1
|
Vakalopoulos A, Basting D, Brechmann M, Teller H, Boultadakis Arapinis M, Straub A, Mittendorf J, Meininghaus M, Müller T, Nowak-Reppel K, Schäfer M, Wittwer M, Kullmann M, Terjung C, Lang D, Poethko T, Marquardt T, Freudenberger T, Mondritzki T, Hüser J, Heckmann M, Tinel H. Discovery of BAY 2413555, First Selective Positive Allosteric Modulator of the M2 Receptor to Restore Cardiac Autonomic Balance. J Med Chem 2024; 67:19165-19187. [PMID: 39463278 DOI: 10.1021/acs.jmedchem.4c01590] [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: 10/29/2024]
Abstract
Autonomic disbalance, i.e., sympathetic overactivation and parasympathetic withdrawal, is a causal driver of disease progression in heart failure. While sympatholytic drugs are established treatments, no drug therapy restoring vagal control of cardiac function is available. We report here the HTS-based discovery of a novel class of 1,8-naphthyridin-4(1H)-one carboxamides acting as positive allosteric modulators (PAMs) of the M2 muscarinic acetylcholine receptor (M2R). M2R is the main postsynaptic myocyte receptor regulating heart rate, electrical conduction, and contractile strength. Extensive optimization of the screening hit in terms of potency, permeation, metabolic stability, and solubility ultimately resulted in the discovery of the first-in-class clinical candidate BAY 2413555 (27). With an overall technical profile compatible with once-daily oral administration in a phase 1 study, no apparent effects on blood pressure, and a mechanism that largely preserves autonomic regulatory capacity, BAY 2413555 could be the tool to finally study the restoration of autonomic balance.
Collapse
Affiliation(s)
- Alexandros Vakalopoulos
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Daniel Basting
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Markus Brechmann
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- CSL Innovation GmbH, Emil-von-Behring-Str. 76, Marburg 35041, Germany
| | - Henrik Teller
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- micromod Partikeltechnologie GmbH, Schillingallee 68, Rostock 18057, Germany
| | | | - Alexander Straub
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Joachim Mittendorf
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Mark Meininghaus
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Thomas Müller
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Katrin Nowak-Reppel
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Martina Schäfer
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- Nuvisan ICB GmbH, Müllerstrasse 178, Berlin 13353, Germany
| | - Matthias Wittwer
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Maximilian Kullmann
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- BioNTech SE, An der Goldgrube 12, Mainz 55131, Germany
| | - Carsten Terjung
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Dieter Lang
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Thorsten Poethko
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Tobias Marquardt
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Till Freudenberger
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Thomas Mondritzki
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- University of Witten/Herdecke, Witten 58455, Germany
| | - Jörg Hüser
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- Axxam S.p.A., Openzone - Via Meucci 3, Bresso, Milan 20091, Italy
| | - Michael Heckmann
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Hanna Tinel
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| |
Collapse
|
2
|
Do HN, Wang J, Miao Y. Deep Learning Dynamic Allostery of G-Protein-Coupled Receptors. JACS AU 2023; 3:3165-3180. [PMID: 38034960 PMCID: PMC10685416 DOI: 10.1021/jacsau.3c00503] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023]
Abstract
G-protein-coupled receptors (GPCRs) make up the largest superfamily of human membrane proteins and represent primary targets of ∼1/3 of currently marketed drugs. Allosteric modulators have emerged as more selective drug candidates compared with orthosteric agonists and antagonists. However, many X-ray and cryo-EM structures of GPCRs resolved so far exhibit negligible differences upon the binding of positive and negative allosteric modulators (PAMs and NAMs). The mechanism of dynamic allosteric modulation in GPCRs remains unclear. In this work, we have systematically mapped dynamic changes in free energy landscapes of GPCRs upon binding of allosteric modulators using the Gaussian accelerated molecular dynamics (GaMD), deep learning (DL), and free energy prOfiling Workflow (GLOW). GaMD simulations were performed for a total of 66 μs on 44 GPCR systems in the presence and absence of the modulator. DL and free energy calculations revealed significantly reduced dynamic fluctuations and conformational space of GPCRs upon modulator binding. While the modulator-free GPCRs often sampled multiple low-energy conformational states, the NAMs and PAMs confined the inactive and active agonist-G-protein-bound GPCRs, respectively, to mostly only one specific conformation for signaling. Such cooperative effects were significantly reduced for binding of the selective modulators to "non-cognate" receptor subtypes. Therefore, GPCR allostery exhibits a dynamic "conformational selection" mechanism. In the absence of available modulator-bound structures as for most current GPCRs, it is critical to use a structural ensemble of representative GPCR conformations rather than a single structure for compound docking ("ensemble docking"), which will potentially improve structure-based design of novel allosteric drugs of GPCRs.
