1
|
Chestnykh D, Kalinichenko LS, von Hörsten S, Kornhuber J, Müller CP. The mirror preference test: A reverse translational approach to study anomalous subjective experience in rats. Brain Res Bull 2025; 222:111247. [PMID: 39921146 DOI: 10.1016/j.brainresbull.2025.111247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Significant advancements have been made in the treatment of psychotic disorders, yet current pharmacotherapy remains inadequate. Symptoms related to the misinterpretation of reality are crucial for diagnosis but pose challenges for preclinical research. In humans, a Mirror-Gazing test is used to examine abnormal self-experience and to predict the risk of schizophrenia. To address this, we developed a task to evaluate anomalous subjective experiences in rats using a Mirror-Preference test. Here we demonstrate that naive rats show a preference for a mirror chamber, which was followed by significant habituation over a series of trials. In subsequent tests, we utilized dimmed lighting and net-covered mirrors to induce incomplete mirror images. Acute stimulation with amphetamine (AMPH, 3 mg/kg, i.p.) further increased the preference for the mirror. In a model of psychosis induced by chronic AMPH administration, rats showed fewer and shorter interactions with the mirror but maintained their preference for the chamber where psychotic-like animals were given additional AMPH stimulation (1.5 mg/kg, i.p.) before testing. Here, a surprising reversal in chamber preference was observed, along with decreased frequency and duration of mirror contact, suggesting mirror avoidance. The AMPH-presensitized rats also exhibited hyperlocomotion and elevated anxiety, indicative of psychotic-like behaviour. Although self-recognition in rodents is debatable, recent studies suggest they can discriminate mirror images. We propose that limited visual perception that meets brain monoamine sensitization may trigger visual illusions as part of a psychotic-like state in rats. This novel approach can be utilized to test intervention strategies for psychosis in rats.
Collapse
Affiliation(s)
- Daria Chestnykh
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stephan von Hörsten
- Department of Experimental Therapy, Preclinical Experimental Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
2
|
Peri L, Matzov D, Huxley DR, Rainish A, Fierro F, Sapir L, Pfeiffer T, Waterloo L, Hübner H, Peleg Y, Gmeiner P, McCormick PJ, Weikert D, Niv MY, Shalev-Benami M. A bitter anti-inflammatory drug binds at two distinct sites of a human bitter taste GPCR. Nat Commun 2024; 15:9991. [PMID: 39557861 PMCID: PMC11574016 DOI: 10.1038/s41467-024-54157-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
Bitter taste receptors (TAS2Rs), a subfamily of G-protein coupled receptors (GPCRs) expressed orally and extraorally, elicit signaling in response to a large set of tastants. Among 25 functional TAS2Rs encoded in the human genome, TAS2R14 is the most promiscuous, and responds to hundreds of chemically diverse ligands. Here we present the cryo-electron microscopy (cryo-EM) structure of the human TAS2R14 in complex with its signaling partner gustducin, and bound to flufenamic acid (FFA), a clinically approved nonsteroidal anti-inflammatory drug. The structure reveals an unusual binding mode, where two copies of FFA are bound at distinct pockets: one at the canonical receptor site within the trans-membrane bundle, and the other in the intracellular facet, bridging the receptor with gustducin. Together with a pocket-specific BRET-based ligand binding assay, these results illuminate bitter taste signaling and provide tools for a site-targeted compound design.
Collapse
Affiliation(s)
- Lior Peri
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Fritz Haber Research Center, and the Harvey M. Kruger Center for Nanoscience & Nanotechnology, Institute of Chemistry, The Hebrew University, Jerusalem, Israel
| | - Donna Matzov
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Dominic R Huxley
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary, University of London, Charterhouse Square, London, UK
| | - Alon Rainish
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Fritz Haber Research Center, and the Harvey M. Kruger Center for Nanoscience & Nanotechnology, Institute of Chemistry, The Hebrew University, Jerusalem, Israel
| | - Fabrizio Fierro
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Fritz Haber Research Center, and the Harvey M. Kruger Center for Nanoscience & Nanotechnology, Institute of Chemistry, The Hebrew University, Jerusalem, Israel
| | - Liel Sapir
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Fritz Haber Research Center, and the Harvey M. Kruger Center for Nanoscience & Nanotechnology, Institute of Chemistry, The Hebrew University, Jerusalem, Israel
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, Israel
| | - Tara Pfeiffer
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Waterloo
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yoav Peleg
- Structural Proteomics Unit (SPU), Life Sciences Core Facilities (LSCF), Weizmann Institute of Science, Rehovot, Israel
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAUNeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Peter J McCormick
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary, University of London, Charterhouse Square, London, UK
- Department of Pharmacology and Therapeutics, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
- FAUNeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
- The Fritz Haber Research Center, and the Harvey M. Kruger Center for Nanoscience & Nanotechnology, Institute of Chemistry, The Hebrew University, Jerusalem, Israel.
| | - Moran Shalev-Benami
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Vogt H, Shinkwin P, Huber ME, Staffen N, Hübner H, Gmeiner P, Schiedel M, Weikert D. Development of a Fluorescent Ligand for the Intracellular Allosteric Binding Site of the Neurotensin Receptor 1. ACS Pharmacol Transl Sci 2024; 7:1533-1545. [PMID: 38751637 PMCID: PMC11092115 DOI: 10.1021/acsptsci.4c00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
The membrane protein family of G protein-coupled receptors (GPCRs) represents a major class of drug targets. Over the last years, the presence of additional intracellular binding sites besides the canonical orthosteric binding pocket has been demonstrated for an increasing number of GPCRs. Allosteric modulators harnessing these pockets may represent valuable alternatives when targeting the orthosteric pocket is not successful for drug development. Starting from SBI-553, a recently discovered intracellular allosteric modulator for neurotensin receptor subtype 1 (NTSR1), we developed the fluorescent molecular probe 14. Compound 14 binds to NTSR1 with an affinity of 0.68 μM in the presence of the agonist NT(8-13). NanoBRET-based ligand binding assays with 14 were established to derive the affinity and structure-activity relationships for allosteric NTSR1 modulators in a direct and nonisotopic manner, thereby facilitating the search for and optimization of novel allosteric NTSR1 ligands. As a consequence of cooperativity between the ligands binding to the allosteric and orthosteric pocket, compound 14 can also be used to investigate orthosteric NTSR1 agonists and antagonists. Moreover, employing 14 as a probe in a drug library screening, we identified novel chemotypes as binders for the intracellular allosteric SBI-553 binding pocket of NTSR1 with single-digit micromolar affinity. These hits may serve as interesting starting points for the development of novel intracellular allosteric ligands for NTSR1 as a highly interesting yet unexploited drug target in the fields of pain and addiction disorder therapy.
Collapse
Affiliation(s)
- Hannah Vogt
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Patrick Shinkwin
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Max E. Huber
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Nico Staffen
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- FAU
NeW − Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Matthias Schiedel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
| | - Dorothee Weikert
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- FAU
NeW − Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
4
|
Zhang M, Chen T, Lu X, Lan X, Chen Z, Lu S. G protein-coupled receptors (GPCRs): advances in structures, mechanisms, and drug discovery. Signal Transduct Target Ther 2024; 9:88. [PMID: 38594257 PMCID: PMC11004190 DOI: 10.1038/s41392-024-01803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of human membrane proteins and an important class of drug targets, play a role in maintaining numerous physiological processes. Agonist or antagonist, orthosteric effects or allosteric effects, and biased signaling or balanced signaling, characterize the complexity of GPCR dynamic features. In this study, we first review the structural advancements, activation mechanisms, and functional diversity of GPCRs. We then focus on GPCR drug discovery by revealing the detailed drug-target interactions and the underlying mechanisms of orthosteric drugs approved by the US Food and Drug Administration in the past five years. Particularly, an up-to-date analysis is performed on available GPCR structures complexed with synthetic small-molecule allosteric modulators to elucidate key receptor-ligand interactions and allosteric mechanisms. Finally, we highlight how the widespread GPCR-druggable allosteric sites can guide structure- or mechanism-based drug design and propose prospects of designing bitopic ligands for the future therapeutic potential of targeting this receptor family.
Collapse
Affiliation(s)
- Mingyang Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xun Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaobing Lan
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziqiang Chen
- Department of Orthopedics, Changhai Hospital, Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Shaoyong Lu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
5
|
Gross F, Mancini A, Breton B, Kobayashi H, Pereira PHS, Le Gouill C, Bouvier M, Schann S, Leroy X, Sabbagh L. EGFR signaling and pharmacology in oncology revealed with innovative BRET-based biosensors. Commun Biol 2024; 7:250. [PMID: 38429428 PMCID: PMC10907714 DOI: 10.1038/s42003-024-05965-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
Mutations of receptor tyrosine kinases (RTKs) are associated with the development of many cancers by modifying receptor signaling and contributing to drug resistance in clinical settings. We present enhanced bystander bioluminescence resonance energy transfer-based biosensors providing new insights into RTK biology and pharmacology critical for the development of more effective RTK-targeting drugs. Distinct SH2-specific effector biosensors allow for real-time and spatiotemporal monitoring of signal transduction pathways engaged upon RTK activation. Using EGFR as a model, we demonstrate the capacity of these biosensors to differentiate unique signaling signatures, with EGF and Epiregulin ligands displaying differences in efficacy, potency, and responses within different cellular compartments. We further demonstrate that EGFR single point mutations found in Glioblastoma or non-small cell lung cancer, impact the constitutive activity of EGFR and response to tyrosine kinase inhibitor. The BRET-based biosensors are compatible with microscopy, and more importantly characterize the next generation of therapeutics directed against RTKs.
