivity in skeletal muscle and a reduction of body weight. Moreover, Tg mice showed improved age-associated glucose intolerance. These results strongly suggest that TGR5 ameliorated glucose metabolism disorder that is caused by diet-induced obesity and aging by enhancing the glucose metabolic capacity of the skeletal muscle. Our study demonstrates that TGR5 activation in the skeletal muscle is effective in improving glucose metabolism and may be beneficial in developing a novel strategy for the prevention or treatment of hyperglycemia.
Collapse
Affiliation(s)
- Takashi Sasaki
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan.
| | - Yuichi Watanabe
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Ayane Kuboyama
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Akira Oikawa
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa, Japan; Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata, Japan
| | - Makoto Shimizu
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Yoshio Yamauchi
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan; Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
13
|
Karl K, Paul MD, Pasquale EB, Hristova K. Ligand bias in receptor tyrosine kinase signaling. J Biol Chem 2020; 295:18494-18507. [PMID: 33122191 PMCID: PMC7939482 DOI: 10.1074/jbc.rev120.015190] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Ligand bias is the ability of ligands to differentially activate certain receptor signaling responses compared with others. It reflects differences in the responses of a receptor to specific ligands and has implications for the development of highly specific therapeutics. Whereas ligand bias has been studied primarily for G protein-coupled receptors (GPCRs), there are also reports of ligand bias for receptor tyrosine kinases (RTKs). However, the understanding of RTK ligand bias is lagging behind the knowledge of GPCR ligand bias. In this review, we highlight how protocols that were developed to study GPCR signaling can be used to identify and quantify RTK ligand bias. We also introduce an operational model that can provide insights into the biophysical basis of RTK activation and ligand bias. Finally, we discuss possible mechanisms underpinning RTK ligand bias. Thus, this review serves as a primer for researchers interested in investigating ligand bias in RTK signaling.
Collapse
Affiliation(s)
- Kelly Karl
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael D Paul
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
14
|
Ahmed F, Zapata-Mercado E, Rahman S, Hristova K. The Biased Ligands NGF and NT-3 Differentially Stabilize Trk-A Dimers. Biophys J 2020; 120:55-63. [PMID: 33285113 DOI: 10.1016/j.bpj.2020.11.2262] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/14/2020] [Accepted: 11/23/2020] [Indexed: 12/23/2022] Open
Abstract
Trk-A is a receptor tyrosine kinase (RTK) that plays an essential role in the development and functioning of the nervous system. Trk-A is expressed in neurons and signals in response to two ligands, NGF and neurotrophin-3 (NT-3), with very different functional consequences. Thus, NGF and NT-3 are "biased" ligands for Trk-A. Because it has been hypothesized that biased RTK ligands induce differential stabilization of RTK dimers, here, we seek to test this hypothesis for NGF and NT-3. In particular, we use Förster resonance energy transfer (FRET) and fluorescence intensity fluctuation spectroscopy to assess the strength of Trk-A interactions and Trk-A oligomer size in the presence of the two ligands. Although the difference in Trk-A behavior in response to the two ligands has been previously attributed to differences in their binding to Trk-A in the endosomes at low pH, here, we further show differences in the stabilities of the NGF- and NT-3-bound Trk-A dimers in the plasma membrane and at neutral pH. We discuss the biological significance of these new findings and their implications for the design of Trk-A ligands with novel functionalities.
Collapse
Affiliation(s)
- Fozia Ahmed
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Elmer Zapata-Mercado
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Sanim Rahman
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland; Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
15
|
Hernandez-Olmos V, Heering J, Planz V, Liu T, Kaps A, Rajkumar R, Gramzow M, Kaiser A, Schubert-Zsilavecz M, Parnham MJ, Windbergs M, Steinhilber D, Proschak E. First Structure-Activity Relationship Study of Potent BLT2 Agonists as Potential Wound-Healing Promoters. J Med Chem 2020; 63:11548-11572. [PMID: 32946232 DOI: 10.1021/acs.jmedchem.0c00588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The first potent leukotriene B4 (LTB4) receptor type 2 (BLT2) agonists, endogenous 12(S)-hydroxyheptadeca-5Z,8E,10E-trienoic acid (12-HHT), and synthetic CAY10583 (CAY) have been recently described to accelerate wound healing by enhanced keratinocyte migration and indirect stimulation of fibroblast activity in diabetic rats. CAY represents a very valuable starting point for the development of novel wound-healing promoters. In this work, the first structure-activity relationship study for CAY scaffold-based BLT2 agonists is presented. The newly prepared derivatives showed promising in vitro wound-healing activity.
Collapse
Affiliation(s)
- Victor Hernandez-Olmos
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Jan Heering
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Viktoria Planz
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Ting Liu
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.,Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Alexander Kaps
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.,Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Rinusha Rajkumar
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.,Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Matthias Gramzow
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.,Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Astrid Kaiser
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Manfred Schubert-Zsilavecz
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Michael J Parnham
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Dieter Steinhilber
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.,Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.,Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| |
Collapse
|
16
|
Zhang B, Zhao S, Yang D, Wu Y, Xin Y, Cao H, Huang XP, Cai X, Sun W, Ye N, Xu Y, Peng Y, Zhao S, Liu ZJ, Zhong G, Wang MW, Shui W. A Novel G Protein-Biased and Subtype-Selective Agonist for a G Protein-Coupled Receptor Discovered from Screening Herbal Extracts. ACS CENTRAL SCIENCE 2020; 6:213-225. [PMID: 32123739 PMCID: PMC7047268 DOI: 10.1021/acscentsci.9b01125] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 05/14/2023]
Abstract
Subtype selectivity and functional bias are vital in current drug discovery for G protein-coupled receptors (GPCRs) as selective and biased ligands are expected to yield drug leads with optimal on-target benefits and minimal side-effects. However, structure-based design and medicinal chemistry exploration remain challenging in part because of highly conserved binding pockets within subfamilies. Herein, we present an affinity mass spectrometry approach for screening herbal extracts to identify active ligands of a GPCR, the 5-HT2C receptor. Using this method, we discovered a naturally occurring aporphine 1857 that displayed strong selectivity for activating 5-HT2C without activating the 5-HT2A or 5-HT2B receptors. Remarkably, this novel ligand exhibited exclusive bias toward G protein signaling for which key residues were identified, and it showed comparable in vivo efficacy for food intake suppression and weight loss as the antiobesity drug, lorcaserin. Our study establishes an efficient approach to discovering novel GPCR ligands by exploring the largely untapped chemical space of natural products.