Collapse
Affiliation(s)
| | - Jinan Wang
- Computational Biology Program
and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, United States
| | | |
Collapse
|
3
|
Qi A, Kling HE, Billard N, Rodriguez AL, Peng L, Dickerson JW, Engers JL, Bender AM, Moehle MS, Lindsley CW, Rook JM, Niswender CM. Development of a Selective and High Affinity Radioligand, [ 3H]VU6013720, for the M 4 Muscarinic Receptor. Mol Pharmacol 2023; 104:195-202. [PMID: 37595966 PMCID: PMC10586508 DOI: 10.1124/molpharm.122.000643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
M4 muscarinic receptors are highly expressed in the striatum and cortex, brain regions that are involved in diseases such as Parkinson's disease, schizophrenia, and dystonia. Despite potential therapeutic advantages of specifically targeting the M4 receptor, it has been historically challenging to develop highly selective ligands, resulting in undesired off-target activity at other members of the muscarinic receptor family. Recently, we have reported first-in-class, potent, and selective M4 receptor antagonists. As an extension of that work, we now report the development and characterization of a radiolabeled M4 receptor antagonist, [3H]VU6013720, with high affinity (pKd of 9.5 ± 0.2 at rat M4, 9.7 at mouse M4, and 10 ± 0.1 at human M4 with atropine to define nonspecific binding) and no significant binding at the other muscarinic subtypes. Binding assays using this radioligand in rodent brain tissues demonstrate loss of specific binding in Chrm4 knockout animals. Dissociation kinetics experiments with various muscarinic ligands show differential effects on the dissociation of [3H]VU6013720 from M4 receptors, suggesting a binding site that is overlapping but may be distinct from the orthosteric site. Overall, these results demonstrate that [3H]VU6013720 is the first highly selective antagonist radioligand for the M4 receptor, representing a useful tool for studying the basic biology of M4 as well for the support of M4 receptor-based drug discovery. SIGNIFICANCE STATEMENT: This manuscript describes the development and characterization of a novel muscarinic (M) acetylcholine subtype 4 receptor antagonist radioligand, [3H]VU6013720. This ligand binds to or overlaps with the acetylcholine binding site, providing a highly selective radioligand for the M4 receptor that can be used to quantify M4 protein expression in vivo and probe the selective interactions of acetylcholine with M4 versus the other members of the muscarinic receptor family.
Collapse
Affiliation(s)
- Aidong Qi
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Haley E Kling
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Natasha Billard
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Alice L Rodriguez
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Li Peng
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Jonathan W Dickerson
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Julie L Engers
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Aaron M Bender
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Mark S Moehle
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Craig W Lindsley
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Jerri M Rook
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| | - Colleen M Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery (A.Q., H.E.K., N.B., A.L.R., L.P., J.W.D., J.L.E., A.M.B., C.W.L., J.M.R., C.M.N.) and Department of Chemistry (C.W.L.), Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N); Vanderbilt Brain Institute (C.M.N.) and Vanderbilt Institute of Chemical Biology (C.W.L., C.M.N.),Vanderbilt University School of Medicine, Nashville, Tennessee; and Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration (M.S.M.), University of Florida, Gainesville, Florida
| |
Collapse
|
4
|
Do H, Wang J, Miao Y. Deep Learning Dynamic Allostery of G-Protein-Coupled Receptors. RESEARCH SQUARE 2023:rs.3.rs-2543463. [PMID: 36865316 PMCID: PMC9980202 DOI: 10.21203/rs.3.rs-2543463/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest superfamily of human membrane proteins and represent primary targets of ~ 1/3 of currently marketed drugs. Allosteric modulators have emerged as more selective drug candidates compared with orthosteric agonists and antagonists. However, many X-ray and cryo-EM structures of GPCRs resolved so far exhibit negligible differences upon binding of positive and negative allosteric modulators (PAMs and NAMs). Mechanism of dynamic allosteric modulation in GPCRs remains unclear. In this work, we have systematically mapped dynamic changes in free energy landscapes of GPCRs upon binding of allosteric modulators using the Gaussian accelerated molecular dynamics (GaMD), Deep Learning (DL) and free energy prOfiling Workflow (GLOW). A total of 18 available high-resolution experimental structures of allosteric modulator-bound class A and B GPCRs were collected for simulations. A number of 8 computational models were generated to examine selectivity of the modulators by changing their target receptors to different subtypes. All-atom GaMD simulations were performed for a total of 66 μs on 44 GPCR systems in the presence/absence of the modulator. DL and free energy calculations revealed significantly reduced conformational space of GPCRs upon modulator binding. While the modulator-free GPCRs often sampled multiple low-energy conformational states, the NAMs and PAMs confined the inactive and active agonist-G protein-bound GPCRs, respectively, to mostly only one specific conformation for signaling. Such cooperative effects were significantly reduced for binding of the selective modulators to "non-cognate" receptor subtypes in the computational models. Therefore, comprehensive DL of extensive GaMD simulations has revealed a general dynamic mechanism of GPCR allostery, which will greatly facilitate rational design of selective allosteric drugs of GPCRs.
Collapse
|
5
|
Do HN, Wang J, Miao Y. Deep Learning Dynamic Allostery of G-Protein-Coupled Receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.15.524128. [PMID: 36711515 PMCID: PMC9882226 DOI: 10.1101/2023.01.15.524128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest superfamily of human membrane proteins and represent primary targets of ~1/3 of currently marketed drugs. Allosteric modulators have emerged as more selective drug candidates compared with orthosteric agonists and antagonists. However, many X-ray and cryo-EM structures of GPCRs resolved so far exhibit negligible differences upon binding of positive and negative allosteric modulators (PAMs and NAMs). Mechanism of dynamic allosteric modulation in GPCRs remains unclear. In this work, we have systematically mapped dynamic changes in free energy landscapes of GPCRs upon binding of allosteric modulators using the Gaussian accelerated molecular dynamics (GaMD), Deep Learning (DL) and free energy prOfiling Workflow (GLOW). A total of 18 available high-resolution experimental structures of allosteric modulator-bound class A and B GPCRs were collected for simulations. A number of 8 computational models were generated to examine selectivity of the modulators by changing their target receptors to different subtypes. All-atom GaMD simulations were performed for a total of 66 μs on 44 GPCR systems in the presence/absence of the modulator. DL and free energy calculations revealed significantly reduced conformational space of GPCRs upon modulator binding. While the modulator-free GPCRs often sampled multiple low-energy conformational states, the NAMs and PAMs confined the inactive and active agonist-G protein-bound GPCRs, respectively, to mostly only one specific conformation for signaling. Such cooperative effects were significantly reduced for binding of the selective modulators to "non-cognate" receptor subtypes in the computational models. Therefore, comprehensive DL of extensive GaMD simulations has revealed a general dynamic mechanism of GPCR allostery, which will greatly facilitate rational design of selective allosteric drugs of GPCRs.