Collapse
Affiliation(s)
- Florence Gross
- Domain Therapeutics North America Inc., 7171 Frederick-Banting, Saint-Laurent, Quebec, H4S 1Z9, Canada
| | - Arturo Mancini
- Domain Therapeutics North America Inc., 7171 Frederick-Banting, Saint-Laurent, Quebec, H4S 1Z9, Canada
| | - Billy Breton
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Hiroyuki Kobayashi
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Pedro Henrique Scarpelli Pereira
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Stephan Schann
- Domain Therapeutics SA, 220 Boulevard Gonthier D'Andernach, 67400, Strasbourg-Illkirch, France
| | - Xavier Leroy
- Domain Therapeutics SA, 220 Boulevard Gonthier D'Andernach, 67400, Strasbourg-Illkirch, France
| | - Laurent Sabbagh
- Domain Therapeutics North America Inc., 7171 Frederick-Banting, Saint-Laurent, Quebec, H4S 1Z9, Canada.
| |
Collapse
|
6
|
Hamed O, Jayasinghe V, Giembycz MA. The β-Blocker Carvedilol and Related Aryloxypropanolamines Promote ERK1/2 Phosphorylation in HEK293 Cells with K A Values Distinct From Their Equilibrium Dissociation Constants as β 2-Adrenoceptor Antagonists: Evidence for Functional Affinity. J Pharmacol Exp Ther 2024; 388:688-700. [PMID: 38129128 DOI: 10.1124/jpet.123.001920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
The determination of affinity by using functional assays is important in drug discovery because it provides a more relevant estimate of the strength of interaction of a ligand to its cognate receptor than radioligand binding. However, empirical evidence for so-called, "functional affinity" is limited. Herein, we determined whether the affinity of carvedilol, a β-adrenoceptor antagonist used to treat heart failure that also promotes extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation, differed between these two pharmacological activities. Four structurally related β-adrenoceptor antagonists (alprenolol, carazolol, pindolol, propranolol) that also activated ERK1/2 were included as comparators to enhance our understanding of how these drugs work in the clinical setting. In HEK293 cells stably expressing the human β 2-adrenoceptor carvedilol and related aryloxypropanolamines were partial agonists of ERK1/2 phosphorylation with potencies ([A]50s) that were lower than their equilibrium dissociation constants (K Bs) as β 2-adrenoceptor antagonists. As the [A]50 of a partial agonist is a good approximation of its K B, then these data indicated that the affinities of carvedilol and related ligands for these two activities were distinct. Moreover, there was a significant negative rank order correlation between the [A]50 of each ligand to activate ERK1/2 and their intrinsic activities (i.e., as intrinsic activity for ERK1/2 phosphorylation increased, so did affinity). Genome editing revealed that the transducer that coupled the β 2-adrenoceptor to ERK1/2 phosphorylation in response to carvedilol and other β 2-adrenoceptor antagonists was Gαs. Collectively, these data support the concept of "functional affinity" and indicate that the ability of the β 2-adrenoceptor to recruit Gαs may influence the affinity of the activating ligand. SIGNIFICANCE STATEMENT: In HEK293 cells overexpressing the human β2-adrenoceptor carvedilol and four related aryloxypropanolamines behaved as β2-adrenoceptor antagonists and partial agonists of ERK1/2 phosphorylation with rank orders of affinity that were distinct. These data imply that carvedilol and other β-blockers can stabilize the β2-adrenoceptor in different affinity conformations that are revealed when functionally distinct responses are measured. This is the basis for the pharmacological concept of "functional affinity."
Collapse
Affiliation(s)
- Omar Hamed
- Lung Health Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Varuna Jayasinghe
- Lung Health Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Lung Health Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Hetzler B, Donthamsetti P, Peitsinis Z, Stanley C, Trauner D, Isacoff EY. Optical Control of Dopamine D2-like Receptors with Cell-Specific Fast-Relaxing Photoswitches. J Am Chem Soc 2023; 145:18778-18788. [PMID: 37586061 PMCID: PMC10472511 DOI: 10.1021/jacs.3c02735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Indexed: 08/18/2023]
Abstract
Dopamine D2-like receptors (D2R, D3R, and D4R) control diverse physiological and behavioral functions and are important targets for the treatment of a variety of neuropsychiatric disorders. Their complex distribution and activation kinetics in the brain make it difficult to target specific receptor populations with sufficient precision. We describe a new toolkit of light-activatable, fast-relaxing, covalently taggable chemical photoswitches that fully activate, partially activate, or block D2-like receptors. This technology combines the spatiotemporal precision of a photoswitchable ligand (P) with cell type and spatial specificity of a genetically encoded membrane anchoring protein (M) to which the P tethers. These tools set the stage for targeting endogenous D2-like receptor signaling with molecular, cellular, and spatiotemporal precision using only one wavelength of light.
Collapse
Affiliation(s)
- Belinda
E. Hetzler
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Prashant Donthamsetti
- Molecular
and Cell Biology, University of California,
Berkeley, Berkeley, California 94720, United States
| | - Zisis Peitsinis
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Cherise Stanley
- Molecular
and Cell Biology, University of California,
Berkeley, Berkeley, California 94720, United States
| | - Dirk Trauner
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Department
of Chemistry and Department of Systems Pharmacology and Translational
Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ehud Y. Isacoff
- Molecular
and Cell Biology, University of California,
Berkeley, Berkeley, California 94720, United States
- Helen
Wills Neuroscience Institute, University
of California, Berkeley, California 94720, United States
- Weill Neurohub, University of California, Berkeley, Berkeley, California 94720, United States
- Molecular
Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
8
|
Graßl F, Bock L, Huete-Huerta González Á, Schiller M, Gmeiner P, König J, Fromm MF, Hübner H, Heinrich MR. Exploring Structural Determinants of Bias among D4 Subtype-Selective Dopamine Receptor Agonists. J Med Chem 2023. [PMID: 37450764 DOI: 10.1021/acs.jmedchem.3c00537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
The high affinity dopamine D4 receptor ligand APH199 and derivatives thereof exhibit bias toward the Gi signaling pathway over β-arrestin recruitment compared to quinpirole. Based on APH199, two novel groups of D4 subtype selective ligands were designed and evaluated, in which the original benzyl phenylsemicarbazide substructure was replaced by either a biphenylmethyl urea or a biphenyl urea moiety. Functional assays revealed a range of different bias profiles among the newly synthesized compounds, namely, with regard to efficacy, potency, and GRK2 dependency, in which bias factors range from 1 to over 300 and activation from 15% to over 98% compared to quinpirole. These observations demonstrate that within bias, an even more precise tuning toward a particular profile is possible, which─in a general sense─could become an important aspect in future drug development. Docking studies enabled further insight into the role of the ECL2 and the EPB in the emergence of bias, thereby taking advantage of the diversity of functionally selective D4 agonists now available.
Collapse
Affiliation(s)
- Fabian Graßl
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Leonard Bock
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Álvaro Huete-Huerta González
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Martin Schiller
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Markus R Heinrich
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
9
|
Zell L, Bretl A, Temml V, Schuster D. Dopamine Receptor Ligand Selectivity-An In Silico/In Vitro Insight. Biomedicines 2023; 11:1468. [PMID: 37239139 PMCID: PMC10216180 DOI: 10.3390/biomedicines11051468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Different dopamine receptor (DR) subtypes are involved in pathophysiological conditions such as Parkinson's Disease (PD), schizophrenia and depression. While many DR-targeting drugs have been approved by the U.S. Food and Drug Administration (FDA), only a very small number are truly selective for one of the DR subtypes. Additionally, most of them show promiscuous activity at related G-protein coupled receptors, thus suffering from diverse side-effect profiles. Multiple studies have shown that combined in silico/in vitro approaches are a valuable contribution to drug discovery processes. They can also be applied to divulge the mechanisms behind ligand selectivity. In this study, novel DR ligands were investigated in vitro to assess binding affinities at different DR subtypes. Thus, nine D2R/D3R-selective ligands (micro- to nanomolar binding affinities, D3R-selective profile) were successfully identified. The most promising ligand exerted nanomolar D3R activity (Ki = 2.3 nM) with 263.7-fold D2R/D3R selectivity. Subsequently, ligand selectivity was rationalized in silico based on ligand interaction with a secondary binding pocket, supporting the selectivity data determined in vitro. The developed workflow and identified ligands could aid in the further understanding of the structural motifs responsible for DR subtype selectivity, thus benefitting drug development in D2R/D3R-associated pathologies such as PD.
Collapse
Affiliation(s)
| | | | | | - Daniela Schuster
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; (L.Z.); (A.B.); (V.T.)
| |
Collapse
|
10
|
Liu R, Qi J, Wang H, Fan L, Zhang P, Yu J, Tan L, Wang S, Cheng J. Transformation of a Dopamine D 2 Receptor Agonist to Partial Agonists as Novel Antipsychotic Agents. J Med Chem 2023; 66:6274-6287. [PMID: 37130037 DOI: 10.1021/acs.jmedchem.3c00098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Designed ligands of G protein-coupled receptors can exert a spectrum of modulating effects, varying from full agonists and partial agonists to antagonists and inverse agonists. For the dopamine D2 receptor (D2R), partial agonist activity is the pharmacological feature of the third-generation antipsychotics, including aripiprazole, brexpiprazole, and cariprazine. Started from a benzofuran-derived D2R full agonist O4LE6 (4), which was identified using a structure-based method by us in previous studies, a series of D2R partial agonists were designed and synthesized by introducing different tail groups. Among them, compound 10b showed excellent activity in D2R pharmacological assays. Further optimizations using a structural rigidification approach led to the discovery of brain-penetrant compounds 29c and 29d, which exhibited potent antipsychotic effects in the mouse hyperlocomotion model. Compound 29c also showed excellent drug-like pharmacokinetic properties in rats and qualifies as an antipsychotic agent that is worth further evaluations.
Collapse
Affiliation(s)
- Ruiquan Liu
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China
| | - Jianzhong Qi
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Huan Wang
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China
| | - Luyu Fan
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Pei Zhang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jing Yu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Liang Tan
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China
| |
Collapse
|
11
|
Chestnykh D, Graßl F, Pfeifer C, Dülk J, Ebner C, Walters M, von Hörsten S, Kornhuber J, Kalinichenko LS, Heinrich M, Müller CP. Behavioural effects of APH199, a selective dopamine D4 receptor agonist, in animal models. Psychopharmacology (Berl) 2023; 240:1011-1031. [PMID: 36854793 PMCID: PMC10006056 DOI: 10.1007/s00213-023-06347-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/21/2023] [Indexed: 03/02/2023]
Abstract
RATIONALE The dopamine D4 receptors (DRD4) play a key role in numerous brain functions and are involved in the pathogenesis of various psychiatric disorders. DRD4 ligands have been shown to moderate anxiety, reward and depression-like behaviours, and cognitive impairments. Despite a series of promising but ambiguous findings, the therapeutic advantages of DRD4 stimulation remain elusive. OBJECTIVES The investigation focused on the behavioural effects of the recently developed DRD4 agonist, APH199, to evaluate its impact on anxiety, anhedonia, behavioural despair, establishment and retrieval of alcohol reinforcement, and amphetamine (AMPH)-induced symptoms. METHODS Male C57BL/6 J mice and Sprague-Dawley rats were examined in five independent experiments. We assessed APH199 (0.1-5 mg/kg, i.p.) effects on a broad range of behavioural parameters in the open field (OF) test, conditioned place preference test (CPP), elevated plus maze (EPM), light-dark box (LDB), novelty suppressed feeding (NSF), forced swim test (FST), sucrose preference test (SPT), AMPH-induced hyperlocomotion test (AIH), and prepulse inhibition (PPI) of the acoustic startle response in AMPH-sensitized rats. RESULTS APH199 caused mild and sporadic anxiolytic and antidepressant effects in EPM and FST, but no remarkable impact on behaviour in other tests in mice. However, we found a significant increase in AMPH-induced hyperactivity, suggesting an exaggeration of the psychotic-like responses in the AMPH-sensitized rats. CONCLUSIONS Our data challenged the hypothesis of the therapeutic benefits of DRD4 agonists, pointing out a possible aggravation of psychosis. We suggest a need for further preclinical studies to ensure the safety of antipsychotics with DRD4 stimulating properties.