Collapse
Affiliation(s)
- Bingjie Zhang
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Simeng Zhao
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Dehua Yang
- The
National Center for Drug Screening and the CAS Key Laboratory of Receptor
Research, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yiran Wu
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Ye Xin
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Haijie Cao
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Xi-Ping Huang
- Department
of Pharmacology, NIMH Psychoactive Drug Screening Program, School
of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Xiaoqing Cai
- The
National Center for Drug Screening and the CAS Key Laboratory of Receptor
Research, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen Sun
- The
National Center for Drug Screening and the CAS Key Laboratory of Receptor
Research, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Na Ye
- Jiangsu
Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical
Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yueming Xu
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Yao Peng
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Suwen Zhao
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of
Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Zhi-Jie Liu
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of
Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Guisheng Zhong
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of
Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
- E-mail:
| | - Ming-Wei Wang
- The
National Center for Drug Screening and the CAS Key Laboratory of Receptor
Research, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School
of
Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
- School
of Pharmacy, Fudan University, Shanghai 201203, China
- E-mail:
| | - Wenqing Shui
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of
Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
- E-mail:
| |
Collapse
|
17
|
Lamichhane R, Liu JJ, White KL, Katritch V, Stevens RC, Wüthrich K, Millar DP. Biased Signaling of the G-Protein-Coupled Receptor β 2AR Is Governed by Conformational Exchange Kinetics. Structure 2020; 28:371-377.e3. [PMID: 31978323 DOI: 10.1016/j.str.2020.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/11/2019] [Accepted: 01/03/2020] [Indexed: 01/14/2023]
Abstract
G-protein-coupled receptors (GPCRs) mediate a wide range of human physiological functions by transducing extracellular ligand binding events into intracellular responses. GPCRs can activate parallel, independent signaling pathways mediated by G proteins or β-arrestins. Whereas "balanced" agonists activate both pathways equally, "biased" agonists dominantly activate one pathway, which is of interest for designing GPCR-targeting drugs because it may mitigate undesirable side effects. Previous studies demonstrated that β-arrestin activation is associated with transmembrane helix VII (TM VII) of GPCRs. Here, single-molecule fluorescence spectroscopy with the β2-adrenergic receptor (β2AR) in the ligand-free state showed that TM VII spontaneously fluctuates between one inactive and one active-like conformation. The presence of the β-arrestin-biased agonist isoetharine prolongs the dwell time of TM VII in the active-like conformation compared with the balanced agonist formoterol, suggesting that ligands can induce signaling bias by modulating the kinetics of receptor conformational exchange.
Collapse
Affiliation(s)
- Rajan Lamichhane
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jeffrey J Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Kate L White
- Departments of Biological Sciences and Chemistry, Bridge Institute, USC Michelson Center for Convergent Bioscience, University of Southern California, Childs Way, MC3502, Los Angeles, CA 90089, USA
| | - Vsevolod Katritch
- Departments of Biological Sciences and Chemistry, Bridge Institute, USC Michelson Center for Convergent Bioscience, University of Southern California, Childs Way, MC3502, Los Angeles, CA 90089, USA
| | - Raymond C Stevens
- Departments of Biological Sciences and Chemistry, Bridge Institute, USC Michelson Center for Convergent Bioscience, University of Southern California, Childs Way, MC3502, Los Angeles, CA 90089, USA
| | - Kurt Wüthrich
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David P Millar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
18
|
Falomir-Lockhart LJ, Cavazzutti GF, Giménez E, Toscani AM. Fatty Acid Signaling Mechanisms in Neural Cells: Fatty Acid Receptors. Front Cell Neurosci 2019; 13:162. [PMID: 31105530 PMCID: PMC6491900 DOI: 10.3389/fncel.2019.00162] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022] Open
Abstract
Fatty acids (FAs) are typically associated with structural and metabolic roles, as they can be stored as triglycerides, degraded by β-oxidation or used in phospholipids’ synthesis, the main components of biological membranes. It has been shown that these lipids exhibit also regulatory functions in different cell types. FAs can serve as secondary messengers, as well as modulators of enzymatic activities and substrates for cytokines synthesis. More recently, it has been documented a direct activity of free FAs as ligands of membrane, cytosolic, and nuclear receptors, and cumulative evidence has emerged, demonstrating its participation in a wide range of physiological and pathological conditions. It has been long known that the central nervous system is enriched with poly-unsaturated FAs, such as arachidonic (C20:4ω-6) or docosohexaenoic (C22:6ω-3) acids. These lipids participate in the regulation of membrane fluidity, axonal growth, development, memory, and inflammatory response. Furthermore, a whole family of low molecular weight compounds derived from FAs has also gained special attention as the natural ligands for cannabinoid receptors or key cytokines involved in inflammation, largely expanding the role of FAs as precursors of signaling molecules. Nutritional deficiencies, and alterations in lipid metabolism and lipid signaling have been associated with developmental and cognitive problems, as well as with neurodegenerative diseases. The molecular mechanism behind these effects still remains elusive. But in the last two decades, different families of proteins have been characterized as receptors mediating FAs signaling. This review focuses on different receptors sensing and transducing free FAs signals in neural cells: (1) membrane receptors of the family of G Protein Coupled Receptors known as Free Fatty Acid Receptors (FFARs); (2) cytosolic transport Fatty Acid-Binding Proteins (FABPs); and (3) transcription factors Peroxisome Proliferator-Activated Receptors (PPARs). We discuss how these proteins modulate and mediate direct regulatory functions of free FAs in neural cells. Finally, we briefly discuss the advantages of evaluating them as potential targets for drug design in order to manipulate lipid signaling. A thorough characterization of lipid receptors of the nervous system could provide a framework for a better understanding of their roles in neurophysiology and, potentially, help for the development of novel drugs against aging and neurodegenerative processes.