Collapse
Affiliation(s)
- Hung N. Do
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| |
Collapse
|
6
|
Myslivecek J. Multitargeting nature of muscarinic orthosteric agonists and antagonists. Front Physiol 2022; 13:974160. [PMID: 36148314 PMCID: PMC9486310 DOI: 10.3389/fphys.2022.974160] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Muscarinic receptors (mAChRs) are typical members of the G protein-coupled receptor (GPCR) family and exist in five subtypes from M1 to M5. Muscarinic receptor subtypes do not sufficiently differ in affinity to orthosteric antagonists or agonists; therefore, the analysis of receptor subtypes is complicated, and misinterpretations can occur. Usually, when researchers mainly specialized in CNS and peripheral functions aim to study mAChR involvement in behavior, learning, spinal locomotor networks, biological rhythms, cardiovascular physiology, bronchoconstriction, gastrointestinal tract functions, schizophrenia, and Parkinson's disease, they use orthosteric ligands and they do not use allosteric ligands. Moreover, they usually rely on manufacturers' claims that could be misleading. This review aimed to call the attention of researchers not deeply focused on mAChR pharmacology to this fact. Importantly, limited selective binding is not only a property of mAChRs but is a general attribute of most neurotransmitter receptors. In this review, we want to give an overview of the most common off-targets for established mAChR ligands. In this context, an important point is a mention the tremendous knowledge gap on off-targets for novel compounds compared to very well-established ligands. Therefore, we will summarize reported affinities and give an outline of strategies to investigate the subtype's function, thereby avoiding ambiguous results. Despite that, the multitargeting nature of drugs acting also on mAChR could be an advantage when treating such diseases as schizophrenia. Antipsychotics are a perfect example of a multitargeting advantage in treatment. A promising strategy is the use of allosteric ligands, although some of these ligands have also been shown to exhibit limited selectivity. Another new direction in the development of muscarinic selective ligands is functionally selective and biased agonists. The possible selective ligands, usually allosteric, will also be listed. To overcome the limited selectivity of orthosteric ligands, the recommended process is to carefully examine the presence of respective subtypes in specific tissues via knockout studies, carefully apply "specific" agonists/antagonists at appropriate concentrations and then calculate the probability of a specific subtype involvement in specific functions. This could help interested researchers aiming to study the central nervous system functions mediated by the muscarinic receptor.
Collapse
Affiliation(s)
- Jaromir Myslivecek
- Institute of Physiology, 1 Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
7
|
Stroganova T, Vasilin VK, Dotsenko VV, Aksenov NA, Morozov PG, Vassiliev PM, Volynkin VA, Krapivin GD. Unusual Oxidative Dimerization in the 3-Aminothieno[2,3- b]pyridine-2-carboxamide Series. ACS OMEGA 2021; 6:14030-14048. [PMID: 34124427 PMCID: PMC8190813 DOI: 10.1021/acsomega.1c00341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/18/2021] [Indexed: 06/12/2023]
Abstract
Noncatalyzed, regio- and stereoselective hypochlorite oxidation of 3-aminothieno[2,3-b]pyridine-2-carboxamides is presented. Unexpectedly, the oxidation proceeded by different mechanistic pathways, and different products were formed, depending on the nature of solvents used. A possible mechanism, the structure of products, kinetics and dynamics of intramolecular processes, and biological activity of products are discussed.
Collapse
Affiliation(s)
- Tatyana
A. Stroganova
- Department
of Bioorganic Chemistry, Kuban State Technological
University, Krasnodar 350072, Russian Federation
| | - Vladimir K. Vasilin
- Department
of Bioorganic Chemistry, Kuban State Technological
University, Krasnodar 350072, Russian Federation
| | - Victor V. Dotsenko
- Department
of Organic Chemistry and Technologies, Kuban
State University, Krasnodar 350040, Russian Federation
- Department
of Organic Chemistry, North Caucasus Federal
University, Stavropol 355009, Russian Federation
| | - Nicolai A. Aksenov
- Department
of Organic Chemistry, North Caucasus Federal
University, Stavropol 355009, Russian Federation
| | - Pavel G. Morozov
- Department
of Chemistry of Natural Compounds, Southern
Federal University, Rostov-on-Don 344006, Russian Federation
| | - Pavel M. Vassiliev
- Volgograd
State Medical University, Volgograd 400131, Russian Federation
| | - Vitaly A. Volynkin
- Department
of Inorganic Chemistry, Kuban State University, Krasnodar 350040, Russian Federation
| | - Gennady D. Krapivin
- Scientific
Research Institute of Chemistry of Heterocyclic Compounds, Kuban State Technological University, Krasnodar 350072, Russian Federation
| |
Collapse
|
8
|
She X, Pegoli A, Gruber CG, Wifling D, Carpenter J, Hübner H, Chen M, Wan J, Bernhardt G, Gmeiner P, Holliday ND, Keller M. Red-Emitting Dibenzodiazepinone Derivatives as Fluorescent Dualsteric Probes for the Muscarinic Acetylcholine M2 Receptor. J Med Chem 2020; 63:4133-4154. [PMID: 32233403 DOI: 10.1021/acs.jmedchem.9b02172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Xueke She
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Andrea Pegoli
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Corinna G. Gruber
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - David Wifling
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Jessica Carpenter
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Derby Road, Nottingham NG7 2UH, U.K
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Mengya Chen
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Jianfei Wan
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Nicholas D. Holliday
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Derby Road, Nottingham NG7 2UH, U.K
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| |
Collapse
|
9
|
Maggi L, Carloni P, Rossetti G. Modeling the allosteric modulation on a G-Protein Coupled Receptor: the case of M2 muscarinic Acetylcholine Receptor in complex with LY211960. Sci Rep 2020; 10:3037. [PMID: 32080232 PMCID: PMC7033091 DOI: 10.1038/s41598-020-59289-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/08/2020] [Indexed: 11/18/2022] Open
Abstract
Allosteric modulation is involved in a plethora of diverse protein functions, which are fundamental for cells' life. This phenomenon can be thought as communication between two topographically distinct site of a protein structure. How this communication occurs is still matter of debate. Many different descriptions have been presented so far. Here we consider a specific case where any significant conformational change is involved upon allosteric modulator binding and the phenomenon is depicted as a vibrational energy diffusion process between distant protein regions. We applied this model, by employing computational tools, to the human muscarinic receptor M2, a transmembrane protein G-protein coupled receptor known to undergo allosteric modulation whose recently X-ray structure has been recently resolved both with and without the presence of a particular allosteric modulator. Our calculations, performed on these two receptor structures, suggest that for this case the allosteric modulator modifies the energy current between functionally relevant regions of the protein; this allows to identify the main residues responsible for this modulation. These results contribute to shed light on the molecular basis of allosteric modulation and may help design new allosteric ligands.