Collapse
Affiliation(s)
- Daria Chestnykh
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Fabian Graßl
- Department of Chemistry and Pharmacy, Friedrich-Alexander-University of Erlangen-Nuremberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Canice Pfeifer
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Jonas Dülk
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Chiara Ebner
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Mona Walters
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Stephan von Hörsten
- Department of Experimental Therapy, Preclinical Experimental Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Palmsanlage 5, 91054, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Markus Heinrich
- Department of Chemistry and Pharmacy, Friedrich-Alexander-University of Erlangen-Nuremberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany.
- Centre for Drug Research, University Sains Malaysia, Penang, Minden, Malaysia.
| |
Collapse
|
12
|
Wenk D, Khan S, Ignatchenko V, Hübner H, Gmeiner P, Weikert D, Pischetsrieder M, Kislinger T. Phosphoproteomic Analysis of Dopamine D2 Receptor Signaling Reveals Interplay of G Protein- and β-Arrestin-Mediated Effects. J Proteome Res 2023; 22:259-271. [PMID: 36508580 PMCID: PMC9831068 DOI: 10.1021/acs.jproteome.2c00707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Leveraging biased signaling of G protein-coupled receptors has been proposed as a promising strategy for the development of drugs with higher specificity. However, the consequences of selectively targeting G protein- or β-arrestin-mediated signaling on cellular functions are not comprehensively understood. In this study, we utilized phosphoproteomics to gain a systematic overview of signaling induced by the four biased and balanced dopamine D2 receptor (D2R) ligands MS308, BM138, quinpirole, and sulpiride in an in vitro D2R transfection model. Quantification of 14,160 phosphosites revealed a low impact of the partial G protein agonist MS308 on cellular protein phosphorylation, as well as surprising similarities between the balanced agonist quinpirole and the inverse agonist sulpiride. Analysis of the temporal profiles of ligand-induced phosphorylation events showed a transient impact of the G protein-selective agonist MS308, whereas the β-arrestin-preferring agonist BM138 elicited a delayed, but more pronounced response. Functional enrichment analysis of ligand-impacted phosphoproteins and treatment-linked kinases confirmed multiple known functions of D2R signaling while also revealing novel effects, for example of MS308 on sterol regulatory element-binding protein-related gene expression. All raw data were deposited in MassIVE (MSV000089457).
Collapse
Affiliation(s)
- Deborah Wenk
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada
| | - Shahbaz Khan
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada
| | - Vladimir Ignatchenko
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada
| | - Harald Hübner
- Medicinal
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Medicinal
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Dorothee Weikert
- Medicinal
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Monika Pischetsrieder
- Food
Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Thomas Kislinger
- Princess
Margaret Cancer Centre, University Health
Network, 101 College
Street, Toronto, Ontario M5G 1L7, Canada,Department
of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada,
| |
Collapse
|
13
|
Juza R, Musilek K, Mezeiova E, Soukup O, Korabecny J. Recent advances in dopamine D 2 receptor ligands in the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:55-211. [PMID: 36111795 DOI: 10.1002/med.21923] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Dopamine is a biologically active amine synthesized in the central and peripheral nervous system. This biogenic monoamine acts by activating five types of dopamine receptors (D1-5 Rs), which belong to the G protein-coupled receptor family. Antagonists and partial agonists of D2 Rs are used to treat schizophrenia, Parkinson's disease, depression, and anxiety. The typical pharmacophore with high D2 R affinity comprises four main areas, namely aromatic moiety, cyclic amine, central linker and aromatic/heteroaromatic lipophilic fragment. From the literature reviewed herein, we can conclude that 4-(2,3-dichlorophenyl), 4-(2-methoxyphenyl)-, 4-(benzo[b]thiophen-4-yl)-1-substituted piperazine, and 4-(6-fluorobenzo[d]isoxazol-3-yl)piperidine moieties are critical for high D2 R affinity. Four to six atoms chains are optimal for D2 R affinity with 4-butoxyl as the most pronounced one. The bicyclic aromatic/heteroaromatic systems are most frequently occurring as lipophilic appendages to retain high D2 R affinity. In this review, we provide a thorough overview of the therapeutic potential of D2 R modulators in the treatment of the aforementioned disorders. In addition, this review summarizes current knowledge about these diseases, with a focus on the dopaminergic pathway underlying these pathologies. Major attention is paid to the structure, function, and pharmacology of novel D2 R ligands, which have been developed in the last decade (2010-2021), and belong to the 1,4-disubstituted aromatic cyclic amine group. Due to the abundance of data, allosteric D2 R ligands and D2 R modulators from patents are not discussed in this review.
Collapse
Affiliation(s)
- Radomir Juza
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Mezeiova
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
14
|
Dimethyl Sulfoxide-Assisted, Iodine- and Ascorbic Acid-Catalyzed One-Pot Synthetic Approach for Constructing Highly Substituted Pyrazolo[1,5- a]quinoline Thioether Derivatives. J Org Chem 2022; 87:13856-13872. [PMID: 36215433 DOI: 10.1021/acs.joc.2c01557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A dimethyl sulfoxide-assisted and iodine/ascorbic acid-catalyzed simple approach to pyrazolo[1,5-a]quinoline thioether derivatives 22 is described. The compounds were identified using 1H NMR, 13C NMR, high-resolution mass spectrometry, and single-crystal X-ray diffractometry. The pyrazolo[1,5-a]quinoline thioether was synthesized in a stepwise fashion through aryl sulfenylation and benzannulation strategies. The generated heteroaryl thioether compounds 23 were exposed to the benzannulation path to produce pyrazolo[1,5-a]quinoline thioether 22. The benzannulation reaction proceeds by way of diazotization of the pyrazole amine derivative 23, radical generation by the removal of nitrogen, and eventually trapping of the aryl radical with the support of phenylacetylene 19. A catalytic amount of ascorbic acid aided the benzannulation reaction. There were several other control studies conducted, including trapping reactions with isopropenyl acetate, tetramethylpiperidine N-oxyl reactions, and reactions without phenylacetylene. Since a change in the substitution has previously demonstrated substantial bioactivity, the core structure of pyrazole was evaluated for functional group tolerance. A reasonable mechanism is then proposed, accompanied by the support of control experiments and scope. A Suzuki reaction was used to create an aryl/heteroaryl compound 35 from one of the synthesized compounds 22b. In the controlled oxidation reaction paths, molecule 22a was selectively transformed into the corresponding sulfoxide 32 and sulfone 33.
Collapse
|
15
|
Fink EA, Xu J, Hübner H, Braz JM, Seemann P, Avet C, Craik V, Weikert D, Schmidt MF, Webb CM, Tolmachova NA, Moroz YS, Huang XP, Kalyanaraman C, Gahbauer S, Chen G, Liu Z, Jacobson MP, Irwin JJ, Bouvier M, Du Y, Shoichet BK, Basbaum AI, Gmeiner P. Structure-based discovery of nonopioid analgesics acting through the α 2A-adrenergic receptor. Science 2022; 377:eabn7065. [PMID: 36173843 PMCID: PMC10360211 DOI: 10.1126/science.abn7065] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Because nonopioid analgesics are much sought after, we computationally docked more than 301 million virtual molecules against a validated pain target, the α2A-adrenergic receptor (α2AAR), seeking new α2AAR agonists chemotypes that lack the sedation conferred by known α2AAR drugs, such as dexmedetomidine. We identified 17 ligands with potencies as low as 12 nanomolar, many with partial agonism and preferential Gi and Go signaling. Experimental structures of α2AAR complexed with two of these agonists confirmed the docking predictions and templated further optimization. Several compounds, including the initial docking hit '9087 [mean effective concentration (EC50) of 52 nanomolar] and two analogs, '7075 and PS75 (EC50 4.1 and 4.8 nanomolar), exerted on-target analgesic activity in multiple in vivo pain models without sedation. These newly discovered agonists are interesting as therapeutic leads that lack the liabilities of opioids and the sedation of dexmedetomidine.
Collapse
Affiliation(s)
- Elissa A. Fink
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Jun Xu
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Joao M. Braz
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Philipp Seemann
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Charlotte Avet
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Veronica Craik
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Maximilian F. Schmidt
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Chase M. Webb
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, CA, USA
| | - Nataliya A. Tolmachova
- Enamine Ltd., 02094 Kyiv, Ukraine
- Institute of Bioorganic Chemistry and Petrochemistry, National Ukrainian Academy of Science, 02660 Kyiv, Ukraine
| | - Yurii S. Moroz
- National Taras Shevchenko University of Kyiv, 01601 Kyiv, Ukraine
- Chemspace, Riga LV-1082, Latvia
| | - Xi-Ping Huang
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Chakrapani Kalyanaraman
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Stefan Gahbauer
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Zheng Liu
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Allan I. Basbaum
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| |
Collapse
|
16
|
Hsiao PY, Chang R, Sue ACH, Chu JH, Liao GW, Lee YH, Huang JY. Synthesis and Mechanistic Investigation of Bipyrazolo[1,5- a]pyridines via Palladium-Catalyzed Cross-Dehydrogenative Coupling of Pyrazolo[1,5- a]pyridines. J Org Chem 2022; 87:9851-9863. [PMID: 35844185 DOI: 10.1021/acs.joc.2c00895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The synthesis of a range of 3,3'-bipyrazolo[1,5-a]pyridine derivatives via direct cross-dehydrogenative coupling of pyrazolo[1,5-a]pyridine precursors is herein presented. This simple and efficient methodology involving palladium(II)-catalyzed C-H bond activation showed good functional group tolerance and product yield (up to 94%). Through the mechanistic insights gained from both kinetic isotope effect experimental studies and density functional theory calculations, a plausible reaction mechanism was outlined. Furthermore, subsequent derivatizations of the resulting 7,7'-diaryl-3,3'-bipyrazolo[1,5-a]pyridines, executed by performing palladium-mediated ortho C-H bond activation followed by hypervalent iodine-induced chlorination, rendered this series of compounds more extended π-conjugation and twisted conformations. Our study on these bipyrazolo[1,5-a]pyridine-based luminogens provides new opportunities for tailor-made organic luminescent materials.