Collapse
Affiliation(s)
- Lisandro Jorge Falomir-Lockhart
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Gian Franco Cavazzutti
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Ezequiel Giménez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Andrés Martín Toscani
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| |
Collapse
|
19
|
Non-Nutritive Sweeteners and Their Implications on the Development of Metabolic Syndrome. Nutrients 2019; 11:nu11030644. [PMID: 30884834 PMCID: PMC6471792 DOI: 10.3390/nu11030644] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/18/2022] Open
Abstract
Individuals widely use non-nutritive sweeteners (NNS) in attempts to lower their overall daily caloric intake, lose weight, and sustain a healthy diet. There are insufficient scientific data that support the safety of consuming NNS. However, recent studies have suggested that NNS consumption can induce gut microbiota dysbiosis and promote glucose intolerance in healthy individuals that may result in the development of type 2 diabetes mellitus (T2DM). This sequence of events may result in changes in the gut microbiota composition through microRNA (miRNA)-mediated changes. The mechanism(s) by which miRNAs alter gene expression of different bacterial species provides a link between the consumption of NNS and the development of metabolic changes. Another potential mechanism that connects NNS to metabolic changes is the molecular crosstalk between the insulin receptor (IR) and G protein-coupled receptors (GPCRs). Here, we aim to highlight the role of NNS in obesity and discuss IR-GPCR crosstalk and miRNA-mediated changes, in the manipulation of the gut microbiota composition and T2DM pathogenesis.
Collapse
|
20
|
Littmann T, Buschauer A, Bernhardt G. Split luciferase-based assay for simultaneous analyses of the ligand concentration- and time-dependent recruitment of β-arrestin2. Anal Biochem 2019; 573:8-16. [PMID: 30853375 DOI: 10.1016/j.ab.2019.02.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022]
Abstract
Functional selectivity of agonists has gained increasing interest in G protein-coupled receptor (GPCR) research, e.g. due to expectations of drugs with reduced adverse effects. Different agonist-dependent GPCR conformations are conceived to selectively activate a balanced or imbalanced intracellular signalling response, involving e.g. different Gα subtypes, Gβγ-subunits and β-arrestins. To discriminate between the different signalling pathways (bias), sensitive techniques are needed that do not interfere with signalling. We applied split luciferase complementation to the GPCR/β-arrestin2 interaction and thoroughly analysed the influence of its implementation on intracellular signalling. This led to an assay enabling the functional characterization of ligands at the hH1R, the hM1,5R and the hNTS1R in live HEK293T cells. As demonstrated at the hM1,5R, the assay was sensitive enough to identify iperoxo as a superagonist. Time-dependent analyses of the recruitment of β-arrestin2 became possible, allowing the identification of class A and class B GPCRs, due to the differential duration of their interaction with β-arrestin2 and their recycling to the cell membrane. The developed β-arrestin2 recruitment assay, which provides concentration- and time-dependent information on the interaction between GPCRs and β-arrestin2 upon stimulation of the receptor, should be broadly applicable and of high value for the analysis of agonist bias.
Collapse
Affiliation(s)
- Timo Littmann
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany.
| | - Armin Buschauer
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany.
| |
Collapse
|
21
|
Tan L, Yan W, McCorvy JD, Cheng J. Biased Ligands of G Protein-Coupled Receptors (GPCRs): Structure-Functional Selectivity Relationships (SFSRs) and Therapeutic Potential. J Med Chem 2018; 61:9841-9878. [PMID: 29939744 DOI: 10.1021/acs.jmedchem.8b00435] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
G protein-coupled receptors (GPCRs) signal through both G-protein-dependent and G-protein-independent pathways, and β-arrestin recruitment is the most recognized one of the latter. Biased ligands selective for either pathway are expected to regulate biological functions of GPCRs in a more precise way, therefore providing new drug molecules with superior efficacy and/or reduced side effects. During the past decade, biased ligands have been discovered and developed for many GPCRs, such as the μ opioid receptor, the angiotensin II receptor type 1, the dopamine D2 receptor, and many others. In this Perspective, recent advances in this field are reviewed by discussing the structure-functional selectivity relationships (SFSRs) of GPCR biased ligands and the therapeutic potential of these molecules. Further understanding of the biological functions associated with each signaling pathway and structural basis for biased signaling will facilitate future drug design in this field.
Collapse
Affiliation(s)
- Liang Tan
- iHuman Institute , ShanghaiTech University , 393 Middle Huaxia Road , Pudong District, Shanghai 201210 , China
| | - Wenzhong Yan
- iHuman Institute , ShanghaiTech University , 393 Middle Huaxia Road , Pudong District, Shanghai 201210 , China
| | - John D McCorvy
- Department of Cell Biology, Neurobiology and Anatomy , Medical College of Wisconsin , 8701 W. Watertown Plank Road , Milwaukee , Wisconsin 53226 , United States
| | - Jianjun Cheng
- iHuman Institute , ShanghaiTech University , 393 Middle Huaxia Road , Pudong District, Shanghai 201210 , China
| |
Collapse
|
22
|
Biased G protein-coupled receptor agonism mediates Neu1 sialidase and matrix metalloproteinase-9 crosstalk to induce transactivation of insulin receptor signaling. Cell Signal 2018; 43:71-84. [DOI: 10.1016/j.cellsig.2017.12.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/26/2017] [Accepted: 12/21/2017] [Indexed: 11/19/2022]
|
23
|
The EphA2 receptor is activated through induction of distinct, ligand-dependent oligomeric structures. Commun Biol 2018; 1:15. [PMID: 30271902 PMCID: PMC6123813 DOI: 10.1038/s42003-018-0017-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/28/2018] [Indexed: 01/19/2023] Open
Abstract
The EphA2 receptor tyrosine kinase is capable of activating multiple diverse signaling pathways with roles in processes such as tissue homeostasis and cancer. EphA2 is known to form activated oligomers in the presence of ephrin-A ligands. Here, we characterize the lateral interactions between full-length EphA2 molecules in the plasma membrane in the presence of three types of ligands (dimeric ephrinA1-Fc, monomeric ephrinA1, and an engineered peptide ligand) as well as in the absence of ligand, using a quantitative FRET technique. The data show that EphA2 forms higher-order oligomers and two different types of dimers that all lead to increased EphA2 tyrosine phosphorylation, which is indicative of increased kinase-dependent signaling. We find that different ligands stabilize conformationally distinct oligomers that are assembled through two different interfaces. Our results suggest that these different oligomeric assemblies could have distinct signaling properties, contributing to the diverse activities of the EphA2 receptor. Deo Singh et al. use Fully Quantified Spectral Imaging-FRET to show that the EphA2 receptor forms dimers or higher order oligomers depending on the type of ligand, and that different ligands stabilize EphA2 dimers through distinct interfaces. These findings may explain how EphA2 activates diverse signaling pathways.