Collapse
Affiliation(s)
- L Maggi
- Computational Biomedicine Section, Institute of Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.
| | - P Carloni
- Computational Biomedicine Section, Institute of Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
- Institute for Neuroscience and Medicine INM-11, Forschungszentrum Jülich, 52428, Jülich, Germany
- Department of Physics, RWTH Aachen University, 52078, Aachen, Germany
| | - G Rossetti
- Computational Biomedicine Section, Institute of Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
- Department of Neurology, University Hospital Aachen, 52078, Aachen, Germany
- Simulation Laboratory Biology, Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
- Department of Oncology, Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| |
Collapse
|
10
|
Xu P, Liu Y, Wang L, Wu Y, Zhou X, Xiao J, Zheng J, Xue M. Phencynonate S-isomer as a eutomer is a novel central anticholinergic drug for anti-motion sickness. Sci Rep 2019; 9:2000. [PMID: 30760797 PMCID: PMC6374516 DOI: 10.1038/s41598-018-38305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/21/2018] [Indexed: 12/04/2022] Open
Abstract
To compare and evaluate the differences of stereoselective activity, the binding affinity, metabolism, transport and molecular docking of phencynonate isomers to muscarinic acetylcholine receptor (mAChR) were investigated in this study. The rotation stimulation and locomotor experiments were used to evaluate anti-motion sickness effects. The competitive affinity with [3H]-QNB and molecular docking were used for studying the interactions between the two isomers and mAChR. The stereoselective mechanism of isomers was investigated by incubation with rat liver microsomes, a protein binding assay and membrane permeability assay across a Caco-2 cell monolayer using a chiral column HPLC method. The results indicated that S-isomer was more effective against motion sickness and had not anxiogenic action at therapeutic doses. S-isomer has the higher affinity and activity for mAChR in cerebral cortex and acted as a competitive mAChR antagonist. The stereoselective elimination of S-isomer was primarily affected by CYP1B1 and 17A1 enzymes, resulting in a higher metabolic stability and slower elimination. Phencynonate S isomer, as a eutomer and central anticholinergic chiral drug, is a novel anti-motion sickness drug with higher efficacy and lower central side effect. Our data assisted the development of a novel drug and eventual use of S-isomer in clinical practice.
Collapse
Affiliation(s)
- Pingxiang Xu
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Nervous System Drugs, Beijing, 100053, China
| | - Ying Liu
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Yanjing Medical College, Capital Medical University, Beijing, 101300, China
| | - Liyun Wang
- Beijing Institutes of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yi Wu
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xuelin Zhou
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Junhai Xiao
- Beijing Institutes of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jianquan Zheng
- Beijing Institutes of Pharmacology and Toxicology, Beijing, 100850, China
| | - Ming Xue
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Beijing Engineering Research Center for Nervous System Drugs, Beijing, 100053, China.
| |
Collapse
|
11
|
Jiménez-Rosés M, Matsoukas MT, Caltabiano G, Cordomí A. Ligand-Triggered Structural Changes in the M 2 Muscarinic Acetylcholine Receptor. J Chem Inf Model 2018; 58:1074-1082. [PMID: 29671585 DOI: 10.1021/acs.jcim.8b00108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The muscarinic M2 acetylcholine receptor, one of the few G-protein coupled receptors that has not only been crystallized in both active and inactive conformations but also in the presence of a positive allosteric modulator, is an interesting system to study the molecular mechanisms of GPCR activation and ligand allosterism. Here, we have employed molecular dynamics (MD) simulations (adding to 14 μs in total) to study conformational changes triggered by the inverse agonist R-(-)-3-quinuclidinyl-benzilate (QNB) in the structure of the active M2 receptor (PBD ID 4MQS ) after replacement of the agonist iperoxo by the inverse agonist QNB. This permitted us to identify the sequence of events in the deactivation mechanism of the M2 acetylcholine receptor, which results first in the rearrangement of the transmission switch, the subsequent opening of the extracellular portion of the receptor and finally, the closure of the intracellular part. We also evaluate the effect of the positive allosteric modulator LY2119620 when bound simultaneously with the orthosteric agonist iperoxo and find that it restricts the conformation of Trp4227.35 in a position that modulates the orientation of the Tyr4267.39 at the orthosteric-binding pocket.