Collapse
Affiliation(s)
- Pu-Yen Hsiao
- Department of Applied Science, National Taitung University, Taitung 95092, Taiwan, R.O.C
| | - Rong Chang
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Andrew C-H Sue
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jean-Ho Chu
- Department of Applied Science, National Taitung University, Taitung 95092, Taiwan, R.O.C
| | - Guan-Wei Liao
- Department of Applied Science, National Taitung University, Taitung 95092, Taiwan, R.O.C
| | - Yi-Hsin Lee
- Department of Applied Science, National Taitung University, Taitung 95092, Taiwan, R.O.C
| | - Jui-Yang Huang
- Department of Applied Science, National Taitung University, Taitung 95092, Taiwan, R.O.C
| |
Collapse
|
17
|
Hsiao PY, Chu JH. Novel bipyrazolo[1,5- a]pyridine luminogens with aggregation-induced emission enhancement properties. Chem Commun (Camb) 2021; 57:12281-12284. [PMID: 34730135 DOI: 10.1039/d1cc05371j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A novel 3,3'-bipyrazolo[1,5-a]pyridine molecular scaffold was obtained as a product of serendipity. Both photophysical characterisations and HOMO-LUMO theoretical calculations indicate its potential as a promising fluorophore with notable intramolecular charge transfer. Nonetheless, the emission properties of this compound suffer from the typical aggregation-caused quenching effect. To overcome this situation, we introduced additional diaryl groups onto the skeleton and synthesised a series of 7,7'-diaryl-3,3'-bipyrazolo[1,5-a]pyridines via palladium-catalysed intermolecular C-H/C-H bond cross-coupling reaction in 35-62% yields. This series of tailor-made luminogens with twisted π-structures display aggregation-induced emission enhancement properties.
Collapse
Affiliation(s)
- Pu-Yen Hsiao
- Department of Applied Science, National Taitung University, Taitung, Taiwan 95092, Taiwan.
| | - Jean-Ho Chu
- Department of Applied Science, National Taitung University, Taitung, Taiwan 95092, Taiwan.
| |
Collapse
|
18
|
Stepniewski TM, Mancini A, Ågren R, Torrens-Fontanals M, Semache M, Bouvier M, Sahlholm K, Breton B, Selent J. Mechanistic insights into dopaminergic and serotonergic neurotransmission - concerted interactions with helices 5 and 6 drive the functional outcome. Chem Sci 2021; 12:10990-11003. [PMID: 34522296 PMCID: PMC8386650 DOI: 10.1039/d1sc00749a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/15/2021] [Indexed: 01/14/2023] Open
Abstract
Brain functions rely on neurotransmitters that mediate communication between billions of neurons. Disruption of this communication can result in a plethora of psychiatric and neurological disorders. In this work, we combine molecular dynamics simulations, live-cell biosensor and electrophysiological assays to investigate the action of the neurotransmitter dopamine at the dopaminergic D2 receptor (D2R). The study of dopamine and closely related chemical probes reveals how neurotransmitter binding translates into the activation of distinct subsets of D2R effectors (i.e.: Gi2, GoB, Gz and β-arrestin 2). Ligand interactions with key residues in TM5 (S5.42) and TM6 (H6.55) in the D2R binding pocket yield a dopamine-like coupling signature, whereas exclusive TM5 interaction is typically linked to preferential G protein coupling (in particular GoB) over β-arrestin. Further experiments for serotonin receptors indicate that the reported molecular mechanism is shared by other monoaminergic neurotransmitter receptors. Ultimately, our study highlights how sequence variation in position 6.55 is used by nature to fine-tune β-arrestin recruitment and in turn receptor signaling and internalization of neurotransmitter receptors.
Collapse
Affiliation(s)
- Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF) Dr Aiguader 88 Barcelona E-08003 Spain
- InterAx Biotech AG, PARK InnovAARE 5234 Villigen Switzerland
| | - Arturo Mancini
- Domain Therapeutics NA Inc 7171 Frederick-Banting Saint-Laurent (QC) H4S 1Z9 Canada
| | - Richard Ågren
- Department of Neuroscience, Karolinska Institute Stockholm Sweden
| | - Mariona Torrens-Fontanals
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF) Dr Aiguader 88 Barcelona E-08003 Spain
| | - Meriem Semache
- Domain Therapeutics NA Inc 7171 Frederick-Banting Saint-Laurent (QC) H4S 1Z9 Canada
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Université de Montréal Montreal QC H3C 3J7 Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal Montréal Québec H3T 1J4 Canada
| | - Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institute Stockholm Sweden
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University 90187 Umeå Sweden
| | - Billy Breton
- Domain Therapeutics NA Inc 7171 Frederick-Banting Saint-Laurent (QC) H4S 1Z9 Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal Montréal Québec H3T 1J4 Canada
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF) Dr Aiguader 88 Barcelona E-08003 Spain
| |
Collapse
|
19
|
Mennie KM, Reutershan MH, White C, Adams B, Becker B, Deng J, Katz JD, LaBlue E, Margrey K, Saurí J. Divergent and Regioselective Synthesis of Pyrazolo[1,5- a]pyridines and Imidazo[1,5- a]pyridines. Org Lett 2021; 23:4694-4698. [PMID: 34037404 DOI: 10.1021/acs.orglett.1c01431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nitrogenous heterocycles are ubiquitous in pharmaceuticals and drug-like compounds; however, regioselective synthesis has proved challenging. Herein we report our efforts to develop a regioselective method for the synthesis of pyrazolo[1,5-a]pyridines and the serendipitous discovery of a protocol for the regioselective formation of imidazo[1,5-a]pyridines. Together, these transformations allow for the rapid and selective formation of two important heterocyclic motifs from a common intermediate.
Collapse
Affiliation(s)
| | | | - Catherine White
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Bruce Adams
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Bridget Becker
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - James Deng
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Jason D Katz
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | | | - Kaila Margrey
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Josep Saurí
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| |
Collapse
|
20
|
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
|
21
|
Ullmann T, Gienger M, Budzinski J, Hellmann J, Hübner H, Gmeiner P, Weikert D. Homobivalent Dopamine D 2 Receptor Ligands Modulate the Dynamic Equilibrium of D 2 Monomers and Homo- and Heterodimers. ACS Chem Biol 2021; 16:371-379. [PMID: 33435665 DOI: 10.1021/acschembio.0c00895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dopamine D2 receptors (D2Rs) are major targets in the treatment of psychiatric and neurodegenerative diseases. As with many other G protein-coupled receptors (GPCRs), D2Rs interact within the cellular membrane, leading to a transient receptor homo- or heterodimerization. These interactions are known to alter ligand binding, signaling, and receptor trafficking. Bivalent ligands are ideally suited to target GPCR dimers and are composed of two pharmacophores connected by a spacer element. If properly designed, bivalent ligands are able to engange the two orthosteric binding sites of a GPCR dimer simultaneously. Taking advantage of previously developed ligands for heterodimers of D2R and the neurotensin receptor 1 (NTSR1), we synthesized homobivalent ligands targeting D2R. Employing bioluminescence resonance energy transfer, we found that the bivalent ligands 3b and 4b comprising a 92-atom spacer are able to foster D2R-homodimerization while simultaneously reducing interactions of D2R with NTSR1. Both receptors are coexpressed in the central nervous system and involved in important physiological processes. The newly developed bivalent ligands are excellent tools to further understand the pharmacological consequences of D2R homo- and heterodimerization. Not limited to the dopaminergic system, modifying class A GPCRs' dynamic equilibrium between monomers, homomers, and heteromers with bivalent ligands may represent a novel pharmacological concept paving the way toward innovative drugs.
Collapse
Affiliation(s)
- Tamara Ullmann
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Marie Gienger
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Julian Budzinski
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Jan Hellmann
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
22
|
Wenk D, Ignatchenko V, Macklin A, Hübner H, Gmeiner P, Weikert D, Pischetsrieder M, Kislinger T. Functionally selective activation of the dopamine receptor D 2 is mirrored by the protein expression profiles. Sci Rep 2021; 11:3501. [PMID: 33568753 PMCID: PMC7875989 DOI: 10.1038/s41598-021-83038-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/28/2021] [Indexed: 01/11/2023] Open
Abstract
The development of functionally selective or biased ligands is a promising approach towards drugs with less side effects. Biased ligands for G protein-coupled receptors can selectively induce G protein activation or β-arrestin recruitment. The consequences of this selective action on cellular functions, however, are not fully understood. Here, we investigated the impact of five biased and balanced dopamine D2 receptor agonists and antagonists on the global protein expression in HEK293T cells by untargeted nanoscale liquid chromatography-tandem mass spectrometry. The proteome analysis detected 5290 protein groups. Hierarchical clustering and principal component analysis based on the expression levels of 1462 differential proteins led to a separation of antagonists and balanced agonist from the control treatment, while the biased ligands demonstrated larger similarities to the control. Functional analysis of affected proteins revealed that the antagonists haloperidol and sulpiride regulated exocytosis and peroxisome function. The balanced agonist quinpirole, but not the functionally selective agonists induced a downregulation of proteins involved in synaptic signaling. The β-arrestin-preferring agonist BM138, however, regulated several proteins related to neuron function and the dopamine receptor-mediated signaling pathway itself. The G protein-selective partial agonist MS308 influenced rather broad functional terms such as DNA processing and mitochondrial translation.
Collapse
Affiliation(s)
- Deborah Wenk
- Food Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Vladimir Ignatchenko
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON, Canada
| | - Andrew Macklin
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON, Canada
| | - Harald Hübner
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Peter Gmeiner
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Dorothée Weikert
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Monika Pischetsrieder
- Food Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany.
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, Canada
| |
Collapse
|
23
|
Willmann M, Ermert J, Prante O, Hübner H, Gmeiner P, Neumaier B. Radiosynthesis and evaluation of 18F-labeled dopamine D 4-receptor ligands. Nucl Med Biol 2021; 92:43-52. [PMID: 32718750 DOI: 10.1016/j.nucmedbio.2020.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/09/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The dopamine D4 receptor (D4R) has attracted considerable attention as potential target for the treatment of a broad range of central nervous system disorders. Although many efforts have been made to improve the performance of putative radioligand candidates, there is still a lack of D4R selective tracers suitable for in vivo PET imaging. Thus, the objective of this work was to develop a D4-selective PET ligand for clinical applications. METHODS Four compounds based on previous and new lead structures were prepared and characterized with regard to their D4R subtype selectivity and predicted lipophilicity. From these, 3-((4-(2-fluorophenyl)piperazin-1-yl)methyl)-1H-pyrrolo[2,3-b]pyridine I and (S)-4-(3-fluoro-4-methoxybenzyl)-2-(phenoxymethyl)morpholine II were selected for labeling with fluorine-18 and subsequent evaluation by in vitro autoradiography to assess their suitability as D4 radioligand candidates for in vivo imaging. RESULTS The radiosynthesis of [18F]I and [18F]II was successfully achieved by copper-mediated radiofluorination with radiochemical yields of 7% and 66%, respectively. The radioligand [18F]II showed specific binding in areas where D4 expression is expected, whereas [18F]I did not show any uptake in distinct brain regions and exhibited an unacceptable degree of non-specific binding. CONCLUSIONS The compounds studied exhibited high D4R subtype selectivity and logP values compatible with high brain uptake, but only ligand [18F]II showed low non-specific binding and is therefore a good candidate for further evaluation. ADVANCES IN KNOWLEDGE The discovery of new lead structures for high-affinity D4 ligands opens up new possibilities for the development of suitable PET-radioligands. IMPLICATIONS FOR PATIENT PET-imaging of dopamine D4-receptors could facilitate understanding, diagnosis and treatment of neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Michael Willmann
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen Straße, 52428 Jülich, Germany
| | - Johannes Ermert
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen Straße, 52428 Jülich, Germany.