Collapse
|
24
|
Human GIP(3-30)NH 2 inhibits G protein-dependent as well as G protein-independent signaling and is selective for the GIP receptor with high-affinity binding to primate but not rodent GIP receptors. Biochem Pharmacol 2018; 150:97-107. [PMID: 29378179 DOI: 10.1016/j.bcp.2018.01.040] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022]
Abstract
GIP(3-30)NH2 is a high affinity antagonist of the GIP receptor (GIPR) in humans inhibiting insulin secretion via G protein-dependent pathways. However, its ability to inhibit G protein-independent signaling is unknown. Here we determine its action on arrestin-recruitment and receptor internalization in recombinant cells. As GIP is adipogenic, we evaluate the inhibitory actions of GIP(3-30)NH2 in human adipocytes. Finally, we determine the receptor selectivity of GIP(3-30)NH2 among other human and animal GPCRs. cAMP accumulation and β-arrestin 1 and 2 recruitment were studied in transiently transfected HEK293 cells and real-time internalization in transiently transfected HEK293A and in HEK293A β-arrestin 1 and 2 knockout cells. Furthermore, human subcutaneous adipocytes were assessed for cAMP accumulation following ligand stimulation. Competition binding was examined in transiently transfected COS-7 cells using human 125I-GIP(3-30)NH2. The selectivity of human GIP(3-30)NH2 was examined by testing for agonistic and antagonistic properties on 62 human GPCRs. Human GIP(3-30)NH2 inhibited GIP(1-42)-induced cAMP and β-arrestin 1 and 2 recruitment on the human GIPR and Schild plot analysis showed competitive antagonism with a pA2 and Hill slope of 16.8 nM and 1.11 ± 0.02 in cAMP, 10.6 nM and 1.15 ± 0.05 in β-arrestin 1 recruitment, and 10.2 nM and 1.06 ± 0.05 in β-arrestin 2 recruitment. Efficient internalization of the GIPR was dependent on the presence of either β-arrestin 1 or 2. Moreover, GIP(3-30)NH2 inhibited GIP(1-42)-induced internalization in a concentration-dependent manner and notably also inhibited GIP-mediated signaling in human subcutaneous adipocytes. Finally, the antagonist was established as GIPR selective among 62 human GPCRs being species-specific with high affinity binding to the human and non-human primate (Macaca fascicularis) GIPRs, and low affinity binding to the rat and mouse GIPRs (Kd values of 2.0, 2.5, 31.6 and 100 nM, respectively). In conclusion, human GIP(3-30)NH2 is a selective and species-specific GIPR antagonist with broad inhibition of signaling and internalization in transfected cells as well as in human adipocytes.
Collapse
|
25
|
Lee Y, Basith S, Choi S. Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled Receptors Utilizing Crystal Structures and Computational Simulations. J Med Chem 2017; 61:1-46. [PMID: 28657745 DOI: 10.1021/acs.jmedchem.6b01453] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) represent the largest and most physiologically important integral membrane protein family, and these receptors respond to a wide variety of physiological and environmental stimuli. GPCRs are among the most critical therapeutic targets for numerous human diseases, and approximately one-third of the currently marketed drugs target this receptor family. The recent breakthroughs in GPCR structural biology have significantly contributed to our understanding of GPCR function, ligand binding, and pharmacological action as well as to the design of new drugs. This perspective highlights the latest advances in GPCR structures with a focus on the receptor-ligand interactions of each receptor family in class A nonrhodopsin GPCRs as well as the structural features for their activation, biased signaling, and allosteric mechanisms. The current state-of-the-art methodologies of structure-based drug design (SBDD) approaches in the GPCR research field are also discussed.