Collapse
Affiliation(s)
- Mireia Jiménez-Rosés
- Laboratori de Medicina Computacional, Unitat de Bioestadística , Facultat de Medicina, Universitat Autònoma de Barcelona , 08193 Bellaterra , Spain
| | | | - Gianluigi Caltabiano
- Laboratori de Medicina Computacional, Unitat de Bioestadística , Facultat de Medicina, Universitat Autònoma de Barcelona , 08193 Bellaterra , Spain
| | - Arnau Cordomí
- Laboratori de Medicina Computacional, Unitat de Bioestadística , Facultat de Medicina, Universitat Autònoma de Barcelona , 08193 Bellaterra , Spain
| |
Collapse
|
12
|
Schober DA, Croy CH, Ruble CL, Tao R, Felder CC. Identification, expression and functional characterization of M4L, a muscarinic acetylcholine M4 receptor splice variant. PLoS One 2017; 12:e0188330. [PMID: 29211764 PMCID: PMC5718406 DOI: 10.1371/journal.pone.0188330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/03/2017] [Indexed: 11/24/2022] Open
Abstract
Rodent genomic alignment sequences support a 2-exon model for muscarinic M4 receptor. Using this model a novel N-terminal extension was discovered in the human muscarinic acetylcholine M4 receptor. An open reading frame was discovered in the human, mouse and rat with a common ATG (methionine start codon) that extended the N-terminus of the muscarinic acetylcholine M4 receptor subtype by 155 amino acids resulting in a longer variant. Transcriptional evidence for this splice variant was confirmed by RNA-Seq and RT-PCR experiments performed from human donor brain prefrontal cortices. We detected a human upstream exon indicating the translation of the mature longer M4 receptor transcript. The predicted size for the longer two-exon M4 receptor splice variant with the additional 155 amino acid N-terminal extension, designated M4L is 69.7 kDa compared to the 53 kDa canonical single exon M4 receptor (M4S). Western blot analysis from a mammalian overexpression system, and saturation radioligand binding with [3H]-NMS (N-methyl-scopolamine) demonstrated the expression of this new splice variant. Comparative pharmacological characterization between the M4L and M4S receptors revealed that both the orthosteric and allosteric binding sites for both receptors were very similar despite the addition of an N-terminal extension.
Collapse
Affiliation(s)
- Douglas A. Schober
- Neuroscience, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Carrie H. Croy
- Neuroscience, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Cara L. Ruble
- Neuroscience, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Ran Tao
- Lieber Institute for Brain Development, Baltimore, Maryland, United States of America
| | - Christian C. Felder
- Neuroscience, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
13
|
She X, Pegoli A, Mayr J, Hübner H, Bernhardt G, Gmeiner P, Keller M. Heterodimerization of Dibenzodiazepinone-Type Muscarinic Acetylcholine Receptor Ligands Leads to Increased M 2R Affinity and Selectivity. ACS OMEGA 2017; 2:6741-6754. [PMID: 30023530 PMCID: PMC6044897 DOI: 10.1021/acsomega.7b01085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/05/2017] [Indexed: 05/13/2023]
Abstract
In search for selective ligands for the muscarinic acetylcholine receptor (MR) subtype M2, the dimeric ligand approach, that is combining two pharmacophores in one and the same molecule, was pursued. Different types (agonists, antagonists, orthosteric, and allosteric) of monomeric MR ligands were combined by various linkers with a dibenzodiazepinone-type MR antagonist, affording five types of heterodimeric compounds ("DIBA-xanomeline," "DIBA-TBPB," "DIBA-77-LH-28-1," "DIBA-propantheline," and "DIBA-4-DAMP"), which showed high M2R affinities (pKi > 8.3). The heterodimeric ligand UR-SK75 (46) exhibited the highest M2R affinity and selectivity [pKi (M1R-M5R): 8.84, 10.14, 7.88, 8.59, and 7.47]. Two tritium-labeled dimeric derivatives ("DIBA-xanomeline"-type: [3H]UR-SK71 ([3H]44) and "DIBA-TBPB"-type: [3H]UR-SK59 ([3H]64)) were prepared to investigate their binding modes at hM2R. Saturation-binding experiments showed that these compounds address the orthosteric binding site of the M2R. The investigation of the effect of various allosteric MR modulators [gallamine (13), W84 (14), and LY2119620 (15)] on the equilibrium (13-15) or saturation (14) binding of [3H]64 suggested a competitive mechanism between [3H]64 and the investigated allosteric ligands, and consequently a dualsteric binding mode of 64 at the M2R.
Collapse
Affiliation(s)
- Xueke She
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Andrea Pegoli
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Judith Mayr
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Emil Fischer Center, Friedrich Alexander University, Schuhstr. 19, D-91052 Erlangen, Germany
| | - Günther Bernhardt
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Emil Fischer Center, Friedrich Alexander University, Schuhstr. 19, D-91052 Erlangen, Germany
| | - Max Keller
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
- E-mail: . Phone: (+49)941-9433329.
Fax: (+49)941-9434820 (M.K.)
| |
Collapse
|
14
|
Bock A, Schrage R, Mohr K. Allosteric modulators targeting CNS muscarinic receptors. Neuropharmacology 2017; 136:427-437. [PMID: 28935216 DOI: 10.1016/j.neuropharm.2017.09.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 12/21/2022]
Abstract
Muscarinic acetylcholine receptors are G protein-coupled receptors (GPCRs) which are broadly expressed in the central nervous system (CNS) and other tissues in the periphery. They emerge as important drug targets for a number of diseases including Alzheimer's disease, Parkinson's disease, and schizophrenia. Muscarinic receptors are divided into five subtypes (M1-M5) of which M1-M4 have been crystalized. All subtypes possess at least one allosteric binding site which is located in the extracellular region of the receptor on top of the ACh (i.e. orthosteric) binding site. The former can be specifically targeted by chemical compounds (mostly small molecules) and binding of such allosteric modulators affects the affinity and/or efficacy of orthosteric ligands. This allows highly specific modulation of GPCR function and, from a drug discovery point of view, may be advantageous in terms of subtype selectivity and biased signaling. There is a plethora of allosteric modulators for all five muscarinic receptor subtypes. This review presents the basic principles of allosteric modulation of GPCRs on both the molecular and structural level focusing on allosteric modulators of the muscarinic receptor family. Further we discuss dualsteric (i.e. bitopic orthosteric/allosteric) ligands emphasizing their potential in modulating muscarinic receptor dynamics and signaling. The common mechanisms of muscarinic receptor allosteric modulation have been proven to be generalizable and are at play at many, if not all GPCRs. Given this paradigmatic role of muscarinic receptors we suggest that also new developments in muscarinic allosteric modulation may also be extended to other members of the GPCR superfamily. This article is part of the Special Issue entitled 'Neuropharmacology on Muscarinic Receptors'.