| | - Olaf Prante
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, 91054 Erlangen, Germany
| | - Harald Hübner
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department Chemistry and Pharmacy, Medicinal Chemistry, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department Chemistry and Pharmacy, Medicinal Chemistry, 91058 Erlangen, Germany
| | - Bernd Neumaier
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen Straße, 52428 Jülich, Germany; University of Colgne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, 50937 Cologne, Germany
| |
Collapse
|
24
|
Uzuneser TC, Weiss EM, Dahlmanns J, Kalinichenko LS, Amato D, Kornhuber J, Alzheimer C, Hellmann J, Kaindl J, Hübner H, Löber S, Gmeiner P, Grömer TW, Müller CP. Presynaptic vesicular accumulation is required for antipsychotic efficacy in psychotic-like rats. J Psychopharmacol 2021; 35:65-77. [PMID: 33274688 PMCID: PMC7770212 DOI: 10.1177/0269881120965908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The therapeutic effects of antipsychotic drugs (APDs) are mainly attributed to their postsynaptic inhibitory functions on the dopamine D2 receptor, which, however, cannot explain the delayed onset of full therapeutic efficacy. It was previously shown that APDs accumulate in presynaptic vesicles during chronic treatment and are released like neurotransmitters in an activity-dependent manner triggering an auto-inhibitory feedback mechanism. Although closely mirroring therapeutic action onset, the functional consequence of the APD accumulation process remained unclear. AIMS Here we tested whether the accumulation of the APD haloperidol (HAL) is required for full therapeutic action in psychotic-like rats. METHODS We designed a HAL analog compound (HAL-F), which lacks the accumulation property of HAL, but retains its postsynaptic inhibitory action on dopamine D2 receptors. RESULTS/OUTCOMES By perfusing LysoTracker fluorophore-stained cultured hippocampal neurons, we confirmed the accumulation of HAL and the non-accumulation of HAL-F. In an amphetamine hypersensitization psychosis-like model in rats, we found that subchronic intracerebroventricularly delivered HAL (0.1 mg/kg/day), but not HAL-F (0.3-1.5 mg/kg/day), attenuates psychotic-like behavior in rats. CONCLUSIONS/INTERPRETATION These findings suggest the presynaptic accumulation of HAL may serve as an essential prerequisite for its full antipsychotic action and may explain the time course of APD action. Targeting accumulation properties of APDs may, thus, become a new strategy to improve APD action.
Collapse
Affiliation(s)
- Taygun C Uzuneser
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Eva-Maria Weiss
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jana Dahlmanns
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Davide Amato
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany,Department of Neuroscience, Medical University of South Carolina, Charleston, USA
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jan Hellmann
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jonas Kaindl
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Löber
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Teja W Grömer
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany,Christian P Müller, Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany.
| |
Collapse
|
25
|
Allikalt A, Purkayastha N, Flad K, Schmidt MF, Tabor A, Gmeiner P, Hübner H, Weikert D. Fluorescent ligands for dopamine D 2/D 3 receptors. Sci Rep 2020; 10:21842. [PMID: 33318558 PMCID: PMC7736868 DOI: 10.1038/s41598-020-78827-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022] Open
Abstract
Fluorescent ligands are versatile tools for the study of G protein-coupled receptors. Depending on the fluorophore, they can be used for a range of different applications, including fluorescence microscopy and bioluminescence or fluorescence resonance energy transfer (BRET or FRET) assays. Starting from phenylpiperazines and indanylamines, privileged scaffolds for dopamine D2-like receptors, we developed dansyl-labeled fluorescent ligands that are well accommodated in the binding pockets of D2 and D3 receptors. These receptors are the target proteins for the therapy for several neurologic and psychiatric disorders, including Parkinson’s disease and schizophrenia. The dansyl-labeled ligands exhibit binding affinities up to 0.44 nM and 0.29 nM at D2R and D3R, respectively. When the dansyl label was exchanged for sterically more demanding xanthene or cyanine dyes, fluorescent ligands 10a-c retained excellent binding properties and, as expected from their indanylamine pharmacophore, acted as agonists at D2R. While the Cy3B-labeled ligand 10b was used to visualize D2R and D3R on the surface of living cells by total internal reflection microscopy, ligand 10a comprising a rhodamine label showed excellent properties in a NanoBRET binding assay at D3R.
Collapse
Affiliation(s)
- Anni Allikalt
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Nirupam Purkayastha
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Khajidmaa Flad
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Maximilian F Schmidt
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Alina Tabor
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany.
| |
Collapse
|
26
|
Signal profiling of the β 1AR reveals coupling to novel signalling pathways and distinct phenotypic responses mediated by β 1AR and β 2AR. Sci Rep 2020; 10:8779. [PMID: 32471984 PMCID: PMC7260363 DOI: 10.1038/s41598-020-65636-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
A comprehensive understanding of signalling downstream of GPCRs requires a broad approach to capture novel signalling modalities in addition to established pathways. Here, using an array of sixteen validated BRET-based biosensors, we analyzed the ability of seven different β-adrenergic ligands to engage five distinct signalling pathways downstream of the β1-adrenergic receptor (β1AR). In addition to generating signalling signatures and capturing functional selectivity for the different ligands toward these pathways, we also revealed coupling to signalling pathways that have not previously been ascribed to the βAR. These include coupling to Gz and G12 pathways. The signalling cascade linking the β1AR to calcium mobilization was also characterized using a combination of BRET-based biosensors and CRISPR-engineered HEK 293 cells lacking the Gαs subunit or with pharmacological or genetically engineered pathway inhibitors. We show that both Gs and G12 are required for the full calcium response. Our work highlights the power of combining signal profiling with genome editing approaches to capture the full complement of GPCR signalling activities in a given cell type and to probe their underlying mechanisms.
Collapse
|
27
|
Newman AH, Battiti FO, Bonifazi A. 2016 Philip S. Portoghese Medicinal Chemistry Lectureship: Designing Bivalent or Bitopic Molecules for G-Protein Coupled Receptors. The Whole Is Greater Than the Sum of Its Parts. J Med Chem 2020; 63:1779-1797. [PMID: 31499001 PMCID: PMC8281448 DOI: 10.1021/acs.jmedchem.9b01105] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The genesis of designing bivalent or bitopic molecules that engender unique pharmacological properties began with Portoghese's work directed toward opioid receptors, in the early 1980s. This strategy has evolved as an attractive way to engineer highly selective compounds for targeted G-protein coupled receptors (GPCRs) with optimized efficacies and/or signaling bias. The emergence of X-ray crystal structures of many GPCRs and the identification of both orthosteric and allosteric binding sites have provided further guidance to ligand drug design that includes a primary pharmacophore (PP), a secondary pharmacophore (SP), and a linker between them. It is critical to note the synergistic relationship among all three of these components as they contribute to the overall interaction of these molecules with their receptor proteins and that strategically designed combinations have and will continue to provide the GPCR molecular tools of the future.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Corresponding author: Amy H. Newman: Phone: (443)-740-2887. Fax: (443)-740-2111.
| | - Francisco O. Battiti
- 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
| | - Alessandro Bonifazi
- 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
|
28
|
Sanchez-Soto M, Verma RK, Willette BKA, Gonye EC, Moore AM, Moritz AE, Boateng CA, Yano H, Free RB, Shi L, Sibley DR. A structural basis for how ligand binding site changes can allosterically regulate GPCR signaling and engender functional selectivity. Sci Signal 2020; 13:13/617/eaaw5885. [PMID: 32019899 DOI: 10.1126/scisignal.aaw5885] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Signaling bias is the propensity for some agonists to preferentially stimulate G protein-coupled receptor (GPCR) signaling through one intracellular pathway versus another. We previously identified a G protein-biased agonist of the D2 dopamine receptor (D2R) that results in impaired β-arrestin recruitment. This signaling bias was predicted to arise from unique interactions of the ligand with a hydrophobic pocket at the interface of the second extracellular loop and fifth transmembrane segment of the D2R. Here, we showed that residue Phe189 within this pocket (position 5.38 using Ballesteros-Weinstein numbering) functions as a microswitch for regulating receptor interactions with β-arrestin. This residue is relatively conserved among class A GPCRs, and analogous mutations within other GPCRs similarly impaired β-arrestin recruitment while maintaining G protein signaling. To investigate the mechanism of this signaling bias, we used an active-state structure of the β2-adrenergic receptor (β2R) to build β2R-WT and β2R-Y1995.38A models in complex with the full β2R agonist BI-167107 for molecular dynamics simulations. These analyses identified conformational rearrangements in β2R-Y1995.38A that propagated from the extracellular ligand binding site to the intracellular surface, resulting in a modified orientation of the second intracellular loop in β2R-Y1995.38A, which is predicted to affect its interactions with β-arrestin. Our findings provide a structural basis for how ligand binding site alterations can allosterically affect GPCR-transducer interactions and result in biased signaling.
Collapse
Affiliation(s)
- Marta Sanchez-Soto
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA
| | - Ravi Kumar Verma
- Computational Chemistry and Molecular Biophysics Unit, NIDA, NIH, TRIAD Technology Center, 333 Cassell Drive, Room 1121, Baltimore, MD 21224, USA
| | - Blair K A Willette
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA
| | - Elizabeth C Gonye
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA
| | - Annah M Moore
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA
| | - Amy E Moritz
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA
| | - Comfort A Boateng
- Basic Pharmaceutical Sciences, High Point University, One University Parkway, High Point, NC 27268, USA
| | - Hideaki Yano
- Computational Chemistry and Molecular Biophysics Unit, NIDA, NIH, TRIAD Technology Center, 333 Cassell Drive, Room 1121, Baltimore, MD 21224, USA
| | - R Benjamin Free
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, NIDA, NIH, TRIAD Technology Center, 333 Cassell Drive, Room 1121, Baltimore, MD 21224, USA.
| | - David R Sibley
- Molecular Neuropharmacology Section, NINDS, NIH, 35 Convent Drive, Room 3A201, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Pirzer AS, Lasch R, Friedrich H, Hübner H, Gmeiner P, Heinrich MR. Benzyl Phenylsemicarbazides: A Chemistry-Driven Approach Leading to G Protein-Biased Dopamine D4 Receptor Agonists with High Subtype Selectivity. J Med Chem 2019; 62:9658-9679. [DOI: 10.1021/acs.jmedchem.9b01085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Anna S. Pirzer
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Roman Lasch
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Heike Friedrich
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Markus R. Heinrich
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
30
|
Martini ML, Ray C, Yu X, Liu J, Pogorelov VM, Wetsel WC, Huang XP, McCorvy JD, Caron MG, Jin J. Designing Functionally Selective Noncatechol Dopamine D 1 Receptor Agonists with Potent In Vivo Antiparkinsonian Activity. ACS Chem Neurosci 2019; 10:4160-4182. [PMID: 31387346 DOI: 10.1021/acschemneuro.9b00410] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dopamine receptors are important G protein-coupled receptors (GPCRs) with therapeutic opportunities for treating Parkinson's Disease (PD) motor and cognitive deficits. Biased D1 dopamine ligands that differentially activate G protein over β-arrestin recruitment pathways are valuable chemical tools for dissecting positive versus negative effects in drugs for PD. Here, we reveal an iterative approach toward modification of a D1-selective noncatechol scaffold critical for G protein-biased agonism. This approach provided enhanced understanding of the structural components critical for activity and signaling bias and led to the discovery of several novel compounds with useful pharmacological properties, including three highly GS-biased partial agonists. Administration of a potent, balanced, and brain-penetrant lead compound from this series results in robust antiparkinsonian effects in a rodent model of PD. This study suggests that the noncatechol ligands developed through this approach are valuable tools for probing D1 receptor signaling biology and biased agonism in models of neurologic disease.