Collapse
Affiliation(s)
- Yoonji Lee
- National Leading Research Laboratory (NLRL) of Molecular Modeling & Drug Design, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul 03760, Republic of Korea
| | - Shaherin Basith
- National Leading Research Laboratory (NLRL) of Molecular Modeling & Drug Design, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul 03760, Republic of Korea
| | - Sun Choi
- National Leading Research Laboratory (NLRL) of Molecular Modeling & Drug Design, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul 03760, Republic of Korea
| |
Collapse
|
26
|
Besserer-Offroy É, Brouillette RL, Lavenus S, Froehlich U, Brumwell A, Murza A, Longpré JM, Marsault É, Grandbois M, Sarret P, Leduc R. The signaling signature of the neurotensin type 1 receptor with endogenous ligands. Eur J Pharmacol 2017; 805:1-13. [DOI: 10.1016/j.ejphar.2017.03.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/17/2022]
|
27
|
Ryba DM, Li J, Cowan CL, Russell B, Wolska BM, Solaro RJ. Long-Term Biased β-Arrestin Signaling Improves Cardiac Structure and Function in Dilated Cardiomyopathy. Circulation 2017; 135:1056-1070. [PMID: 28104714 DOI: 10.1161/circulationaha.116.024482] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Biased agonism of the angiotensin II receptor is known to promote cardiac contractility. Our laboratory indicated that these effects may be attributable to changes at the level of the myofilaments. However, these signaling mechanisms remain unknown. Because a common finding in dilated cardiomyopathy is a reduction in the myofilament-Ca2+ response, we hypothesized that β-arrestin signaling would increase myofilament-Ca2+ responsiveness in a model of familial dilated cardiomyopathy and improve cardiac function and morphology. METHODS We treated a dilated cardiomyopathy-linked mouse model expressing a mutant tropomyosin (Tm-E54K) for 3 months with either TRV120067, a β-arrestin 2-biased ligand of the angiotensin II receptor, or losartan, an angiotensin II receptor blocker. At the end of the treatment protocol, we assessed cardiac function using echocardiography, the myofilament-Ca2+ response of detergent-extracted fiber bundles, and used proteomic approaches to understand changes in posttranslational modifications of proteins that may explain functional changes. We also assessed signaling pathways altered in vivo and by using isolated myocytes. RESULTS TRV120067- treated Tm-E54K mice showed improved cardiac structure and function, whereas losartan-treated mice had no improvement. Myofilaments of TRV120067-treated Tm-E54K mice had significantly improved myofilament-Ca2+ responsiveness, which was depressed in untreated Tm-E54K mice. We attributed these changes to increased MLC2v and MYPT1/2 phosphorylation seen only in TRV120067-treated mice. We found that the functional changes were attributable to an activation of ERK1/2-RSK3 signaling, mediated through β-arrestin, which may have a novel role in increasing MLC2v phosphorylation through a previously unrecognized interaction of β-arrestin localized to the sarcomere. CONCLUSIONS Long-term β-arrestin 2-biased agonism of the angiotensin II receptor may be a viable approach to the treatment of dilated cardiomyopathy by not only preventing maladaptive signaling, but also improving cardiac function by altering the myofilament-Ca2+ response via β-arrestin signaling pathways.
Collapse
Affiliation(s)
- David M Ryba
- From Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago (D.M.R., J.L., B.R., B.M.W., R.J.S.); Department of Medicine, Division of Cardiology, University of Illinois at Chicago (B.M.W.); and Trevena, Inc. King of Prussia, PA (B.M.W.)
| | - Jieli Li
- From Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago (D.M.R., J.L., B.R., B.M.W., R.J.S.); Department of Medicine, Division of Cardiology, University of Illinois at Chicago (B.M.W.); and Trevena, Inc. King of Prussia, PA (B.M.W.)
| | - Conrad L Cowan
- From Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago (D.M.R., J.L., B.R., B.M.W., R.J.S.); Department of Medicine, Division of Cardiology, University of Illinois at Chicago (B.M.W.); and Trevena, Inc. King of Prussia, PA (B.M.W.)
| | - Brenda Russell
- From Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago (D.M.R., J.L., B.R., B.M.W., R.J.S.); Department of Medicine, Division of Cardiology, University of Illinois at Chicago (B.M.W.); and Trevena, Inc. King of Prussia, PA (B.M.W.)
| | - Beata M Wolska
- From Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago (D.M.R., J.L., B.R., B.M.W., R.J.S.); Department of Medicine, Division of Cardiology, University of Illinois at Chicago (B.M.W.); and Trevena, Inc. King of Prussia, PA (B.M.W.)
| | - R John Solaro
- From Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago (D.M.R., J.L., B.R., B.M.W., R.J.S.); Department of Medicine, Division of Cardiology, University of Illinois at Chicago (B.M.W.); and Trevena, Inc. King of Prussia, PA (B.M.W.).
| |
Collapse
|
28
|
Váradi A, Marrone GF, Palmer TC, Narayan A, Szabó MR, Le Rouzic V, Grinnell SG, Subrath JJ, Warner E, Kalra S, Hunkele A, Pagirsky J, Eans SO, Medina JM, Xu J, Pan YX, Borics A, Pasternak GW, McLaughlin JP, Majumdar S. Mitragynine/Corynantheidine Pseudoindoxyls As Opioid Analgesics with Mu Agonism and Delta Antagonism, Which Do Not Recruit β-Arrestin-2. J Med Chem 2016; 59:8381-97. [PMID: 27556704 DOI: 10.1021/acs.jmedchem.6b00748] [Citation(s) in RCA: 232] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Natural products found in Mitragyna speciosa, commonly known as kratom, represent diverse scaffolds (indole, indolenine, and spiro pseudoindoxyl) with opioid activity, providing opportunities to better understand opioid pharmacology. Herein, we report the pharmacology and SAR studies both in vitro and in vivo of mitragynine pseudoindoxyl (3), an oxidative rearrangement product of the corynanthe alkaloid mitragynine. 3 and its corresponding corynantheidine analogs show promise as potent analgesics with a mechanism of action that includes mu opioid receptor agonism/delta opioid receptor antagonism. In vitro, 3 and its analogs were potent agonists in [(35)S]GTPγS assays at the mu opioid receptor but failed to recruit β-arrestin-2, which is associated with opioid side effects. Additionally, 3 developed analgesic tolerance more slowly than morphine, showed limited physical dependence, respiratory depression, constipation, and displayed no reward or aversion in CPP/CPA assays, suggesting that analogs might represent a promising new generation of novel pain relievers.