Collapse
Affiliation(s)
- Andreas Bock
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany.
| | - Ramona Schrage
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany
| | - Klaus Mohr
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany
| |
Collapse
|
15
|
Pegoli A, She X, Wifling D, Hübner H, Bernhardt G, Gmeiner P, Keller M. Radiolabeled Dibenzodiazepinone-Type Antagonists Give Evidence of Dualsteric Binding at the M2 Muscarinic Acetylcholine Receptor. J Med Chem 2017; 60:3314-3334. [PMID: 28388054 DOI: 10.1021/acs.jmedchem.6b01892] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Andrea Pegoli
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Xueke She
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - David Wifling
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Emil Fischer Center, Friedrich Alexander University, Schuhstrasse 19, D-91052 Erlangen, Germany
| | - Günther Bernhardt
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Emil Fischer Center, Friedrich Alexander University, Schuhstrasse 19, D-91052 Erlangen, Germany
| | - Max Keller
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| |
Collapse
|
16
|
Campbell AP, Wakelin LPG, Denny WA, Finch AM. Homobivalent Conjugation Increases the Allosteric Effect of 9-aminoacridine at the α1-Adrenergic Receptors. Mol Pharmacol 2017; 91:135-144. [PMID: 27903755 DOI: 10.1124/mol.116.105874] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022] Open
Abstract
The α1-adrenergic receptors are targets for a number of cardiovascular and central nervous system conditions, but the current drugs for these receptors lack specificity to be of optimal clinical value. Allosteric modulators offer an alternative mechanism of action to traditional α1-adrenergic ligands, yet there is little information describing this drug class at the α1-adrenergic receptors. We have identified a series of 9-aminoacridine compounds that demonstrate allosteric modulation of the α1A- and α1B-adrenergic receptors. The 9-aminoacridines increase the rate of [3H]prazosin dissociation from the α1A- and α1B-adrenergic receptors and noncompetitively inhibit receptor activation by the endogenous agonist norepinephrine. The structurally similar compound, tacrine, which is a known allosteric modulator of the muscarinic receptors, is also shown to be a modulator of the α1-adrenergic receptors, which suggests a general lack of selectivity for allosteric binding sites across aminergic G protein-coupled receptor. Conjugation of two 9-aminoacridine pharmacophores, using linkers of varying length, increases the potency and efficacy of the allosteric effects of this ligand, likely through optimization of bitopic engagement of the allosteric and orthosteric binding sites of the receptor. Such a bivalent approach may provide a mechanism for fine tuning the efficacy of allosteric compounds in future drug design efforts.
Collapse
Affiliation(s)
- Adrian P Campbell
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| | - Laurence P G Wakelin
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| | - William A Denny
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| | - Angela M Finch
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| |
Collapse
|
17
|
Jeon WJ, Dean B, Scarr E, Gibbons A. The Role of Muscarinic Receptors in the Pathophysiology of Mood Disorders: A Potential Novel Treatment? Curr Neuropharmacol 2016; 13:739-49. [PMID: 26630954 PMCID: PMC4759313 DOI: 10.2174/1570159x13666150612230045] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/30/2014] [Accepted: 02/28/2015] [Indexed: 12/21/2022] Open
Abstract
The central cholinergic system has been implicated in the pathophysiology of mood disorders. An imbalance in central cholinergic neurotransmitter activity has been proposed to contribute to the manic and depressive episodes typical of these disorders. Neuropharmacological studies into the effects of cholinergic agonists and antagonists on mood state have provided considerable support for this hypothesis. Furthermore, recent clinical studies have shown that the pan-CHRM antagonist, scopolamine, produces rapid-acting antidepressant effects in individuals with either major depressive disorder (MDD) or bipolar disorder (BPD), such as bipolar depression, contrasting the delayed therapeutic response of conventional mood stabilisers and antidepressants. This review presents recent data from neuroimaging, post-mortem and genetic studies supporting the involvement of muscarinic cholinergic receptors (CHRMs), particularly CHRM2, in the pathophysiology of MDD and BPD. Thus, novel drugs that selectively target CHRMs with negligible effects in the peripheral nervous system might produce more rapid and robust clinical improvement in patients with BPD and MDD.
Collapse
Affiliation(s)
| | - Brian Dean
- Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, Victoria 3052, Australia.