Collapse
Affiliation(s)
- Michael L. Martini
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Caroline Ray
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Xufen Yu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Vladimir M. Pogorelov
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
- Departments of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - William C. Wetsel
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
- Departments of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Xi-Ping Huang
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - John D. McCorvy
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Marc G. Caron
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
31
|
Silva RR, Parreiras-E-Silva LT, Pompeu TET, Duarte DA, Fraga CAM, Barreiro EJ, Menegatti R, Costa-Neto CM, Noël F. Evaluation of Functional Selectivity of Haloperidol, Clozapine, and LASSBio-579, an Experimental Compound With Antipsychotic-Like Actions in Rodents, at G Protein and Arrestin Signaling Downstream of the Dopamine D 2 Receptor. Front Pharmacol 2019; 10:628. [PMID: 31214037 PMCID: PMC6558205 DOI: 10.3389/fphar.2019.00628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 05/15/2019] [Indexed: 12/28/2022] Open
Abstract
LASSBio-579, an N-phenylpiperazine antipsychotic lead compound, has been previously reported as a D2 receptor (D2R) ligand with antipsychotic-like activities in rodent models of schizophrenia. In order to better understand the molecular mechanism of action of LASSBio-579 and of its main metabolite, LQFM 037, we decided to address the hypothesis of functional selectivity at the D2R. HEK-293T cells transiently coexpressing the human long isoform of D2 receptor (D2LR) and bioluminescence resonance energy transfer (BRET)-based biosensors were used. The antagonist activity was evaluated using different concentrations of the compounds in the presence of a submaximal concentration of dopamine (DA), after 5 and 20 min. For both signaling pathways, haloperidol, clozapine, and our compounds act as DA antagonists in a concentration-dependent manner, with haloperidol being by far the most potent, consistent with its nanomolar D2R affinity measured in binding assays. In our experimental conditions, only haloperidol presented a robust functional selectivity, being four- to fivefold more efficient for inhibiting translocation of β-arrestin-2 (β-arr2) than for antagonizing Gi activation. Present data are the first report on the effects of LASSBio-579 and LQFM 037 on the β-arr2 signaling pathway and further illustrate that the functional activity could vary depending on the assay conditions and approaches used.
Collapse
Affiliation(s)
- Rafaela R Silva
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas T Parreiras-E-Silva
- Laboratory of structure and function of 7 Transmembrane Receptors (7TMR), Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thais E T Pompeu
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego A Duarte
- Laboratory of structure and function of 7 Transmembrane Receptors (7TMR), Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos A M Fraga
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliezer J Barreiro
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Menegatti
- Laboratório de Química Farmacêutica Medicinal, Pharmacy School, Federal University of Goiás, Goiânia, Brazil
| | - Claudio M Costa-Neto
- Laboratory of structure and function of 7 Transmembrane Receptors (7TMR), Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - François Noël
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Shen Y, McCorvy JD, Martini ML, Rodriguiz RM, Pogorelov VM, Ward KM, Wetsel WC, Liu J, Roth BL, Jin J. D 2 Dopamine Receptor G Protein-Biased Partial Agonists Based on Cariprazine. J Med Chem 2019; 62:4755-4771. [PMID: 30964661 PMCID: PMC6509010 DOI: 10.1021/acs.jmedchem.9b00508] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Functionally selective G protein-coupled receptor ligands are valuable tools for deciphering the roles of downstream signaling pathways that potentially contribute to therapeutic effects versus side effects. Recently, we discovered both Gi/o-biased and β-arrestin2-biased D2 receptor agonists based on the Food and Drug Administration (FDA)-approved drug aripiprazole. In this work, based on another FDA-approved drug, cariprazine, we conducted a structure-functional selectivity relationship study and discovered compound 38 (MS1768) as a potent partial agonist that selectively activates the Gi/o pathway over β-arrestin2. Unlike the dual D2R/D3R partial agonist cariprazine, compound 38 showed selective agonist activity for D2R over D3R. In fact, compound 38 exhibited potent antagonism of dopamine-stimulated β-arrestin2 recruitment. In our docking studies, compound 38 directly interacts with S1935.42 on TM5 but has no interactions with extracellular loop 2, which appears to be in contrast to the binding poses of D2R β-arrestin2-biased ligands. In in vivo studies, compound 38 showed high D2R receptor occupancy in mice and effectively inhibited phencyclidine-induced hyperlocomotion.
Collapse
Affiliation(s)
- Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - John D. McCorvy
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Michael L. Martini
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ramona M. Rodriguiz
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Vladimir M. Pogorelov
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Karen M. Ward
- Worldwide Research and Development, Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts 02139, United States
| | - William C. Wetsel
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Bryan L. Roth
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
33
|
Seyedabadi M, Ghahremani MH, Albert PR. Biased signaling of G protein coupled receptors (GPCRs): Molecular determinants of GPCR/transducer selectivity and therapeutic potential. Pharmacol Ther 2019; 200:148-178. [PMID: 31075355 DOI: 10.1016/j.pharmthera.2019.05.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
G protein coupled receptors (GPCRs) convey signals across membranes via interaction with G proteins. Originally, an individual GPCR was thought to signal through one G protein family, comprising cognate G proteins that mediate canonical receptor signaling. However, several deviations from canonical signaling pathways for GPCRs have been described. It is now clear that GPCRs can engage with multiple G proteins and the line between cognate and non-cognate signaling is increasingly blurred. Furthermore, GPCRs couple to non-G protein transducers, including β-arrestins or other scaffold proteins, to initiate additional signaling cascades. Receptor/transducer selectivity is dictated by agonist-induced receptor conformations as well as by collateral factors. In particular, ligands stabilize distinct receptor conformations to preferentially activate certain pathways, designated 'biased signaling'. In this regard, receptor sequence alignment and mutagenesis have helped to identify key receptor domains for receptor/transducer specificity. Furthermore, molecular structures of GPCRs bound to different ligands or transducers have provided detailed insights into mechanisms of coupling selectivity. However, receptor dimerization, compartmentalization, and trafficking, receptor-transducer-effector stoichiometry, and ligand residence and exposure times can each affect GPCR coupling. Extrinsic factors including cell type or assay conditions can also influence receptor signaling. Understanding these factors may lead to the development of improved biased ligands with the potential to enhance therapeutic benefit, while minimizing adverse effects. In this review, evidence for ligand-specific GPCR signaling toward different transducers or pathways is elaborated. Furthermore, molecular determinants of biased signaling toward these pathways and relevant examples of the potential clinical benefits and pitfalls of biased ligands are discussed.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Pharmacology, School of Medicine, Bushehr University of Medical Sciences, Iran; Education Development Center, Bushehr University of Medical Sciences, Iran
| | | | - Paul R Albert
- Ottawa Hospital Research Institute, Neuroscience, University of Ottawa, Canada.
| |
Collapse
|
34
|
Abstract
A great deal of experimental evidence suggests that ligands can stabilize different receptor active states that go on to interact with cellular signaling proteins to form a range of different complexes in varying quantities. In pleiotropically linked receptor systems, this leads to selective activation of some signaling pathways at the expense of others (biased signaling). This article summarizes the current knowledge about the complex components of receptor systems, the evidence that biased signaling is used in natural physiology to fine-tune signaling, and the current thoughts on how this mechanism may be applied to the design of better drugs. Although this is a fairly newly discovered phenomenon, theoretical and experimental data suggest that it is a ubiquitous behavior of ligands and receptors and to be expected. Biased signaling is simple to detect in vitro and there are numerous methods to quantify the effect with scales that can be used to optimize this activity in structure-activity medicinal chemistry studies. At present, the major hurdle in the application of this mechanism to therapeutics is the translation of in vitro bias to in vivo effect; this is because of the numerous factors that can modify measures of bias in natural physiologic systems. In spite of this, biased signaling still has the potential to justify revisiting of receptor targets previously thought to be intractable and also furnishes the means to pursue targets previously thought to be forbidden due to deleterious physiology (as these may be eliminated through biased signaling).
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
35
|
Martini ML, Liu J, Ray C, Yu X, Huang XP, Urs A, Urs N, McCorvy JD, Caron MG, Roth BL, Jin J. Defining Structure-Functional Selectivity Relationships (SFSR) for a Class of Non-Catechol Dopamine D 1 Receptor Agonists. J Med Chem 2019; 62:3753-3772. [PMID: 30875219 DOI: 10.1021/acs.jmedchem.9b00351] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
G protein-coupled receptors (GPCRs) are capable of downstream signaling through distinct noncanonical pathways such as β-arrestins in addition to the canonical G protein-dependent pathways. GPCR ligands that differentially activate the downstream signaling pathways are termed functionally selective or biased ligands. A class of novel non-catechol G protein-biased agonists of the dopamine D1 receptor (D1R) was recently disclosed. We conducted the first comprehensive structure-functional selectivity relationship study measuring GS and β-arrestin2 recruitment activities focused on four regions of this scaffold, resulting in over 50 analogs with diverse functional selectivity profiles. Some compounds became potent full agonists of β-arrestin2 recruitment, while others displayed enhanced GS bias compared to the starting compound. Pharmacokinetic testing of an analog with an altered functional selectivity profile demonstrated excellent blood-brain barrier penetration. This study provides novel tools for studying ligand bias at D1R and paves the way for developing the next generation of biased D1R ligands.