Collapse
Affiliation(s)
- András Váradi
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Gina F Marrone
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Travis C Palmer
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Ankita Narayan
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Márton R Szabó
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged, H-6726 Hungary
| | - Valerie Le Rouzic
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Steven G Grinnell
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Joan J Subrath
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Evelyn Warner
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Sanjay Kalra
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Amanda Hunkele
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Jeremy Pagirsky
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Shainnel O Eans
- Department of Pharmacodyanamics, University of Florida , Gainesville, Florida 032610, United States
| | - Jessica M Medina
- Department of Pharmacodyanamics, University of Florida , Gainesville, Florida 032610, United States
| | - Jin Xu
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Ying-Xian Pan
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Attila Borics
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged, H-6726 Hungary
| | - Gavril W Pasternak
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Jay P McLaughlin
- Department of Pharmacodyanamics, University of Florida , Gainesville, Florida 032610, United States
| | - Susruta Majumdar
- Molecular Pharmacology and Chemistry Program and Department of Neurology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| |
Collapse
|
29
|
Ogawa S, Watanabe T, Moriyuki K, Goto Y, Yamane S, Watanabe A, Tsuboi K, Kinoshita A, Okada T, Takeda H, Tani K, Maruyama T. Structural optimization and structure–functional selectivity relationship studies of G protein-biased EP2 receptor agonists. Bioorg Med Chem Lett 2016; 26:2446-2449. [DOI: 10.1016/j.bmcl.2016.03.110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 01/14/2023]
|
30
|
Ogawa S, Watanabe T, Sugimoto I, Moriyuki K, Goto Y, Yamane S, Watanabe A, Tsuboi K, Kinoshita A, Kigoshi H, Tani K, Maruyama T. Discovery of G Protein-Biased EP2 Receptor Agonists. ACS Med Chem Lett 2016; 7:306-11. [PMID: 26985320 DOI: 10.1021/acsmedchemlett.5b00455] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/03/2016] [Indexed: 01/14/2023] Open
Abstract
To identify G protein-biased and highly subtype-selective EP2 receptor agonists, a series of bicyclic prostaglandin analogues were designed and synthesized. Structural hybridization of EP2/4 dual agonist 5 and prostacyclin analogue 6, followed by simplification of the ω chain enabled us to discover novel EP2 agonists with a unique prostacyclin-like scaffold. Further optimization of the ω chain was performed to improve EP2 agonist activity and subtype selectivity. Phenoxy derivative 18a showed potent agonist activity and excellent subtype selectivity. Furthermore, a series of compounds were identified as G protein-biased EP2 receptor agonists. These are the first examples of biased ligands of prostanoid receptors.
Collapse
Affiliation(s)
- Seiji Ogawa
- Department of Chemistry,
Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8571, Japan
| | | | | | | | | | | | | | | | | | - Hideo Kigoshi
- Department of Chemistry,
Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8571, Japan
| | | | | |
Collapse
|
31
|
Rankovic Z, Brust TF, Bohn LM. Biased agonism: An emerging paradigm in GPCR drug discovery. Bioorg Med Chem Lett 2016; 26:241-250. [PMID: 26707396 PMCID: PMC5595354 DOI: 10.1016/j.bmcl.2015.12.024] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 01/11/2023]
Abstract
G protein coupled receptors have historically been one of the most druggable classes of cellular proteins. The members of this large receptor gene family couple to primary effectors, G proteins, that have built in mechanisms for regeneration and amplification of signaling with each engagement of receptor and ligand, a kinetic event in itself. In recent years GPCRs, have been found to interact with arrestin proteins to initiate signal propagation in the absence of G protein interactions. This pinnacle observation has changed a previously held notion of the linear spectrum of GPCR efficacy and uncovered a new paradigm in GPCR research and drug discovery that relies on multidimensionality of GPCR signaling. Ligands were found that selectively confer activity in one pathway over another, and this phenomenon has been referred to as 'biased agonism' or 'functional selectivity'. While great strides in the understanding of this phenomenon have been made in recent years, two critical questions still dominate the field: How can we rationally design biased GPCR ligands, and ultimately, which physiological responses are due to G protein versus arrestin interactions? This review will discuss the current understanding of some of the key aspects of biased signaling that are related to these questions, including mechanistic insights in the nature of biased signaling and methods for measuring ligand bias, as well as relevant examples of drug discovery applications and medicinal chemistry strategies that highlight the challenges and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Zoran Rankovic
- Discovery Chemistry and Research Technologies, Eli Lilly and Company, 893 South Delaware Street, Indianapolis, IN 46285, USA.
| | - Tarsis F Brust
- Department of Molecular Therapeutics, and Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Laura M Bohn
- Department of Molecular Therapeutics, and Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
32
|
Namkung Y, Radresa O, Armando S, Devost D, Beautrait A, Le Gouill C, Laporte SA. Quantifying biased signaling in GPCRs using BRET-based biosensors. Methods 2016; 92:5-10. [DOI: 10.1016/j.ymeth.2015.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/11/2015] [Indexed: 01/14/2023] Open
|
33
|
Quantification of adenosine A 1 receptor biased agonism: Implications for drug discovery. Biochem Pharmacol 2016; 99:101-12. [DOI: 10.1016/j.bcp.2015.11.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022]
|
34
|
Wierońska JM, Zorn SH, Doller D, Pilc A. Metabotropic glutamate receptors as targets for new antipsychotic drugs: Historical perspective and critical comparative assessment. Pharmacol Ther 2015; 157:10-27. [PMID: 26549541 DOI: 10.1016/j.pharmthera.2015.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this review, we aim to present, discuss and clarify our current understanding regarding the prediction of possible antipsychotic effects of metabotropic glutamate (mGlu) receptor ligands. The number of preclinical trials clearly indicates, that this group of compounds constitutes an excellent alternative to presently used antipsychotic therapy, being effective not only to positive, but also negative and cognitive symptoms of schizophrenia. Although the results of clinical trials that were performed for the group of mGlu2/3 agonists were not so enthusiastic as in animal studies, they still showed that mGlu ligands do not induced variety of side effects typical for presently used antipsychotics, and were generally well tolerated. The lack of satisfactory effectiveness towards schizophrenia symptoms of mGlu2/3 activators in humans could be a result of variety of uncontrolled factors and unidentified biomarkers different for each schizophrenia patient, that should be taken into consideration in the future set of clinical trials. The subject is still open for further research, and the novel classes of mGlu5 or mGlu2/3 agonists/PAMs were recently introduced, including the large group of compounds from the third group of mGlu receptors, especially of mGlu4 subtype. Finally, more precise treatment based on simultaneous administration of minimal doses of the ligands for two or more receptors, seems to be promising in the context of symptoms-specific schizophrenia treatment.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | | | | | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland.