| | | | | |
Collapse
|
18
|
Smith DL, Davoren JE, Edgerton JR, Lazzaro JT, Lee CW, Neal S, Zhang L, Grimwood S. Characterization of a Novel M1 Muscarinic Acetylcholine Receptor Positive Allosteric Modulator Radioligand, [3H]PT-1284. Mol Pharmacol 2016; 90:177-87. [PMID: 27382013 DOI: 10.1124/mol.116.104737] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/30/2016] [Indexed: 02/14/2025] Open
Abstract
Selective activation of the M1 muscarinic acetylcholine receptor (mAChR) via a positive allosteric modulator (PAM) is a new approach for the treatment of the cognitive impairments associated with schizophrenia and Alzheimer's disease. Herein, we describe the characterization of an M1 PAM radioligand, 8-((1S,2S)-2-hydroxycyclohexyl)-5-((6-(methyl-t3)pyridin-3-yl)methyl)-8,9-dihydro-7H-pyrrolo[3,4-hour]quinolin-7-one ([(3)H]PT-1284), as a tool for characterizing the M1 allosteric binding site, as well as profiling novel M1 PAMs. 8-((1S,2S)-2-Hydroxycyclohexyl)-5-((6-methylpyridin-3-yl)methyl)-8,9-dihydro-7H-pyrrolo[3,4-hour]quinolin-7-one (PT-1284 ( 1: )) was shown to potentiate acetylcholine (ACh) in an M1 fluorometric imaging plate reader (FLIPR) functional assay (EC50, 36 nM) and carbachol in a hippocampal slice electrophysiology assay (EC50, 165 nM). PT-1284 ( 1: ) also reduced the concentration of ACh required to inhibit [(3)H]N-methylscopolamine ([(3)H]NMS) binding to M1, left-shifting the ACh Ki approximately 19-fold at 10 μM. Saturation analysis of a human M1 mAChR stable cell line showed that [(3)H]PT-1284 bound to M1 mAChR in the presence of 1 mM ACh with Kd, 4.23 nM, and saturable binding capacity (Bmax), 6.38 pmol/mg protein. M1 selective PAMs were shown to inhibit [(3)H]PT-1284 binding in a concentration-responsive manner, whereas M1 allosteric and orthosteric agonists showed weak affinity (>30 μM). A strong positive correlation (R(2) = 0.86) was found to exist between affinity values generated for nineteen M1 PAMs in the [(3)H]PT-1284 binding assay and the EC50 values of these ligands in a FLIPR functional potentiation assay. These data indicate that there is a strong positive correlation between M1 PAM binding affinity and functional activity, and that [(3)H]PT-1284 can serve as a tool for pharmacological investigation of M1 mAChR PAMs.
Collapse
Affiliation(s)
- Deborah L Smith
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - Jennifer E Davoren
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - Jeremy R Edgerton
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - John T Lazzaro
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - Che-Wah Lee
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - Sarah Neal
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - Lei Zhang
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| | - Sarah Grimwood
- Neuroscience and Pain Research Unit (D.L.S., J.R.E., S.N., S.G.) and Worldwide Medicinal Chemistry (J.E.D., L.Z.), Pfizer Inc., Cambridge, Massachusetts; Primary Pharmacology Group (J.T.L.) and Worldwide Medicinal Chemistry (C.-W.L.), Pfizer Inc., Groton, Connecticut
| |
Collapse
|
19
|
Yu Z, Liu J, van Veldhoven JPD, IJzerman AP, Schalij MJ, Pijnappels DA, Heitman LH, de Vries AAF. Allosteric Modulation of Kv11.1 (hERG) Channels Protects Against Drug-Induced Ventricular Arrhythmias. Circ Arrhythm Electrophysiol 2016; 9:e003439. [PMID: 27071825 DOI: 10.1161/circep.115.003439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 02/04/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Ventricular arrhythmias as a result of unintentional blockade of the Kv11.1 (hERG [human ether-à-go-go-related gene]) channel are a major safety concern in drug development. In past years, several highly prescribed drugs have been withdrawn for their ability to cause such proarrhythmia. Here, we investigated whether the proarrhythmic risk of existing drugs could be reduced by Kv11.1 allosteric modulators. METHODS AND RESULTS Using [(3)H]dofetilide-binding assays with membranes of human Kv11.1-expressing human embryonic kidney 293 cells, 2 existing compounds (VU0405601 and ML-T531) and a newly synthesized compound (LUF7244) were found to be negative allosteric modulators of dofetilide binding to the Kv11.1 channel, with LUF7244 showing the strongest effect at 10 μmol/L. The Kv11.1 affinities of typical blockers (ie, dofetilide, astemizole, sertindole, and cisapride) were significantly decreased by LUF7244. Treatment of confluent neonatal rat ventricular myocyte (NRVM) monolayers with astemizole or sertindole caused heterogeneous prolongation of action potential duration and a high incidence of early afterdepolarizations on 1-Hz electric point stimulation, occasionally leading to unstable, self-terminating tachyarrhythmias. Pretreatment of NRVMs with LUF7244 prevented these proarrhythmic effects. NRVM monolayers treated with LUF7244 alone displayed electrophysiological properties indistinguishable from those of untreated NRVM cultures. Prolonged exposure of NRVMs to LUF7244 or LUF7244 plus astemizole did not affect their viability, excitability, and contractility as assessed by molecular, immunological, and electrophysiological assays. CONCLUSIONS Allosteric modulation of the Kv11.1 channel efficiently suppresses drug-induced ventricular arrhythmias in vitro by preventing potentially arrhythmogenic changes in action potential characteristics, raising the possibility to resume the clinical use of unintended Kv11.1 blockers via pharmacological combination therapy.
Collapse
Affiliation(s)
- Zhiyi Yu
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Jia Liu
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Jacobus P D van Veldhoven
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Adriaan P IJzerman
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Martin J Schalij
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Daniël A Pijnappels
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Laura H Heitman
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.).
| | - Antoine A F de Vries
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.).
| |
Collapse
|
20
|
Thal DM, Sun B, Feng D, Nawaratne V, Leach K, Felder CC, Bures MG, Evans DA, Weis WI, Bachhawat P, Kobilka TS, Sexton PM, Kobilka BK, Christopoulos A. Crystal structures of the M1 and M4 muscarinic acetylcholine receptors. Nature 2016; 531:335-40. [PMID: 26958838 DOI: 10.1038/nature17188] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022]
Abstract
Muscarinic M1-M5 acetylcholine receptors are G-protein-coupled receptors that regulate many vital functions of the central and peripheral nervous systems. In particular, the M1 and M4 receptor subtypes have emerged as attractive drug targets for treatments of neurological disorders, such as Alzheimer's disease and schizophrenia, but the high conservation of the acetylcholine-binding pocket has spurred current research into targeting allosteric sites on these receptors. Here we report the crystal structures of the M1 and M4 muscarinic receptors bound to the inverse agonist, tiotropium. Comparison of these structures with each other, as well as with the previously reported M2 and M3 receptor structures, reveals differences in the orthosteric and allosteric binding sites that contribute to a role in drug selectivity at this important receptor family. We also report identification of a cluster of residues that form a network linking the orthosteric and allosteric sites of the M4 receptor, which provides new insight into how allosteric modulation may be transmitted between the two spatially distinct domains.