Collapse
Affiliation(s)
| | | | | | | | - Xi-Ping Huang
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Aarti Urs
- Department of Pharmacology and Therapeutics, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - Nikhil Urs
- Department of Pharmacology and Therapeutics, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - John D McCorvy
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States.,Department of Cell Biology, Neurobiology and Anatomy , Medical College of Wisconsin , Milwaukee , Wisconsin 53226 , United States
| | | | - Bryan L Roth
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | | |
Collapse
|
36
|
Bonifazi A, Yano H, Guerrero AM, Kumar V, Hoffman AF, Lupica CR, Shi L, Newman AH. Novel and Potent Dopamine D 2 Receptor Go-Protein Biased Agonists. ACS Pharmacol Transl Sci 2019; 2:52-65. [PMID: 30775693 PMCID: PMC6371206 DOI: 10.1021/acsptsci.8b00060] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Indexed: 12/18/2022]
Abstract
![]()
The
discovery of functionally biased and physiologically beneficial
ligands directed toward G-protein coupled receptors (GPCRs) has provided
the impetus to design dopamine D2 receptor (D2R) targeted molecules that may be therapeutically advantageous for
the treatment of certain neuropsychiatric or basal ganglia related
disorders. Here we describe the synthesis of a novel series of D2R agonists linking the D2R unbiased agonist sumanirole
with privileged secondary molecular fragments. The resulting ligands
demonstrate improved D2R affinity and selectivity over
sumanirole. Extensive in vitro functional studies
and bias factor analysis led to the identification of a novel class
of highly potent Go-protein biased full D2R agonists with
more than 10-fold and 1000-fold bias selectivity toward activation
of specific G-protein subtypes and β-arrestin, respectively.
Intracellular electrophysiological recordings from midbrain dopamine
neurons demonstrated that Go-protein selective agonists can elicit
prolonged ligand-induced GIRK activity via D2Rs, which
may be beneficial in the treatment of dyskinesias associated with
dopamine system dysfunction.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- 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
| | - Hideaki Yano
- Computational Chemistry and Molecular Biophysics Unit, 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
| | - Adrian M Guerrero
- 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
| | - Vivek Kumar
- 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
| | - Alexander F Hoffman
- Electrophysiology Research Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Carl R Lupica
- Electrophysiology Research Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, 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
| | - 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
|
37
|
Uzuneser TC, Speidel J, Kogias G, Wang AL, de Souza Silva MA, Huston JP, Zoicas I, von Hörsten S, Kornhuber J, Korth C, Müller CP. Disrupted-in-Schizophrenia 1 (DISC1) Overexpression and Juvenile Immune Activation Cause Sex-Specific Schizophrenia-Related Psychopathology in Rats. Front Psychiatry 2019; 10:222. [PMID: 31057438 PMCID: PMC6465888 DOI: 10.3389/fpsyt.2019.00222] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023] Open
Abstract
Synaptic pruning is a critical refinement step during neurodevelopment, and schizophrenia has been associated with overpruning of cortical dendritic spines. Both human studies and animal models implicate disrupted-in-schizophrenia 1 (DISC1) gene as a strong susceptibility factor for schizophrenia. Accumulating evidence supports the involvement of DISC1 protein in the modulation of synaptic elimination during critical periods of neurodevelopment and of dopamine D2-receptor-mediated signaling during adulthood. In many species, synaptic pruning occurs during juvenile and adolescent periods and is mediated by microglia, which can be over-activated by an immune challenge, giving rise to overpruning. Therefore, we sought to investigate possible interactions between a transgenic DISC1 model (tgDISC1) and juvenile immune activation (JIA) by the bacterial cell wall endotoxin lipopolysaccharide on the induction of schizophrenia-related behavioral and neurochemical disruptions in adult female and male rats. We examined possible behavioral aberrations along three major symptom dimensions of schizophrenia including psychosis, social and emotional disruptions, and cognitive impairments. We detected significant gene-environment interactions in the amphetamine-induced locomotion in female animals and in the amphetamine-induced anxiety in male animals. Surprisingly, gene-environment interactions improved social memory in both male and female animals. JIA alone disrupted spatial memory and recognition memory, but only in male animals. DISC1 overexpression alone induced an improvement in sensorimotor gating, but only in female animals. Our neurochemical analyses detected sex- and manipulation-dependent changes in the postmortem monoamine content of animals. Taken together, we here report sex-specific effects of environment and genotype as well as their interaction on behavioral phenotypes and neurochemical profiles relevant for schizophrenia.
Collapse
Affiliation(s)
- Taygun C Uzuneser
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jil Speidel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Georgios Kogias
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - An-Li Wang
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf, Germany
| | - Maria A de Souza Silva
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf, Germany
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf, Germany
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Stephan von Hörsten
- Department of Experimental Therapy, Preclinical Experimental Center, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Carsten Korth
- Department of Neuropathology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
38
|
Schizophrenia dimension-specific antipsychotic drug action and failure in amphetamine-sensitized psychotic-like rats. Eur Neuropsychopharmacol 2018; 28:1382-1393. [PMID: 30243682 DOI: 10.1016/j.euroneuro.2018.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
Abstract
Schizophrenic patients suffer from various disruptions in their psyche, mood and cognition, most of which cannot be effectively treated with the available antipsychotic drugs. Some dimensions of the schizophrenia syndrome in man can be mimicked in animals by the amphetamine (AMPH)-sensitization-induced psychosis model. Using such a sensitization procedure, we induced a psychosis-like syndrome in rats, measured as a deficit in sensory information processing and memory deficits. We then investigated the possible restorative effects of continuous treatment with haloperidol (HAL), a typical antipsychotic drug, on distinct dimensions of the syndrome. We found that, continuous infusion of a clinically relevant dose of HAL (0.5 mg/kg/day) effectively ameliorated AMPH-sensitization-induced sensorimotor gating disruptions after seven days of treatment. However, the sensory information processing deficit reappeared after prolonged HAL treatment, suggesting a treatment failure in this dimension of the syndrome. HAL had at this dose little beneficial effects on the cognitive deficits. In contrast, a continuously administered low dose of HAL (0.05 mg/kg/day) successfully attenuated cognitive deficits, but aggravated the sensorimotor gating deficit under both short- or long-term treatment conditions. Post mortem neurochemical analysis revealed that the psychotic-like behavior induced by our manipulations might be explained by altered monoamine levels in distinct brain regions. These findings provide evidence for dissociating and dose-dependent HAL treatment action and failure at different dimensions of schizophrenia.
Collapse
|
39
|
Zhang P, Lv H, Duan G, Dong J, Ge Y. A novel pyrazolo[1,5- a]pyridine fluorophore and its application to detect pH in cells. RSC Adv 2018; 8:30732-30735. [PMID: 35548712 PMCID: PMC9085528 DOI: 10.1039/c8ra06191b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 08/17/2018] [Indexed: 01/09/2023] Open
Abstract
A new fluorescent probe based on pyrazolo[1,5-a]pyridine was synthesized and used to monitor the pH in cells. This probe exhibited a fast response to acidic pH (less than 10 s), a high quantum yield (φ = 0.64), and high selectivity and sensitivity. The response mechanism of the fluorescent probes relies on the ICT change.
Collapse
Affiliation(s)
- Ping Zhang
- School of Chemistry and Pharmaceutical Engineering, Taishan Medical University Taian Shandong 271016 P. R. China +86-538-6229741 +86-538-6229741
| | - Huaying Lv
- Laigang Hospital Affiliated to Taishan Medical University Lai'wu 271126 P. R. China
| | - Guiyun Duan
- School of Chemistry and Pharmaceutical Engineering, Taishan Medical University Taian Shandong 271016 P. R. China +86-538-6229741 +86-538-6229741
| | - Jian Dong
- School of Chemistry and Pharmaceutical Engineering, Taishan Medical University Taian Shandong 271016 P. R. China +86-538-6229741 +86-538-6229741
| | - Yanqing Ge
- School of Chemistry and Pharmaceutical Engineering, Taishan Medical University Taian Shandong 271016 P. R. China +86-538-6229741 +86-538-6229741
| |
Collapse
|
40
|
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
|
41
|
Yan P, Duan G, Ji R, Ge Y. A simple and efficient synthesis of new fluorophore 4-hydroxy pyrazolo[1,5-a]pyridines through a tandem reaction. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.05.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Weïwer M, Xu Q, Gale JP, Lewis M, Campbell AJ, Schroeder FA, Van de Bittner GC, Walk M, Amaya A, Su P, D Ordevic L, Sacher JR, Skepner A, Fei D, Dennehy K, Nguyen S, Faloon PW, Perez J, Cottrell JR, Liu F, Palmer M, Pan JQ, Hooker JM, Zhang YL, Scolnick E, Wagner FF, Holson EB. Functionally Biased D2R Antagonists: Targeting the β-Arrestin Pathway to Improve Antipsychotic Treatment. ACS Chem Biol 2018; 13:1038-1047. [PMID: 29485852 DOI: 10.1021/acschembio.8b00168] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a severe neuropsychiatric disease that lacks completely effective and safe therapies. As a polygenic disorder, genetic studies have only started to shed light on its complex etiology. To date, the positive symptoms of schizophrenia are well-managed by antipsychotic drugs, which primarily target the dopamine D2 receptor (D2R). However, these antipsychotics are often accompanied by severe side effects, including motoric symptoms. At D2R, antipsychotic drugs antagonize both G-protein dependent (Gαi/o) signaling and G-protein independent (β-arrestin) signaling. However, the relevant contributions of the distinct D2R signaling pathways to antipsychotic efficacy and on-target side effects (motoric) are still incompletely understood. Recent evidence from mouse genetic and pharmacological studies point to β-arrestin signaling as the major driver of antipsychotic efficacy and suggest that a β-arrestin biased D2R antagonist could achieve an additional level of selectivity at D2R, increasing the therapeutic index of next generation antipsychotics. Here, we characterize BRD5814, a highly brain penetrant β-arrestin biased D2R antagonist. BRD5814 demonstrated good target engagement via PET imaging, achieving efficacy in an amphetamine-induced hyperlocomotion mouse model with strongly reduced motoric side effects in a rotarod performance test. This proof of concept study opens the possibility for the development of a new generation of pathway selective antipsychotics at D2R with reduced side effect profiles for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Michel Weïwer
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Qihong Xu
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Jennifer P Gale
- Center for the Development of Therapeutics , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Michael Lewis
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Frederick A Schroeder
- Department of Radiology, MGH , Athinoula A. Martinos Center for Biomedical Imaging , Charlestown , Massachusetts 02129 , United States
| | - Genevieve C Van de Bittner
- Department of Radiology, MGH , Athinoula A. Martinos Center for Biomedical Imaging , Charlestown , Massachusetts 02129 , United States
| | - Michelle Walk
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Aldo Amaya
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health , University of Toronto , Toronto , Ontario M5T1R8 , Canada
| | - Luka D Ordevic
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Joshua R Sacher
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Adam Skepner
- Center for the Development of Therapeutics , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - David Fei
- Center for the Development of Therapeutics , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Kelly Dennehy
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Shannon Nguyen
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Patrick W Faloon
- Center for the Development of Therapeutics , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Jose Perez
- Center for the Development of Therapeutics , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Jeffrey R Cottrell
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health , University of Toronto , Toronto , Ontario M5T1R8 , Canada
| | - Michelle Palmer
- Center for the Development of Therapeutics , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Jen Q Pan
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Jacob M Hooker
- Department of Radiology, MGH , Athinoula A. Martinos Center for Biomedical Imaging , Charlestown , Massachusetts 02129 , United States
| | - Yan-Ling Zhang
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Edward Scolnick
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Florence F Wagner
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| | - Edward B Holson
- Stanley Center for Psychiatric Research , Broad Institute of MIT and Harvard , Cambridge , Massachusetts 02142 , United States
| |
Collapse
|
43
|
Rajacic SK, Schwall G, Penjisevic J, Andric D, Sukalovic V, Soskic V. Identification of NQO1 and ferrochelatase as interaction partners for neuroprotective N-{[2-(4-phenyl-piperazin-1-yl)-ethyl]-phenyl}-arylamides. Chem Biol Drug Des 2018. [PMID: 29543381 DOI: 10.1111/cbdd.13193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Affinity chromatography was used to identify potential cellular targets that are responsible for neuroprotective activity of N-{[2-(4-phenyl-piperazin-1-yl)-ethyl]-phenyl}-arylamides. Active and inactive representatives of N-{[2-(4-phenyl-piperazin-1-yl)-ethyl]-phenyl}-arylamides bearing an extended linker were synthesized and immobilized on an agarose-based matrix. This was followed by the identification of specifically bound proteins isolated out of the whole rat brain extract. Inducible flavoprotein NAD(P)H:quinone oxidoreductase (NQO1) was identified as candidates for cellular targets.