| |
Collapse
|
35
|
Kassel S, Schwed JS, Stark H. Dopamine D3 receptor agonists as pharmacological tools. Eur Neuropsychopharmacol 2015; 25:1480-99. [PMID: 25498414 DOI: 10.1016/j.euroneuro.2014.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/23/2014] [Accepted: 11/04/2014] [Indexed: 01/10/2023]
Abstract
Dysregulation of the dopaminergic innervation in the central nervous system plays a key role in different neurological disorders like Parkinson´s disease, restless legs syndrome, schizophrenia etc. Although dopamine D3 receptors have been recognized as an important target in these diseases, their full pharmacological properties need further investigations. With focus on dopamine D3 receptor full agonists, this review has divided the ergoline and non-ergoline ligands in dissimilar chemical subclasses describing their pharmacodynamic properties on different related receptors, on species differences and their functional properties on different signaling mechanism. This is combined with a short description of structure-activity relationships for each class. Therefore, this overview should support the rational choice for the optimal compound selection based on affinity, selectivity and efficacy data in biochemical and pharmacological studies.
Collapse
Affiliation(s)
- S Kassel
- Heinrich-Heine-University, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - J S Schwed
- Heinrich-Heine-University, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - H Stark
- Heinrich-Heine-University, Universitaetsstr. 1, 40225 Duesseldorf, Germany.
| |
Collapse
|
36
|
Heifetz A, Schertler GFX, Seifert R, Tate CG, Sexton PM, Gurevich VV, Fourmy D, Cherezov V, Marshall FH, Storer RI, Moraes I, Tikhonova IG, Tautermann CS, Hunt P, Ceska T, Hodgson S, Bodkin MJ, Singh S, Law RJ, Biggin PC. GPCR structure, function, drug discovery and crystallography: report from Academia-Industry International Conference (UK Royal Society) Chicheley Hall, 1-2 September 2014. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2015; 388:883-903. [PMID: 25772061 PMCID: PMC4495723 DOI: 10.1007/s00210-015-1111-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/24/2015] [Indexed: 01/14/2023]
Abstract
G-protein coupled receptors (GPCRs) are the targets of over half of all prescribed drugs today. The UniProt database has records for about 800 proteins classified as GPCRs, but drugs have only been developed against 50 of these. Thus, there is huge potential in terms of the number of targets for new therapies to be designed. Several breakthroughs in GPCRs biased pharmacology, structural biology, modelling and scoring have resulted in a resurgence of interest in GPCRs as drug targets. Therefore, an international conference, sponsored by the Royal Society, with world-renowned researchers from industry and academia was recently held to discuss recent progress and highlight key areas of future research needed to accelerate GPCR drug discovery. Several key points emerged. Firstly, structures for all three major classes of GPCRs have now been solved and there is increasing coverage across the GPCR phylogenetic tree. This is likely to be substantially enhanced with data from x-ray free electron sources as they move beyond proof of concept. Secondly, the concept of biased signalling or functional selectivity is likely to be prevalent in many GPCRs, and this presents exciting new opportunities for selectivity and the control of side effects, especially when combined with increasing data regarding allosteric modulation. Thirdly, there will almost certainly be some GPCRs that will remain difficult targets because they exhibit complex ligand dependencies and have many metastable states rendering them difficult to resolve by crystallographic methods. Subtle effects within the packing of the transmembrane helices are likely to mask and contribute to this aspect, which may play a role in species dependent behaviour. This is particularly important because it has ramifications for how we interpret pre-clinical data. In summary, collaborative efforts between industry and academia have delivered significant progress in terms of structure and understanding of GPCRs and will be essential for resolving problems associated with the more difficult targets in the future.
Collapse
Key Words
- gpcrs, g-protein coupled receptors
- β2ar, β2-adrenergic receptor
- glp-1, glucagon-like peptide-1 receptor
- cck2r, cholecystokinin receptor-2
- δ-or, delta-opioid receptor
- crf1, corticotropin releasing factor receptor 1
- cxcr1, cxcr2, ccr4 and ccr5, chemokine receptors
- 5-ht2b and 5-ht2c, human 5-hydroxytryptamine receptors 2b and 2c, respectively
- h1, histamine receptor 1
- hm3r, human muscarinic m3 receptor
- dopamine d2 receptor
- α1b adrenergic receptor
- t4l, t4-lysozyme
- bril, apocytochrome b562ril
- xfels, x-ray free electron lasers
- sdm, site-directed mutagenesis
- md, molecular dynamic simulations
- 3d, three-dimensional
- 7tm, seven-transmembrane domain
- tm, trans-membrane helix
- ecl, extracellular loop
- hgmp, hierarchical gpcr modelling protocol
- glas, gpcr-likeness assessment score
- pros, pairwise protein similarity method
- pdb, protein data bank
Collapse
Affiliation(s)
- Alexander Heifetz
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ UK
| | - Gebhard F. X. Schertler
- D-BIOL, ETH Zurich Paul Scherrer Institute, Laboratory of Biomolecular Research, LBR OFLC 109, CH-5232 Villigen PSI, Switzerland
| | - Roland Seifert
- Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Christopher G. Tate
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH UK
| | - Patrick M. Sexton
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences 381 Royal Parade, Parkville, VIC 3052 Australia
| | - Vsevolod V. Gurevich
- Vanderbilt University Medical Center, 2200 Pierce Avenue, Preston Research Building, Nashville, TN 37232 USA
| | - Daniel Fourmy
- University of Toulouse, Bat L3, I2MC/INSERM U1048, Avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Vadim Cherezov
- Department of Chemistry, Bridge Institute, Los Angeles, CA USA
| | - Fiona H. Marshall
- Heptares Therapeutics, Biopark, Welwyn Garden City, Hertfordshire AL7 3AX UK
| | - R. Ian Storer
- Pfizer Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park, Cambridge, CB21 6GS UK
| | - Isabel Moraes
- Membrane Protein Laboratory at Diamond Light Source, Didcot, UK
| | - Irina G. Tikhonova
- Molecular Therapeutics, School of Pharmacy, Medical Biology Centre, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | | | - Peter Hunt