Collapse
Affiliation(s)
- David M Thal
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, Victoria, Australia
| | - Bingfa Sun
- ConfometRx, 3070 Kenneth Street, Santa Clara, California 95054, USA
| | - Dan Feng
- ConfometRx, 3070 Kenneth Street, Santa Clara, California 95054, USA
| | - Vindhya Nawaratne
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, Victoria, Australia
| | - Katie Leach
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, Victoria, Australia
| | | | - Mark G Bures
- Computational Chemistry and Chemoinformatics, Eli Lilly, Indianapolis, Indiana 46285, USA
| | - David A Evans
- Computational Chemistry and Chemoinformatics, Eli Lilly, Sunninghill Road, Windlesham GU20 6PH, UK
| | - William I Weis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Priti Bachhawat
- ConfometRx, 3070 Kenneth Street, Santa Clara, California 95054, USA
| | - Tong Sun Kobilka
- ConfometRx, 3070 Kenneth Street, Santa Clara, California 95054, USA
| | - Patrick M Sexton
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, Victoria, Australia
| | - Brian K Kobilka
- ConfometRx, 3070 Kenneth Street, Santa Clara, California 95054, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Arthur Christopoulos
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, Victoria, Australia
| |
Collapse
|
21
|
Abstract
Radioligand binding assays provide sensitive and quantitative information about guanine nucleotide protein G protein-coupled receptor (GPCR) expression and affinity for a wide variety of ligands, making them essential for drug structure-activity studies and basic GPCR research. Three basic radioligand binding protocols, saturation, indirect (competition, displacement, or modulation), and kinetic binding assays, are used to assess GPCR expression (Bmax), equilibrium dissociation constants for radioligands (Kd) and nonradioactive ligands (Ki), association and dissociation rates, and to distinguish competitive and allosteric mechanisms of GPCR-ligand interactions. Nonspecific radioligand binding may be mitigated by appropriate choices of reaction conditions. Radioligand depletion (bound radioactivity >10% of total radioligand), which compromises accuracy of Kd and Ki measurements, can be limited by adjusting receptor concentration and appropriate radioligand choice. Accurate Kd and Ki values in saturation and indirect binding assays depend on binding equilibrium. Equilibration time for high-affinity ligands, with slow dissociation rates, may require much extended incubation times or increased incubation temperature.
Collapse
Affiliation(s)
- Colleen A Flanagan
- School of Physiology and Medical Research Council Receptor Biology Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Wits Parktown, Johannesburg, South Africa.
| |
Collapse
|
22
|
Croy CH, Schober DA, Xiao H, Quets A, Christopoulos A, Felder CC. Characterization of the novel positive allosteric modulator, LY2119620, at the muscarinic M(2) and M(4) receptors. Mol Pharmacol 2014; 86:106-15. [PMID: 24807965 DOI: 10.1124/mol.114.091751] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The M(4) receptor is a compelling therapeutic target, as this receptor modulates neural circuits dysregulated in schizophrenia, and there is clinical evidence that muscarinic agonists possess both antipsychotic and procognitive efficacy. Recent efforts have shifted toward allosteric ligands to maximize receptor selectivity and manipulate endogenous cholinergic and dopaminergic signaling. In this study, we present the pharmacological characterization of LY2119620 (3-amino-5-chloro-N-cyclopropyl-4-methyl-6-[2-(4-methylpiperazin-1-yl)-2-oxoethoxy] thieno[2,3-b]pyridine-2-carboxamide), a M(2)/M(4) receptor-selective positive allosteric modulator (PAM), chemically evolved from hits identified through a M4 allosteric functional screen. Although unsuitable as a therapeutic due to M(2) receptor cross-reactivity and, thus, potential cardiovascular liability, LY2119620 surpassed previous congeners in potency and PAM activity and broadens research capabilities through its development into a radiotracer. Characterization of LY2119620 revealed evidence of probe dependence in both binding and functional assays. Guanosine 5'-[γ-(35)S]-triphosphate assays displayed differential potentiation depending on the orthosteric-allosteric pairing, with the largest cooperativity observed for oxotremorine M (Oxo-M) LY2119620. Further [(3)H]Oxo-M saturation binding, including studies with guanosine-5'-[(β,γ)-imido]triphosphate, suggests that both the orthosteric and allosteric ligands can alter the population of receptors in the active G protein-coupled state. Additionally, this work expands the characterization of the orthosteric agonist, iperoxo, at the M(4) receptor, and demonstrates that an allosteric ligand can positively modulate the binding and functional efficacy of this high efficacy ligand. Ultimately, it was the M(2) receptor pharmacology and PAM activity with iperoxo that made LY2119620 the most suitable allosteric partner for the M(2) active-state structure recently solved (Kruse et al., 2013), a structure that provides crucial insights into the mechanisms of orthosteric activation and allosteric modulation of muscarinic receptors.
Collapse
Affiliation(s)
- Carrie H Croy
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (C.H.C., D.A.S., H.X., A.Q., C.C.F.); and Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (A.C.)
| | - Douglas A Schober
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (C.H.C., D.A.S., H.X., A.Q., C.C.F.); and Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (A.C.)
| | - Hongling Xiao
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (C.H.C., D.A.S., H.X., A.Q., C.C.F.); and Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (A.C.)
| | - Anne Quets
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (C.H.C., D.A.S., H.X., A.Q., C.C.F.); and Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (A.C.)
| | - Arthur Christopoulos
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (C.H.C., D.A.S., H.X., A.Q., C.C.F.); and Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (A.C.)
| | - Christian C Felder
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (C.H.C., D.A.S., H.X., A.Q., C.C.F.); and Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (A.C.)
| |
Collapse
|