Collapse
Affiliation(s)
| | | | - Jelena Penjisevic
- ICTM - Center of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Deana Andric
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | | | | |
Collapse
|
44
|
Chun LS, Vekariya RH, Free RB, Li Y, Lin DT, Su P, Liu F, Namkung Y, Laporte SA, Moritz AE, Aubé J, Frankowski KJ, Sibley DR. Structure-Activity Investigation of a G Protein-Biased Agonist Reveals Molecular Determinants for Biased Signaling of the D 2 Dopamine Receptor. Front Synaptic Neurosci 2018. [PMID: 29515433 PMCID: PMC5826336 DOI: 10.3389/fnsyn.2018.00002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The dopamine D2 receptor (D2R) is known to elicit effects through activating two major signaling pathways mediated by either G proteins (Gi/o) or β-arrestins. However, the specific role of each pathway in physiological or therapeutic activities is not known with certainty. One approach to the dissection of these pathways is through the use of drugs that can selectively modulate one pathway vs. the other through a mechanism known as functional selectivity or biased signaling. Our laboratory has previously described a G protein signaling-biased agonist, MLS1547, for the D2R using a variety of in vitro functional assays. To further evaluate the biased signaling activity of this compound, we investigated its ability to promote D2R internalization, a process known to be mediated by β-arrestin. Using multiple cellular systems and techniques, we found that MLS1547 promotes little D2R internalization, which is consistent with its inability to recruit β-arrestin. Importantly, we validated these results in primary striatal neurons where the D2R is most highly expressed suggesting that MLS1547 will exhibit biased signaling activity in vivo. In an effort to optimize and further explore structure-activity relationships (SAR) for this scaffold, we conducted an iterative chemistry campaign to synthesize and characterize novel analogs of MLS1547. The resulting analysis confirmed previously described SAR requirements for G protein-biased agonist activity and, importantly, elucidated new structural features that are critical for agonist efficacy and signaling bias of the MLS1547 scaffold. One of the most important determinants for G protein-biased signaling is the interaction of a hydrophobic moiety of the compound with a defined pocket formed by residues within transmembrane five and extracellular loop two of the D2R. These results shed new light on the mechanism of biased signaling of the D2R and may lead to improved functionally-selective molecules.
Collapse
Affiliation(s)
- Lani S Chun
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Rakesh H Vekariya
- Department of Medicinal Chemistry and Specialized Chemistry Center, University of Kansas, Lawrence, KS, United States
| | - R Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Yun Li
- Neural Engineering Unit, Behavior Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Da-Ting Lin
- Neural Engineering Unit, Behavior Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Ping Su
- Molecular Neuroscience, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Fang Liu
- Molecular Neuroscience, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Yoon Namkung
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC, Canada
| | - Stephane A Laporte
- Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal, QC, Canada
| | - Amy E Moritz
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey Aubé
- Department of Medicinal Chemistry and Specialized Chemistry Center, University of Kansas, Lawrence, KS, United States
| | - Kevin J Frankowski
- Department of Medicinal Chemistry and Specialized Chemistry Center, University of Kansas, Lawrence, KS, United States
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
45
|
Fish I, Stößel A, Eitel K, Valant C, Albold S, Huebner H, Möller D, Clark MJ, Sunahara RK, Christopoulos A, Shoichet BK, Gmeiner P. Structure-Based Design and Discovery of New M 2 Receptor Agonists. J Med Chem 2017; 60:9239-9250. [PMID: 29094937 DOI: 10.1021/acs.jmedchem.7b01113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Muscarinic receptor agonists are characterized by apparently strict restraints on their tertiary or quaternary amine and their distance to an ester or related center. On the basis of the active state crystal structure of the muscarinic M2 receptor in complex with iperoxo, we explored potential agonists that lacked the highly conserved functionalities of previously known ligands. Using structure-guided pharmacophore design followed by docking, we found two agonists (compounds 3 and 17), out of 19 docked and synthesized compounds, that fit the receptor well and were predicted to form a hydrogen-bond conserved among known agonists. Structural optimization led to compound 28, which was 4-fold more potent than its parent 3. Fortified by the discovery of this new scaffold, we sought a broader range of chemotypes by docking 2.2 million fragments, which revealed another three micromolar agonists unrelated either to 28 or known muscarinics. Even pockets as tightly defined and as deeply studied as that of the muscarinic reveal opportunities for the structure-based design and the discovery of new chemotypes.
Collapse
Affiliation(s)
- Inbar Fish
- Department of Pharmaceutical Chemistry, University of California, San Francisco , San Francisco, California 94158, United States.,Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel-Aviv University , Ramat Aviv, Israel
| | - Anne Stößel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Katrin Eitel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville Victoria 3052, Australia
| | - Sabine Albold
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville Victoria 3052, Australia
| | - Harald Huebner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Dorothee Möller
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| | - Mary J Clark
- Department of Pharmacology, University of California, San Diego , La Jolla, California 92093, United States
| | - Roger K Sunahara
- Department of Pharmacology, University of California, San Diego , La Jolla, California 92093, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville Victoria 3052, Australia
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco , San Francisco, California 94158, United States
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University , Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
46
|
Männel B, Jaiteh M, Zeifman A, Randakova A, Möller D, Hübner H, Gmeiner P, Carlsson J. Structure-Guided Screening for Functionally Selective D 2 Dopamine Receptor Ligands from a Virtual Chemical Library. ACS Chem Biol 2017; 12:2652-2661. [PMID: 28846380 DOI: 10.1021/acschembio.7b00493] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Functionally selective ligands stabilize conformations of G protein-coupled receptors (GPCRs) that induce a preference for signaling via a subset of the intracellular pathways activated by the endogenous agonists. The possibility to fine-tune the functional activity of a receptor provides opportunities to develop drugs that selectively signal via pathways associated with a therapeutic effect and avoid those causing side effects. Animal studies have indicated that ligands displaying functional selectivity at the D2 dopamine receptor (D2R) could be safer and more efficacious drugs against neuropsychiatric diseases. In this work, computational design of functionally selective D2R ligands was explored using structure-based virtual screening. Molecular docking of known functionally selective ligands to a D2R homology model indicated that such compounds were anchored by interactions with the orthosteric site and extended into a common secondary pocket. A tailored virtual library with close to 13 000 compounds bearing 2,3-dichlorophenylpiperazine, a privileged orthosteric scaffold, connected to diverse chemical moieties via a linker was docked to the D2R model. Eighteen top-ranked compounds that occupied both the orthosteric and allosteric site were synthesized, leading to the discovery of 16 partial agonists. A majority of the ligands had comparable maximum effects in the G protein and β-arrestin recruitment assays, but a subset displayed preference for a single pathway. In particular, compound 4 stimulated β-arrestin recruitment (EC50 = 320 nM, Emax = 16%) but had no detectable G protein signaling. The use of structure-based screening and virtual libraries to discover GPCR ligands with tailored functional properties will be discussed.
Collapse
Affiliation(s)
- Barbara Männel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Mariama Jaiteh
- Science
for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| | - Alexey Zeifman
- Science
for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| | - Alena Randakova
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Dorothee Möller
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Jens Carlsson
- Science
for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|
47
|
Männel B, Hübner H, Möller D, Gmeiner P. β-Arrestin biased dopamine D2 receptor partial agonists: Synthesis and pharmacological evaluation. Bioorg Med Chem 2017; 25:5613-5628. [DOI: 10.1016/j.bmc.2017.08.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/04/2017] [Accepted: 08/20/2017] [Indexed: 01/11/2023]
|
48
|
Visualization of ligand-induced dopamine D 2S and D 2L receptor internalization by TIRF microscopy. Sci Rep 2017; 7:10894. [PMID: 28883522 PMCID: PMC5589927 DOI: 10.1038/s41598-017-11436-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/23/2017] [Indexed: 01/11/2023] Open
Abstract
G protein-coupled receptors (GPCRs), including the dopamine receptors, represent a group of important pharmacological targets. Upon agonist binding, GPCRs frequently undergo internalization, a process that is known to attenuate functional responses upon prolonged exposure to agonists. In this study, internalization was visualized by means of total internal reflection fluorescence (TIRF) microscopy at a level of discrete single events near the plasma membrane with high spatial resolution. A novel method has been developed to determine the relative extent of internalized fluorescent receptor-ligand complexes by comparative fluorescence quantification in living CHO cells. The procedure entails treatment with the reducing agent sodium borohydride, which converts cyanine-based fluorescent ligands on the membrane surface to a long-lived reduced form. Because the highly polar reducing agent is not able to pass the cell membrane, the fluorescent receptor-ligand complexes located in internalized compartments remain fluorescent under TIRF illumination. We applied the method to investigate differences of the short (D2S) and the long (D2L) isoforms of dopamine D2 receptors in their ability to undergo agonist-induced internalization.
Collapse
|
49
|
Männel B, Dengler D, Shonberg J, Hübner H, Möller D, Gmeiner P. Hydroxy-Substituted Heteroarylpiperazines: Novel Scaffolds for β-Arrestin-Biased D2R Agonists. J Med Chem 2017; 60:4693-4713. [DOI: 10.1021/acs.jmedchem.7b00363] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Barbara Männel
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nuernberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Daniela Dengler
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nuernberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Jeremy Shonberg
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nuernberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nuernberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Dorothee Möller
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nuernberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and
Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nuernberg, Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|