- Optibrium Ltd, 7221 Cambridge Research Park, Beach Drive, Cambridge, CB25 9TL UK
| | - Tom Ceska
- UCB Pharma, 216 Bath Road, Slough, SL1 3WE UK
| | - Simon Hodgson
- Hodgson Pharma Consulting Ltd, Stevenage Bioscience Catalyst, Stevenage, SG1 2FX UK
| | - Mike J. Bodkin
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ UK
| | - Shweta Singh
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ UK
| | - Richard J. Law
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ UK
| | - Philip C. Biggin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU UK
| |
Collapse
|
37
|
Sutou I, Nakatani T, Hashimoto T, Saito Y. Fentanyl tolerance in the treatment of cancer pain: a case of successful opioid switching from fentanyl to oxycodone at a reduced equivalent dose. J Pain Palliat Care Pharmacother 2015; 29:161-5. [PMID: 26095488 DOI: 10.3109/15360288.2015.1035832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Opioids are not generally deemed to have an analgesic ceiling effect on cancer pain. However, there have been occasional reports of tolerance to opioid development induced by multiple doses of fentanyl. The authors report a case of suspected tolerance to the analgesic effect of opioid, in which an increasing dose of fentanyl failed to relieve the patient's cancer pain symptoms, but opioid switching to oxycodone injections enabled a dose reduction to below the equivalent dose conversion ratio. The patient was a 60-year-old man diagnosed with pancreatic body carcinoma with multiple metastases. The base dose consisted of 12 mg/day of transdermal fentanyl patches (equivalent to 3.6 mg/day, 150 μg/h fentanyl injection), and rescue therapy consisted of 10 mg immediate-release oxycodone powders. Despite the total daily dose of fentanyl reaching 5.6 mg (equivalent to 560 mg oral morphine), the analgesic effect was inadequate; thus, an urgent adjustment was necessary. Due to the moderate dose of fentanyl, the switch to oxycodone injection was done incrementally at a daily dose equivalent to 25% of the fentanyl injection. The total dose of oxycodone was replaced approximately 53.5% of the dose of fentanyl prior to opioid switching.
Collapse
|
38
|
Keck TM, John WS, Czoty PW, Nader MA, Newman AH. Identifying Medication Targets for Psychostimulant Addiction: Unraveling the Dopamine D3 Receptor Hypothesis. J Med Chem 2015; 58:5361-80. [PMID: 25826710 PMCID: PMC4516313 DOI: 10.1021/jm501512b] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The dopamine D3 receptor (D3R) is a target for developing medications to treat substance use disorders. D3R-selective compounds with high affinity and varying efficacies have been discovered, providing critical research tools for cell-based studies that have been translated to in vivo models of drug abuse. D3R antagonists and partial agonists have shown especially promising results in rodent models of relapse-like behavior, including stress-, drug-, and cue-induced reinstatement of drug seeking. However, to date, translation to human studies has been limited. Herein, we present an overview and illustrate some of the pitfalls and challenges of developing novel D3R-selective compounds toward clinical utility, especially for treatment of cocaine abuse. Future research and development of D3R-selective antagonists and partial agonists for substance abuse remains critically important but will also require further evaluation and development of translational animal models to determine the best time in the addiction cycle to target D3Rs for optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Thomas M Keck
- †Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - William S John
- §Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157-1083, United States
| | - Paul W Czoty
- §Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157-1083, United States
| | - Michael A Nader
- §Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157-1083, United States
| | - Amy Hauck Newman
- †Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
39
|
Weichert D, Banerjee A, Hiller C, Kling RC, Hübner H, Gmeiner P. Molecular Determinants of Biased Agonism at the Dopamine D2 Receptor. J Med Chem 2015; 58:2703-17. [DOI: 10.1021/jm501889t] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Dietmar Weichert
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Ashutosh Banerjee
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Christine Hiller
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Ralf C. Kling
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
40
|
Konoura K, Fujii H, Imaide S, Gouda H, Hirayama S, Hirono S, Nagase H. Transformation of naltrexone into mesembrane and investigation of the binding properties of its intermediate derivatives to opioid receptors. Bioorg Med Chem 2015; 23:439-48. [DOI: 10.1016/j.bmc.2014.12.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/15/2014] [Accepted: 12/15/2014] [Indexed: 11/28/2022]
|
41
|
Hudson BD, Shimpukade B, Milligan G, Ulven T. The molecular basis of ligand interaction at free fatty acid receptor 4 (FFA4/GPR120). J Biol Chem 2014; 289:20345-58. [PMID: 24860101 PMCID: PMC4106347 DOI: 10.1074/jbc.m114.561449] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The long-chain fatty acid receptor FFA4 (previously GPR120) is receiving substantial interest as a novel target for the treatment of metabolic and inflammatory disease. This study examines for the first time the detailed mode of binding of both long-chain fatty acid and synthetic agonist ligands at FFA4 by integrating molecular modeling, receptor mutagenesis, and ligand structure-activity relationship approaches in an iterative format. In doing so, residues required for binding of fatty acid and synthetic agonists to FFA4 have been identified. This has allowed for the refinement of a well validated model of the mode of ligand-FFA4 interaction that will be invaluable in the identification of novel ligands and the future development of this receptor as a therapeutic target. The model reliably predicted the effects of substituent variations on agonist potency, and it was also able to predict the qualitative effect of binding site mutations in the majority of cases.
Collapse
Affiliation(s)
- Brian D Hudson
- From the Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom and
| | - Bharat Shimpukade
- the Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Graeme Milligan
- From the Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom and
| | - Trond Ulven
- the Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| |
Collapse
|